1
|
Benne N, Ter Braake D, Stoppelenburg AJ, Broere F. Nanoparticles for Inducing Antigen-Specific T Cell Tolerance in Autoimmune Diseases. Front Immunol 2022; 13:864403. [PMID: 35392079 PMCID: PMC8981588 DOI: 10.3389/fimmu.2022.864403] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Autoimmune diseases affect many people worldwide. Current treatment modalities focus on the reduction of disease symptoms using anti-inflammatory drugs which can lead to side effects due to systemic immune suppression. Restoration of immune tolerance by down-regulating auto-reactive cells in an antigen-specific manner is currently the “holy grail” for the treatment of autoimmune diseases. A promising strategy is the use of nanoparticles that can deliver antigens to antigen-presenting cells which in turn can enhance antigen-specific regulatory T cells. In this review, we highlight some promising cell targets (e.g. liver sinusoidal endothelial cells and splenic marginal zone macrophages) for exploiting natural immune tolerance processes, and several strategies by which antigen-carrying nanoparticles can target these cells. We also discuss how nanoparticles carrying immunomodulators may be able to activate tolerance in other antigen-presenting cell types. Finally, we discuss some important aspects that must be taken into account when translating data from animal studies to patients.
Collapse
Affiliation(s)
- Naomi Benne
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Daniëlle Ter Braake
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Arie Jan Stoppelenburg
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Clinical Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
2
|
Karrich JJ, Romera-Hernández M, Papazian N, Veenbergen S, Cornelissen F, Aparicio-Domingo P, Stenhouse FH, Peddie CD, Hoogenboezem RM, den Hollander CWJ, Gaskell T, Medley T, Boon L, Blackburn CC, Withers DR, Samsom JN, Cupedo T. Expression of Plet1 controls interstitial migration of murine small intestinal dendritic cells. Eur J Immunol 2018; 49:290-301. [PMID: 30537036 PMCID: PMC6492104 DOI: 10.1002/eji.201847671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/30/2018] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
Abstract
Under homeostatic conditions, dendritic cells (DCs) continuously patrol the intestinal lamina propria. Upon antigen encounter, DCs initiate C‐C motif chemokine receptor 7 (CCR7) expression and migrate into lymph nodes to direct T cell activation and differentiation. The mechanistic underpinnings of DC migration from the tissues to lymph nodes have been largely elucidated, contributing greatly to our understanding of DC functionality and intestinal immunity. In contrast, the molecular mechanisms allowing DCs to efficiently migrate through the complex extracellular matrix of the intestinal lamina propria prior to antigen encounter are still incompletely understood. Here we show that small intestinal murine CD11b+CD103+ DCs express Placenta‐expressed transcript 1 (Plet1), a glycophoshatidylinositol (GPI)‐anchored surface protein involved in migration of keratinocytes during wound healing. In the absence of Plet1, CD11b+CD103+ DCs display aberrant migratory behavior, and accumulate in the small intestine, independent of CCR7 responsiveness. RNA‐sequencing indicated involvement of Plet1 in extracellular matrix‐interactiveness, and subsequent in‐vitro migration assays revealed that Plet1 augments the ability of DCs to migrate through extracellular matrix containing environments. In conclusion, our findings reveal that expression of Plet1 facilitates homeostatic interstitial migration of small intestinal DCs.
Collapse
Affiliation(s)
- Julien J Karrich
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sharon Veenbergen
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ferry Cornelissen
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Frances H Stenhouse
- MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - C Diana Peddie
- MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Terri Gaskell
- MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Tanya Medley
- MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | - C Clare Blackburn
- MRC, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - David R Withers
- MRC, Centre for Immune Regulation, University of Birmingham, Birmingham, UK
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Roghanian A, Stopforth RJ, Dahal LN, Cragg MS. New revelations from an old receptor: Immunoregulatory functions of the inhibitory Fc gamma receptor, FcγRIIB (CD32B). J Leukoc Biol 2018; 103:1077-1088. [PMID: 29406570 DOI: 10.1002/jlb.2mir0917-354r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The Fc gamma receptor IIB (FcγRIIB/CD32B) was generated million years ago during evolution. It is the sole inhibitory receptor for IgG, and has long been associated with the regulation of humoral immunity and innate immune homeostasis. However, new and surprising functions of FcγRIIB are emerging. In particular, FcγRIIB has been shown to perform unexpected activatory roles in both immune-signaling and monoclonal antibody (mAb) immunotherapy. Furthermore, although ITIM signaling is an integral part of FcγRIIB regulatory activity, it is now clear that inhibition/activation of immune responses can occur independently of the ITIM. In light of these new findings, we present an overview of the established and noncanonical functions of FcγRIIB and discuss how this knowledge might be exploited therapeutically.
Collapse
Affiliation(s)
- Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard J Stopforth
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
4
|
Abstract
In this issue Ohsaki et al. (https://doi.org/10.1084/jem.20171163) explain how breastfeeding can prevent the onset of food allergies in offspring by instructing T reg formation via neonatal Fc receptor (FcRn)-mediated transfer and uptake of allergen-containing IgG immune complexes (Ig-ICs) by gut dendritic cells (DCs).
Collapse
Affiliation(s)
- Bart N Lambrecht
- VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
| |
Collapse
|
5
|
Howe SE, Sowa G, Konjufca V. Systemic and Mucosal Antibody Responses to Soluble and Nanoparticle-Conjugated Antigens Administered Intranasally. Antibodies (Basel) 2016; 5:antib5040020. [PMID: 31558001 PMCID: PMC6698832 DOI: 10.3390/antib5040020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/16/2016] [Accepted: 09/18/2016] [Indexed: 02/03/2023] Open
Abstract
Nanoparticles (NPs) are increasingly being used for drug delivery, as well as antigen carriers and immunostimulants for the purpose of developing vaccines. In this work, we examined how intranasal (i.n.) priming followed by i.n. or subcutaneous (s.c.) boosting immunization affects the humoral immune response to chicken ovalbumin (Ova) and Ova conjugated to 20 nm NPs (NP-Ova). We show that i.n. priming with 20 mg of soluble Ova, a dose known to trigger oral tolerance when administered via gastric gavage, induced substantial systemic IgG1 and IgG2c, as well as mucosal antibodies. These responses were further boosted following a s.c. immunization with Ova and complete Freund’s adjuvant (Ova+CFA). In contrast, 100 µg of Ova delivered via NPs induced an IgG1-dominated systemic response, and primed the intestinal mucosa for secretion of IgA. Following a secondary s.c. or i.n. immunization with Ova+CFA or NP-Ova, systemic IgG1 titers significantly increased, and serum IgG2c and intestinal antibodies were induced in mice primed nasally with NP-Ova. Only Ova- and NP-Ova-primed mice that were s.c.-boosted exhibited substantial systemic and mucosal titers for up to 6 months after priming, whereas the antibodies of i.n.-boosted mice declined over time. Our results indicate that although the amount of Ova delivered by NPs was 1000-fold less than Ova delivered in soluble form, the antigen-specific antibody responses, both systemic and mucosal, are essentially identical by 6 months following the initial priming immunization. Additionally, both i.n.- and s.c.-boosting strategies for NP-Ova-primed mice were capable of inducing a polarized Th1/Th2 immune response, as well as intestinal antibodies; however, it is only by using a heterogeneous prime-boost strategy that long-lasting antibody responses were initiated. These results provide valuable insight for future mucosal vaccine development, as well as furthering our understanding of mucosal antibody responses.
Collapse
Affiliation(s)
- Savannah E Howe
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA.
| | - Gavin Sowa
- Department of Chemistry, Southern Illinois University, Carbondale, IL 62901, USA.
| | - Vjollca Konjufca
- Department of Microbiology, Southern Illinois University, Carbondale, IL 62901, USA.
| |
Collapse
|
6
|
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]
|
7
|
Tasaniyananda N, Chaisri U, Tungtrongchitr A, Chaicumpa W, Sookrung N. Mouse Model of Cat Allergic Rhinitis and Intranasal Liposome-Adjuvanted Refined Fel d 1 Vaccine. PLoS One 2016; 11:e0150463. [PMID: 26954254 PMCID: PMC4783078 DOI: 10.1371/journal.pone.0150463] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Cats (Felis domesticus) are rich source of airborne allergens that prevailed in the environment and sensitized a number of people to allergy. In this study, a mouse model of allergic rhinitis caused by the cat allergens was developed for the first time and the model was used for testing therapeutic efficacy of a novel intranasal liposome-entrapped vaccines made of native Fel d 1 (major cat allergen) in comparison with the vaccine made of crude cat hair extract (cCE). BALB/c mice were sensitized with cCE mixed with alum intraperitoneally and intranasally. The allergic mice were treated with eight doses of either liposome (L)-entrapped native Fel d 1 (L-nFD1), L-cCE), or placebo on every alternate day. Vaccine efficacy evaluation was performed one day after provoking the treated mice with aerosolic cCE. All allergenized mice developed histological features of allergic rhinitis with rises of serum specific-IgE and Th2 cytokine gene expression. Serum IgE and intranasal mucus production of allergic mice reduced significantly after vaccination in comparison with the placebo mice. The vaccines also caused a shift of the Th2 response (reduction of Th2 cytokine expressions) towards the non-pathogenic responses: Th1 (down-regulation of the Th1 suppressive cytokine gene, IL-35) and Treg (up-regulation of IL-10 and TGF-β). In conclusions, a mouse model of allergic rhinitis to cat allergens was successfully developed. The intranasal, liposome-adjuvanted vaccines, especially the refined single allergen formulation, assuaged the allergic manifestations in the modeled mice. The prototype vaccine is worthwhile testing further for clinical use in the pet allergic patients.
Collapse
Affiliation(s)
- Natt Tasaniyananda
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Anchalee Tungtrongchitr
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nitat Sookrung
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- * E-mail:
| |
Collapse
|
8
|
Krishnamoorthy S, Liu T, Drager D, Patarroyo-White S, Chhabra ES, Peters R, Josephson N, Lillicrap D, Blumberg RS, Pierce GF, Jiang H. Recombinant factor VIII Fc (rFVIIIFc) fusion protein reduces immunogenicity and induces tolerance in hemophilia A mice. Cell Immunol 2016; 301:30-9. [PMID: 26775174 PMCID: PMC4936482 DOI: 10.1016/j.cellimm.2015.12.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/25/2015] [Accepted: 12/28/2015] [Indexed: 12/15/2022]
Abstract
Anti-factor VIII (FVIII) antibodies is a major complication of FVIII replacement therapy for hemophilia A. We investigated the immune response to recombinant human factor VIII Fc (rFVIIIFc) in comparison to BDD-rFVIII and full-length rFVIII (FL-rFVIII) in hemophilia A mice. Repeated administration of therapeutically relevant doses of rFVIIIFc in these mice resulted in significantly lower antibody responses to rFVIII compared to BDD-rFVIII and FL-rFVIII and reduced antibody production upon subsequent challenge with high doses of rFVIIIFc. The induction of a tolerogenic response by rFVIIIFc was associated with higher percentage of regulatory T-cells, a lower percentage of pro-inflammatory splenic T-cells, and up-regulation of tolerogenic cytokines and markers. Disruption of Fc interactions with either FcRn or Fcγ receptors diminished tolerance induction, suggesting the involvement of these pathways. These results indicate that rFVIIIFc reduces immunogenicity and imparts tolerance to rFVIII demonstrating that recombinant therapeutic proteins may be modified to influence immunogenicity and facilitate tolerance.
Collapse
Affiliation(s)
- Sriram Krishnamoorthy
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States,Corresponding authors. (S. Krishnamoorthy), (H. Jiang)
| | - Tongyao Liu
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Douglas Drager
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | | | - Ekta Seth Chhabra
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Robert Peters
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Neil Josephson
- Division of Hematology, University of Washington School of Medicine, Puget Sound Blood Center, Seattle, WA 98104, United States
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Canada
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Glenn F. Pierce
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Haiyan Jiang
- Hematology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States,Corresponding authors. (S. Krishnamoorthy), (H. Jiang)
| |
Collapse
|
9
|
Kaufman GN, Massoud AH, Dembele M, Yona M, Piccirillo CA, Mazer BD. Induction of Regulatory T Cells by Intravenous Immunoglobulin: A Bridge between Adaptive and Innate Immunity. Front Immunol 2015; 6:469. [PMID: 26441974 PMCID: PMC4566032 DOI: 10.3389/fimmu.2015.00469] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/28/2015] [Indexed: 12/25/2022] Open
Abstract
Intravenous immunoglobulin (IVIg) is a polyclonal immunoglobulin G preparation with potent immunomodulatory properties. The mode of action of IVIg has been investigated in multiple disease states, with various mechanisms described to account for its benefits. Recent data indicate that IVIg increases both the number and the suppressive capacity of regulatory T cells, a subpopulation of T cells that are essential for immune homeostasis. IVIg alters dendritic cell function, cytokine and chemokine networks, and T lymphocytes, leading to development of regulatory T cells. The ability of IVIg to influence Treg induction has been shown both in animal models and in human diseases. In this review, we discuss data on the potential mechanisms contributing to the interaction between IVIg and the regulatory T-cell compartment.
Collapse
Affiliation(s)
- Gabriel N Kaufman
- Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada
| | - Amir H Massoud
- Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada ; Laboratory of Cellular and Molecular Immunology, University of Montreal Hospital Research Centre , Montreal, QC , Canada
| | - Marieme Dembele
- Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada
| | - Madelaine Yona
- Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada
| | - Ciriaco A Piccirillo
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada
| | - Bruce D Mazer
- Translational Research in Respiratory Diseases Program, The Research Institute of the McGill University Health Centre , Montreal, QC , Canada ; Department of Pediatrics, Faculty of Medicine, McGill University , Montreal, QC , Canada
| |
Collapse
|
10
|
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: 82] [Impact Index Per Article: 8.2] [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
|
11
|
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
|
12
|
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
|
13
|
Jackson CM, Lim M, Drake CG. Immunotherapy for brain cancer: recent progress and future promise. Clin Cancer Res 2014; 20:3651-9. [PMID: 24771646 DOI: 10.1158/1078-0432.ccr-13-2057] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immunotherapy is emerging as the newest pillar of cancer treatment, with the potential to assume a place alongside surgical debulking, radiotherapy, and chemotherapy. Early experiences with antitumor vaccines demonstrated the feasibility and potential efficacy of this approach, and newer agents, such as immune checkpoint blocking antibodies and modern vaccine platforms, have ushered in a new era. These efforts are headlined by work in melanoma, prostate cancer, and renal cell carcinoma; however, substantial progress has been achieved in a variety of other cancers, including high-grade gliomas. A recurrent theme of this work is that immunotherapy is not a one-size-fits-all solution. Rather, dynamic, tumor-specific interactions within the tumor microenvironment continually shape the immunologic balance between tumor elimination and escape. High-grade gliomas are a particularly fascinating example. These aggressive, universally fatal tumors are highly resistant to radiotherapy and chemotherapy and inevitably recur after surgical resection. Located in the immune-privileged central nervous system, high-grade gliomas also use an array of defenses that serve as direct impediments to immune attack. Despite these challenges, vaccines have shown activity against high-grade gliomas, and anecdotal, preclinical, and early clinical data bolster the notion that durable remission is possible with immunotherapy. Realizing this potential, however, will require an approach tailored to the unique aspects of glioma biology.
Collapse
Affiliation(s)
| | - Michael Lim
- Authors' Affiliations: Departments of Neurosurgery and
| | - Charles G Drake
- Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
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: 481] [Impact Index Per Article: 43.7] [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
|
15
|
Feller L, Altini M, Khammissa R, Chandran R, Bouckaert M, Lemmer J. Oral mucosal immunity. Oral Surg Oral Med Oral Pathol Oral Radiol 2013; 116:576-83. [DOI: 10.1016/j.oooo.2013.07.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/20/2013] [Accepted: 07/17/2013] [Indexed: 12/15/2022]
|
16
|
Sun JB, Xiang Z, Smith KGC, Holmgren J. Important role for FcγRIIB on B lymphocytes for mucosal antigen-induced tolerance and Foxp3+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:4412-22. [PMID: 24038083 DOI: 10.4049/jimmunol.1301324] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FcγRIIB, the only FcγR expressed on B cells, is important in the maintenance of immunological tolerance to self-Ags. In this study, we investigated the role of FcγRIIB in Ag-specific CD4 T cell tolerance induced by mucosally administered Ag (OVA) coupled to cholera toxin B subunit (Ag/CTB) or given alone. We found that sublingual administration of Ag/CTB conjugate or intragastric administration of a >100-fold higher dose of Ag alone efficiently suppressed parenteral immunization-induced Ag-specific T cell proliferation and delayed-type hypersensitivity responses in FcγRIIB-expressing wild-type (WT), but not FcγRIIB(-/-), mice. Such mucosally induced tolerance (oral tolerance) associated with induction of Ag-specific Foxp3(+) regulatory T cells was restored in FcγRIIB(-/-) mice by adoptive transfer of either WT B cells or WT dendritic cells before the mucosal Ag/CTB treatment; it was even more pronounced in μMT mice that received FcγRIIB-overexpressing B cells before treatment. Furthermore, cell transfer in either WT or μMT mice of WT but not FcγRIIB(-/-) B cells pretreated for 1 h in vitro with Ag/CTB conjugate induced Ag-specific immunological tolerance, which was further enhanced by adoptive transfer of WT B cells pretreated with anti-Ag IgG immune complexed Ag/CTB. We conclude that FcγRIIB expression on B cells, in addition to dendritic cells, is important for mucosal induction of Ag-specific immune tolerance.
Collapse
Affiliation(s)
- Jia-Bin Sun
- University of Gothenburg Vaccine Institute, SE405 30 Göteborg, Sweden
| | | | | | | |
Collapse
|
17
|
Laman JD, Weller RO. Drainage of cells and soluble antigen from the CNS to regional lymph nodes. J Neuroimmune Pharmacol 2013; 8:840-56. [PMID: 23695293 PMCID: PMC7088878 DOI: 10.1007/s11481-013-9470-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/28/2013] [Indexed: 12/25/2022]
Abstract
Despite the absence of conventional lymphatics, there is efficient drainage of both cerebrospinal fluid (CSF) and interstitial fluid (ISF) from the CNS to regional lymph nodes. CSF drains from the subarachnoid space by channels that pass through the cribriform plate of the ethmoid bone to the nasal mucosa and cervical lymph nodes in animals and in humans; antigen presenting cells (APC) migrate along this pathway to lymph nodes. ISF and solutes drain from the brain parenchyma to cervical lymph nodes by a separate route along 100–150 nm wide basement membranes in the walls of cerebral capillaries and arteries. This pathway is too narrow for the migration of APC so it is unlikely that APC traffic directly from brain parenchyma to lymph nodes by this route. We present a model for the pivotal involvement of regional lymph nodes in immunological reactions of the CNS. The role of regional lymph nodes in immune reactions of the CNS in virus infections, the remote influence of the gut microbiota, multiple sclerosis and stroke are discussed. Evidence is presented for the role of cervical lymph nodes in the induction of tolerance and its influence on neuroimmunological reactions. We look to the future by examining how nanoparticle technology will enhance our understanding of CNS-lymph node connections and by reviewing the implications of lymphatic drainage of the brain for diagnosis and therapy of diseases of the CNS ranging from neuroimmunological disorders to dementias. Finally, we review the challenges and opportunities for progress in CNS-lymph node interactions and their involvement in disease processes.
Collapse
Affiliation(s)
- Jon D. Laman
- Department of Immunology, room NB-1148a Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Roy O. Weller
- Clinical Neurosciences, Faculty of Medicine, Southampton University, Mailpoint 813, Southampton General Hospital, Southampton, SO16 6YD UK
| |
Collapse
|
18
|
Birmingham JM, Patil S, Li XM, Busse PJ. The effect of oral tolerance on the allergic airway response in younger and aged mice. J Asthma 2013; 50:122-32. [PMID: 23298269 DOI: 10.3109/02770903.2012.753455] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The effect of increased age on the induction of oral tolerance by low-dose antigen feeding and its effect on the response to antigen airway challenge in aged mice have not been well characterized. OBJECTIVE To determine whether oral tolerance can be induced in aged mice and its impact on the development of allergic airway inflammation. METHODS Younger (6 weeks old) and aged (18 months old) mice were fed ovalbumin (OVA) prior to sensitization to induce antigen tolerance. Serum antigen-specific immunoglobulins (Igs), bronchoalveolar lavage fluid (BALF), lung histology, enumeration of CD4 + Foxp3+ Treg cells, and airway hyperresponsiveness (AHR) were determined after the final antigen challenge. RESULTS Feeding antigen to aged mice prior to sensitization induced oral tolerance as determined by a decrease in antigen-specific IgE and IgG(1); however, the effect was greater in younger mice. Induction of oral tolerance was associated with a greater increase in airway Treg cells in the younger mice. Despite these differences, oral tolerance significantly suppressed features of asthma in aged mice, including BALF total cell and eosinophil numbers, cytokine production, and AHR. CONCLUSIONS Aged mice developed oral tolerance to antigen, which suppressed several features of allergic airway inflammation.
Collapse
Affiliation(s)
- Janette M Birmingham
- Division of Clinical Immunology, Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | |
Collapse
|
19
|
Williams JW, Tjota MY, Sperling AI. The contribution of allergen-specific IgG to the development of th2-mediated airway inflammation. J Allergy (Cairo) 2012; 2012:236075. [PMID: 23150737 PMCID: PMC3485540 DOI: 10.1155/2012/236075] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 09/18/2012] [Indexed: 01/01/2023] Open
Abstract
In both human asthmatics and animal models of allergy, allergen-specific IgG can contribute to Th2-mediated allergic inflammation. Mouse models have elucidated an important role for IgG and Fc-gamma receptor (FcγR) signaling on antigen presenting cells (APC) for the induction of airway inflammation. These studies suggest a positive feedback loop between IgG produced by the adaptive B cell response and FcγR signaling on innate immune cells. Studies of IgG and FcγRs in humans with asthma or allergic lung disease have been more controversial. Some reports have identified associations between allergen-specific IgG and severity of allergic responses, while other studies have found associations of IgG subclass IgG4 with allergic tolerance. In this paper, we review the literature to help define the nature of IgG and FcγR signaling on innate immune cells and how it contributes to the development of allergic immune responses.
Collapse
Affiliation(s)
- Jesse W. Williams
- Committee on Molecular Pathology and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Melissa Y. Tjota
- Interdisciplinary Scientist Training Program and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Anne I. Sperling
- Committee on Molecular Pathology and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
- Interdisciplinary Scientist Training Program and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
20
|
Rate A, Bosco A, McKenna KL, Holt PG, Upham JW. Airway epithelial cells condition dendritic cells to express multiple immune surveillance genes. PLoS One 2012; 7:e44941. [PMID: 22984588 PMCID: PMC3439377 DOI: 10.1371/journal.pone.0044941] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 08/14/2012] [Indexed: 01/18/2023] Open
Abstract
Increasing evidence suggests that crosstalk between airway epithelial cells (AEC) and adjacent dendritic cells (DC) tightly regulates airway mucosal DC function in steady state. AEC are known to express multiple immmuno-modulatory factors, though detailed information on how this influences human DC function remains incomplete. We recently demonstrated using an in vitro coculture model that AEC alter differentiation of monocytes into DC in a manner that inhibits expression of potentially damaging Th2 effector function. In the current study, we have extended these findings to examine other aspects of DC function. Using micro-array technology we show that multiple genes important for immune surveillance are significantly over expressed in purified AEC-conditioned DC, compared to control DC. These findings were confirmed by quantitative real time PCR or flow cytometry in an independent sample set. In particular, AEC-conditioned DC showed selective upregulation of chemokines that recruit Th1 cells, but minimal change in chemokines linked to Th2 cell recruitment. AEC-conditioned DC were also characterized by enhanced expression of complement family genes (C1QB, C2, CD59 and SERPING1), Fcγ receptor genes (FCGR1A, FCGR2A, FCGR2B and FCGR2C), signaling lymphocytic activation molecule family member 1 (SLAM), programmed death ligands 1 and 2, CD54 and CD200R1, relative to control DC. These findings suggest that AEC conditioning facilitates the capacity of DC to react to danger signals, to enhance leukocyte recruitment, especially of Th1 effector cells, and to interact with other immune cell populations while minimizing the risks of excessive inflammation leading to tissue damage.
Collapse
Affiliation(s)
- Angela Rate
- Telethon Institute for Child Health Research, and Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Anthony Bosco
- Telethon Institute for Child Health Research, and Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Kathy L. McKenna
- Telethon Institute for Child Health Research, and Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Patrick G. Holt
- Telethon Institute for Child Health Research, and Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - John W. Upham
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
21
|
Development of mucosal immunity in children: a rationale for sublingual immunotherapy? J Allergy (Cairo) 2011; 2012:492761. [PMID: 22121386 PMCID: PMC3205711 DOI: 10.1155/2012/492761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 08/22/2011] [Indexed: 01/14/2023] Open
Abstract
The mucosal immune system has bidirectional tasks to mount an effective defense against invading harmful pathogens and to suppress the immune response to alimentary antigens and commensal bacterial flora. Oral tolerance is a suppression of the mucosal immune pathway related to a specific immunophenotype of the dendritic cells and an induction of the regulatory T cells as well as with the silencing of the effector T cell response by anergy and deletion. The physiological dynamic process of the anatomical and functional maturation of the immune system occurring in children during pre- and postnatal periods is a significant factor, having an impact on the fine balance between the activation and the suppression of the immune response. In this paper, mechanisms of mucosal immunity and tolerance induction in terms of maturational issues are discussed with a special emphasis on the implications for a novel therapeutic intervention in allergic diseases via the sublingual route.
Collapse
|
22
|
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
|
23
|
Abstract
The term oral (or mucosal) tolerance has been classically defined as the suppression of T- and B-cell responses to an antigen by prior administration of the antigen by the oral route. In recent years, it has become clear that both innate and acquired regulatory immune responses are essential for the development of oral tolerance. As such, mucosal microenvironmental factors such as transforming growth factor- β, prostaglandins but also dietary vitamin A create conditioning of an adaptive regulatory T-cell response that suppresses subsequent antigen-specific responses. Particular resident subsets of antigen presenting dendritic cells are pivotal to convey conditioning signals next to the presentation of antigen. This review discusses the primary mechanisms of adaptive regulatory T-cell induction to ingested soluble protein antigen. However, we also discuss the limitations of our knowledge with respect to understanding the very common food hypersensitivity Celiac disease caused by an aberrant adaptive immune response to the food protein gluten.
Collapse
Affiliation(s)
- M F du Pré
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, ErasmusMC University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
24
|
van Dieren JM, Simons-Oosterhuis Y, Raatgeep HCR, Lindenbergh-Kortleve DJ, Lambers MEH, van der Woude CJ, Kuipers EJ, Snoek GT, Potman R, Hammad H, Lambrecht BN, Samsom JN, Nieuwenhuis EES. Anti-inflammatory actions of phosphatidylinositol. Eur J Immunol 2011; 41:1047-57. [PMID: 21360703 DOI: 10.1002/eji.201040899] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 01/11/2011] [Accepted: 01/20/2011] [Indexed: 01/02/2023]
Abstract
Chronic inflammatory T-cell-mediated diseases such as inflammatory bowel disease (IBD) are often treated with immunosuppressants including corticosteroids. In addition to the intended T-cell suppression, these farmacons give rise to many side effects. Recently, immunosuppressive phospholipids have been proposed as less-toxic alternatives. We aimed to investigate the immunoregulatory capacities of the naturally occurring phospholipid phosphatidylinositol (PI). Systemic PI treatment dramatically reduced disease severity and intestinal inflammation in murine 2,4,6-trinitrobenzene sulfonic acid (TNBS) colitis. Moreover, PI treatment inhibited the inflammatory T-cell response in these mice, as T cells derived from colon-draining LN of PI-treated mice secreted less IL-17 and IFN-γ upon polyclonal restimulation when compared to those of saline-treated mice. Further characterization of the suppressive capacity of PI revealed that the phospholipid suppressed Th cell differentiation in vitro irrespective of their cytokine profile by inhibiting proliferation and IL-2 release. In particular, PI diminished IL-2 mRNA expression and inhibited ERK1-, ERK-2-, p38- and JNK-phosphorylation. Crucially, PI did not ablate Treg differentiation or the antigen-presenting capacity of DCs in vitro. These data validate PI as a pluripotent inhibitor that can be applied mucosally as well as systemically. Its compelling functions render PI a promising novel physiological immune suppressant.
Collapse
Affiliation(s)
- Jolanda M van Dieren
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zwiers A, Fuss IJ, Seegers D, Konijn T, Garcia-Vallejo JJ, Samsom JN, Strober W, Kraal G, Bouma G. A polymorphism in the coding region of Il12b promotes IL-12p70 and IL-23 heterodimer formation. THE JOURNAL OF IMMUNOLOGY 2011; 186:3572-80. [PMID: 21321105 DOI: 10.4049/jimmunol.1001330] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
IL-12 and IL-23 are heterodimeric cytokines involved in the induction of Th1 and Th17 immune responses. Previous work indicated that a region on chromosome 11 encoding the IL-12p40 subunit regulates strain differences in susceptibility to murine trinitrobenzene sulfonic acid-induced colitis. In addition, this region determines strain differences in LPS-induced IL-12 responses. In this study, we investigated how polymorphisms in the coding region of murine Il12b influence IL-12 and IL-23 heterodimer formation. Transfection studies using constructs containing IL-12p35 linked to IL-12p40 from the colitis-resistant C57BL/6 strain or to the polymorphic p40 variant from the colitis-susceptible SJL/J strain demonstrated that SJL/J-derived p40 constructs synthesized significantly more IL-12p70 than did constructs harboring the C57BL/6-p40 variant. This could not be attributed to differences in synthesis rate or secretion, implicating a greater affinity of SJL/J-derived IL-12p40 for its IL-12p35 subunit. This greater affinity is also associated with increased IL-23 synthesis. In addition, C57BL/6 mice transgenic for the SJL/J 40 variant synthesized significantly more IL-12p70 upon LPS challenge and were more prone to develop colonic inflammation than did C57BL/6 mice transgenic for the C57BL/6-p40 variant. The more efficient binding of the polymorphic Il12b variant to p35 and p19 is most likely due to conformational changes following differential glycosylation as a consequence of the polymorphism. The high synthesis rate of the mature cytokines resulting from this efficient binding can lead to rapid proinflammatory skewing of immune responses and distortion of the homeostatic balance underlying the greater susceptibility for colitis.
Collapse
Affiliation(s)
- Antonie Zwiers
- Department of Gastroenterology and Hepatology, Vrije Universiteit Medical Center, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mosconi E, Rekima A, Seitz-Polski B, Kanda A, Fleury S, Tissandie E, Monteiro R, Dombrowicz DD, Julia V, Glaichenhaus N, Verhasselt V. Breast milk immune complexes are potent inducers of oral tolerance in neonates and prevent asthma development. Mucosal Immunol 2010; 3:461-74. [PMID: 20485331 DOI: 10.1038/mi.2010.23] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Allergic asthma is a chronic lung disease resulting from an inappropriate T helper (Th)-2 response to environmental antigens. Early tolerance induction is an attractive approach for primary prevention of asthma. Here, we found that breastfeeding by antigen-sensitized mothers exposed to antigen aerosols during lactation induced a robust and long-lasting antigen-specific protection from asthma. Protection was more profound and persistent than the one induced by antigen-exposed non-sensitized mothers. Milk from antigen-exposed sensitized mothers contained antigen-immunoglobulin (Ig) G immune complexes that were transferred to the newborn through the neonatal Fc receptor resulting in the induction of antigen-specific FoxP3(+) CD25(+) regulatory T cells. The induction of oral tolerance by milk immune complexes did not require the presence of transforming growth factor-beta in milk in contrast to tolerance induced by milk-borne free antigen. Furthermore, neither the presence of IgA in milk nor the expression of the inhibitory FcgammaRIIb in the newborn was required for tolerance induction. This study provides new insights on the mechanisms of tolerance induction in neonates and highlights that IgG immune complexes found in breast milk are potent inducers of oral tolerance. These observations may pave the way for the identification of key factors for primary prevention of immune-mediated diseases such as asthma.
Collapse
|
27
|
Shirinbak S, Taher YA, Maazi H, Gras R, van Esch BCAM, Henricks PAJ, Samsom JN, Verbeek JS, Lambrecht BN, van Oosterhout AJM, Nawijn MC. Suppression of Th2-driven airway inflammation by allergen immunotherapy is independent of B cell and Ig responses in mice. THE JOURNAL OF IMMUNOLOGY 2010; 185:3857-65. [PMID: 20802147 DOI: 10.4049/jimmunol.0903909] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Allergen-specific immunotherapy (IT) uniquely renders long-term relief from allergic symptoms and is associated with elevated serum levels of allergen-specific IgG and IgA. The allergen-specific IgG response induced by IT treatment was shown to be critical for suppression of the immediate phase of the allergic response in mice, and this suppression was partially dependent on signaling through FcγRIIB. To investigate the relevance of the allergen-specific IgG responses for suppression of the Th2-driven late-phase allergic response, we performed IT in a mouse model of allergic asthma in the absence of FcγRIIB or FcγRI/FcγRIII signaling. We found that suppression of Th2 cell activity, allergic inflammation, and allergen-specific IgE responses is independent of FcγRIIB and FcγRI/FcγRIII signaling. Moreover, we show that the IT-induced allergen-specific systemic IgG or IgA responses and B cell function are dispensable for suppression of the late-phase allergic response by IT treatment. Finally, we found that the secretory mucosal IgA response also is not required for suppression of the Th2-driven allergic inflammation by IT. These data are in contrast to the suppression of the immediate phase of the allergic response, which is critically dependent on the induced allergen-specific serum IgG response. Hence, IT-induced suppression of the immediate and late phases of the allergic response is governed by divergent and independent mechanisms. Our data show that the IT-induced suppression of the Th2 cell-dependent late-phase allergic response is independent of the allergen-specific IgG and IgA responses that are associated with IT treatment.
Collapse
Affiliation(s)
- Soheila Shirinbak
- Laboratory of Allergology and Pulmonary Diseases, Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD Research, University Medical Center Groningen, University of Groningen, Groningen
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Rha YH, Choi SH. The effects of early allergen/endotoxin exposure on subsequent allergic airway inflammation to allergen in mouse model of asthma. KOREAN JOURNAL OF PEDIATRICS 2010. [DOI: 10.3345/kjp.2010.53.4.481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Yeong-Ho Rha
- Departement of Pediatrics, School of Medicine, Kyunghee University, Seoul, Korea
| | - Sun-Hee Choi
- Departement of Pediatrics, School of Medicine, Kyunghee University, Seoul, Korea
| |
Collapse
|
29
|
van Dieren JM, Lambers MEH, Kuipers EJ, Samsom JN, van der Woude CJ, Nieuwenhuis EES. Local immune regulation of mucosal inflammation by tacrolimus. Dig Dis Sci 2010; 55:2514-9. [PMID: 19949865 PMCID: PMC2914281 DOI: 10.1007/s10620-009-1047-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Accepted: 10/27/2009] [Indexed: 12/09/2022]
Abstract
PURPOSE Tacrolimus is a potent immunomodulator that is effective in the treatment of inflammatory bowel disease (IBD). However, potential toxicity and systemic effects with oral intake limit its use. Local tacrolimus treatment is effective in a subgroup of proctitis patients. This study aimed to evaluate whether colonic mucosal immune cells are susceptible to locally applied tacrolimus in vitro. Our in vivo studies aimed at evaluating whether local tacrolimus treatment in mice would bring about local immune suppression and to compare colonic and systemic tacrolimus levels after locally and systemically applied tacrolimus. RESULTS In vitro tacrolimus inhibited the activation of multiple cell types present in colonic tissue; lamina propria T cells, NKT cells, and both classical- and non- classical antigen presenting cells. However, the cytokine production of epithelial cells was not inhibited by tacrolimus at these concentrations. After rectal administration in mice, tacrolimus blood levels were comparable to those obtained by oral intake. However, rectally treated mice exhibited a 14-fold higher concentration of tacrolimus within their colonic tissue than orally treated mice. Moreover, rectally applied tacrolimus resulted in a local but not a systemic immune suppression in mice. CONCLUSIONS Tacrolimus inhibits activation of several pivotal immune cells of the intestinal mucosa. Murine studies indicate that colonic application of tacrolimus induces local rather than systemic immune suppression.
Collapse
Affiliation(s)
- Jolanda M. van Dieren
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, ‘s Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands ,Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Margaretha E. H. Lambers
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Ernst J. Kuipers
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, ‘s Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Janneke N. Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - C. Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, ‘s Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Edward E. S. Nieuwenhuis
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus MC, University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| |
Collapse
|
30
|
Abboud G, Staumont-Sallé D, Kanda A, Roumier T, Deruytter N, Lavogiez C, Fleury S, Rémy P, Papin JP, Capron M, Dombrowicz D. Fc(epsilon)RI and FcgammaRIII/CD16 differentially regulate atopic dermatitis in mice. THE JOURNAL OF IMMUNOLOGY 2009; 182:6517-26. [PMID: 19414806 DOI: 10.4049/jimmunol.0801055] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The high-affinity IgE receptor Fc(epsilon)RI and, in some models, the low-affinity IgG receptor Fc(epsilon)RIIII/CD16 play an essential role in allergic diseases. In human skin, they are present on APCs and effector cells recruited into the inflamed dermis. FcRgamma is a subunit shared, among other FcRs, by Fc(epsilon)RI and CD16 and is essential to their assembly and signal transduction. Using an experimental model reproducing some features of human atopic dermatitis and specific FcR-deficient mice, we have herein delineated the respective contribution of Fc(epsilon)RIand Fc(epsilon)RIII/CD16 to the pathology. We demonstrate that symptoms of atopic dermatitis are completely absent in FcRgamma-deficient animals but only partially inhibited in either Fc(epsilon)RI- or FcgammaRIII/CD16-deficient animals. Absence or attenuation of the pathology is correlated to increased skin expression of regulatory IL-10 and Foxp3. While Fc(epsilon)RI controls both Th1 and Th2 skin response, mast cell recruitment into draining lymph nodes and IgE production, CD16 regulates only Th2 skin response, as well as T cell proliferation and IgG1 production. This isotype-specific regulation by the cognate FcR is associated to a differential regulation of IL-4 and IL-21 expression in the draining lymph nodes. Fc(epsilon)RIand CD16 thus contribute to atopic dermatitis but differentially regulate immune responses associated with the disease. Targeting both IgE/Fc(epsilon)RI and IgG/CD16 interactions might represent an efficient therapeutic strategy for allergic diseases.
Collapse
Affiliation(s)
- Georges Abboud
- Institut National de la Santé et de la Recherche Médicale Unité 547, Institut Pasteur de Lille, and Université Lille 2, Lille, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Broere F, du Pré MF, van Berkel LA, Garssen J, Schmidt-Weber CB, Lambrecht BN, Hendriks RW, Nieuwenhuis EES, Kraal G, Samsom JN. Cyclooxygenase-2 in mucosal DC mediates induction of regulatory T cells in the intestine through suppression of IL-4. Mucosal Immunol 2009; 2:254-64. [PMID: 19262503 DOI: 10.1038/mi.2009.2] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Oral intake of protein leads to tolerance through the induction of regulatory T cells (Tr cells) in mesenteric lymph nodes (MLNs). Here we show that the inhibition of cyclooxygenase-2 (COX-2) in vivo suppressed oral tolerance and was associated with enhanced differentiation of interleukin (IL)-4-producing T cells and reduced Foxp3(+) Tr-cell differentiation in MLN. As a result, the functional suppressive capacity of these differentiated mucosal T cells was lost. IL-4 was causally related to loss of tolerance as treatment of mice with anti-IL-4 antibodies during COX-2 inhibition restored tolerance. Dendritic cells (DCs) in the MLN differentially expressed COX-2 and reductionist experiments revealed that selective inhibition of the enzyme in these cells inhibited Foxp3(+) Tr-cell differentiation in vitro. Importantly, the inhibition of COX-2 in MLN-DC caused increased GATA-3 expression and enhanced IL-4 release by T cells, which was directly related to impaired Tr-cell differentiation. These data provide crucial insights into the mechanisms driving de novo Tr-cell induction and tolerance in the intestine.
Collapse
Affiliation(s)
- F Broere
- Department of Molecular Cell Biology and Immunology, VU Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Oral tolerance: intestinal homeostasis and antigen-specific regulatory T cells. Trends Immunol 2008; 29:532-40. [DOI: 10.1016/j.it.2008.09.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Revised: 08/26/2008] [Accepted: 09/01/2008] [Indexed: 01/27/2023]
|
33
|
He J, Zhao J, Li Z. Mucosal administration of alpha-fodrin inhibits experimental Sjögren's syndrome autoimmunity. Arthritis Res Ther 2008; 10:R44. [PMID: 18419828 PMCID: PMC2453764 DOI: 10.1186/ar2403] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2007] [Revised: 03/26/2008] [Accepted: 04/18/2008] [Indexed: 11/20/2022] Open
Abstract
Introduction α-Fodrin is an autoantigen in Sjögren's syndrome. We hypothesized that mucosal administration of α-fodrin might prevent the disease. Methods Four-week-old NOD mice were immunized (intranasal) with a 1 μg or 10 μg dose of α-fodrin every other day. PBS 10 μl/dose and Glutathione transferase (GST 10 μg/dose (control mice) were intranasally administrated by the same procedure. The salivary flow was maintained in immunized animals. The animals were analyzed for the presence of anti-Sjögren's syndrome A, anti-Sjögren's syndrome B, rheumatoid factor and antinuclear, anti-α-fodrin, and anti-type 3 muscarinic acetylcholine receptor polypeptide (anti-M3RP) by immunofluorescence or ELISA. The cytokines IFNγ and IL-10 were measured by ELISA. Salivary glands were examined by H&E staining and immunohistochemical analysis. The water-volume intake was calculated for each group. The induction of regulatory T cells was assessed by fluorescence-activated cell sorting analysis for the frequency of Foxp3+ cells among peripheral CD4+CD25+ T cells. Results The appearance of anti-α-fodrin and anti-M3RP antibodies was delayed in mice immunized with α-fodrin. The titers of anti-α-fodrin and anti-M3RP antibodies were lower in immunized mice (P < 0.05), but there was no significant difference between the low-dose or high-dose immunization groups. Five out of eight mice in the GST group, five of eight mice in the PBS group, two of eight mice in the α-fodrin 1 μg/dose group, and three out of eight mice in the α-fodrin 10 μg/dose were positive for antinuclear antibodies. The levels of serum IFNγ in mice immunized with 1 μg/dose or 10 μg/dose α-fodrin, with PBS, and with GST were 41.9 ± 16.2 pg/ml, 37.1 ± 15.4 pg/ml, 86.8 ± 17.8 pg/ml and 71.6 ± 11.1 pg/ml, respectively, while we found no difference in the levels of serum IL-10 among the groups. The number of Foxp3+ CD4+CD25+ regulatory T cells was higher in the α-fodrin groups compared with the PBS and GST control groups (P < 0.05). Lymphocytic infiltration and expression of α-fodrin in the salivary glands was decreased in α-fodrin-treated groups. The fluid intake of mice in the 1 μg/dose α-fodrin, 10 μg/dose α-fodrin, PBS, and GST groups was 39.2 ± 2.1 ml, 40.4 ± 2.5 ml, 49.3 ± 3.1 ml and 51.6 ± 2.8 ml, respectively. Conclusion Mucosal administration of α-fodrin effectively inhibited the progression of experimental Sjögren's syndrome autoimmunity.
Collapse
Affiliation(s)
- Jing He
- Department of Rheumatology & Immunology, People's Hospital, Peking University Medical School, 11 Xizhimen South Street, Beijing 100044, China.
| | | | | |
Collapse
|
34
|
Rynda A, Maddaloni M, Mierzejewska D, Ochoa-Repáraz J, Maślanka T, Crist K, Riccardi C, Barszczewska B, Fujihashi K, McGhee JR, Pascual DW. Low-dose tolerance is mediated by the microfold cell ligand, reovirus protein sigma1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:5187-200. [PMID: 18390700 PMCID: PMC2629740 DOI: 10.4049/jimmunol.180.8.5187] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mucosal tolerance induction generally requires multiple or large Ag doses. Because microfold (M) cells have been implicated as being important for mucosal tolerance induction and because reovirus attachment protein sigma1 (psigma1) is capable of binding M cells, we postulated that targeting a model Ag to M cells via psigma1 could induce a state of unresponsiveness. Accordingly, a genetic fusion between OVA and the M cell ligand, reovirus psigma1, termed OVA-psigma1, was developed to enhance tolerogen uptake. When applied nasally, not parenterally, as little as a single dose of OVA-psigma1 failed to induce OVA-specific Abs even in the presence of adjuvant. Moreover, the mice remained unresponsive to peripheral OVA challenge, unlike mice given multiple nasal OVA doses that rendered them responsive to OVA. The observed unresponsiveness to OVA-psigma1 could be adoptively transferred using cervical lymph node CD4(+) T cells, which failed to undergo proliferative or delayed-type hypersensitivity responses in recipients. To discern the cytokines responsible as a mechanism for this unresponsiveness, restimulation assays revealed increased production of regulatory cytokines, IL-4, IL-10, and TGF-beta1, with greatly reduced IL-17 and IFN-gamma. The induced IL-10 was derived predominantly from FoxP3(+)CD25(+)CD4(+) T cells. No FoxP3(+)CD25(+)CD4(+) T cells were induced in OVA-psigma1-dosed IL-10-deficient (IL-10(-/-)) mice, and despite showing increased TGF-beta1 synthesis, these mice were responsive to OVA. These data demonstrate the feasibility of using psigma1 as a mucosal delivery platform specifically for low-dose tolerance induction.
Collapse
Affiliation(s)
- Agnieszka Rynda
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Massimo Maddaloni
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Dagmara Mierzejewska
- Department of Food Chemistry, Institute of Food Research, Polish Academy of Science, Olsztyn, Poland
| | - Javier Ochoa-Repáraz
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Tomasz Maślanka
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Kathryn Crist
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Carol Riccardi
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Beata Barszczewska
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| | - Kohtaro Fujihashi
- Departments of Microbiology and Pediatric Dentistry, Immunobiology Vaccine Center, University of Alabama at Birmingham, Birmingham AL 35294
| | - Jerry R. McGhee
- Departments of Microbiology and Pediatric Dentistry, Immunobiology Vaccine Center, University of Alabama at Birmingham, Birmingham AL 35294
| | - David W. Pascual
- Veterinary Molecular Biology, Montana State University, 960 Technology Blvd. Bozeman, MT 59718
| |
Collapse
|
35
|
Novak N, Haberstok J, Bieber T, Allam JP. The immune privilege of the oral mucosa. Trends Mol Med 2008; 14:191-8. [PMID: 18396104 DOI: 10.1016/j.molmed.2008.03.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 02/25/2008] [Accepted: 03/04/2008] [Indexed: 01/22/2023]
Abstract
Despite high bacterial colonization and frequent allergen contact, acute inflammatory and allergic reactions are rarely seen in the oral mucosa. Therefore we assert that immune tolerance predominates at this site and antigen presenting cells, such as dendritic cells and different T cell subtypes, serve as key players in oral mucosal tolerance induction. In this article we describe the mechanisms that lead to tolerance induced in the oral mucosa and how they differ from tolerance induced in the lower gastrointestinal tract. Furthermore we discuss ways in which novel nonparenteral approaches for immune intervention, such as allergen-specific immunotherapy applied by way of the sublingual route, might be improved to target the tolerogenic properties of the sophisticated oral mucosal immune network.
Collapse
Affiliation(s)
- Natalija Novak
- Department of Dermatology and Allergy, University of Bonn, Sigmund-Freud-Str 25, 53105 Bonn, Germany.
| | | | | | | |
Collapse
|
36
|
Samsom JN, van der Marel APJ, van Berkel LA, van Helvoort JMLM, Simons-Oosterhuis Y, Jansen W, Greuter M, Nelissen RLH, Meeuwisse CML, Nieuwenhuis EES, Mebius RE, Kraal G. Secretory leukoprotease inhibitor in mucosal lymph node dendritic cells regulates the threshold for mucosal tolerance. THE JOURNAL OF IMMUNOLOGY 2007; 179:6588-95. [PMID: 17982048 DOI: 10.4049/jimmunol.179.10.6588] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The notion that the mucosal immune system maintains a tolerogenic response to harmless Ags while continually being challenged with microbial products seems an enigma. The aim of this study was to unravel mechanisms that are involved in regulating the development of tolerance under constant microbial pressure. The tolerogenic response to Ags administered via the nasal mucosa is dependent on the organized lymphoid tissue of the cervical lymph nodes (LN). We show that cervical LN differentially express secretory leukoprotease inhibitor (SLPI) compared with peripheral LN. SLPI was expressed by dendritic cells (DCs) and because SLPI is known to suppress LPS responsiveness, it was hypothesized that its expression in mucosal DCs may be required to regulate cellular activation to microbial products. Indeed, compared with wild-type controls, bone marrow-derived DCs from SLPI(-/-) mice released more inflammatory cytokines and enhanced T cell proliferation after stimulation with low dose LPS. This increased sensitivity to LPS was accompanied by increased NF-kappaB p65 activation in SLPI(-/-) DCs. In vivo, nasal application of OVA with LPS to SLPI(-/-) mice resulted in enhanced DC activation in the cervical LN reflected by increased costimulatory molecule expression and release of inflammatory cytokines. This led to failure to maintain tolerance to nasal OVA application in the presence of low doses of LPS. We propose that expression of SLPI functions as a rheostat by controlling the level of bacterial stimuli that induce mucosal DC activation. As such, it regulates the quality of the ensuing Ag-specific immune response in the mucosa draining LN.
Collapse
Affiliation(s)
- Janneke N Samsom
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
van der Marel APJ, Samsom JN, Greuter M, van Berkel LA, O'Toole T, Kraal G, Mebius RE. Blockade of IDO inhibits nasal tolerance induction. THE JOURNAL OF IMMUNOLOGY 2007; 179:894-900. [PMID: 17617580 DOI: 10.4049/jimmunol.179.2.894] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The amino acid tryptophan is essential for the proliferation and survival of cells. Modulation of tryptophan metabolism has been described as an important regulatory mechanism for the control of immune responses. The enzyme IDO degrades the indole moiety of tryptophan, not only depleting tryptophan but also producing immunomodulatory metabolites called kynurenines, which have apoptosis-inducing capabilities. In this study, we show that IDO is more highly expressed in nonplasmacytoid dendritic cells of the nose draining lymph nodes (LNs), which form a unique environment to induce tolerance to inhaled Ags, when compared with other peripheral LNs. Upon blockade of IDO during intranasal OVA administration, Ag-specific immune tolerance was abrogated. Analysis of Ag-specific T cells in the LNs revealed that inhibition of IDO resulted in enhanced survival at 48 h after antigenic stimulation, although this result was not mediated through alterations in apoptosis or cell proliferation. Furthermore, no differences were found in CD4(+) T cells expressing FoxP3. Our data suggest that the level of IDO expression in dendritic cells, present in nose draining LNs, allows for the generation of a sufficient number of regulatory T cells to control and balance effector T cells in such a way that immune tolerance is induced, whereas upon IDO blockade, effector T cells will outnumber regulatory T cells, leading to immunity.
Collapse
Affiliation(s)
- Arnold P J van der Marel
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
38
|
Bandukwala HS, Clay BS, Tong J, Mody PD, Cannon JL, Shilling RA, Verbeek JS, Weinstock JV, Solway J, Sperling AI. Signaling through Fc gamma RIII is required for optimal T helper type (Th)2 responses and Th2-mediated airway inflammation. ACTA ACUST UNITED AC 2007; 204:1875-89. [PMID: 17664287 PMCID: PMC2118666 DOI: 10.1084/jem.20061134] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Although inhibitory Fc gamma receptors have been demonstrated to promote mucosal tolerance, the role of activating Fc gamma receptors in modulating T helper type (Th)2-dependent inflammatory responses characteristic of asthma and allergies remains unclear. Here, we demonstrate that signaling via activating Fc gamma receptors in conjunction with Toll-like receptor 4 stimulation modulated cytokine production from bone marrow-derived dendritic cells (DCs) and augmented their ability to promote Th2 responses. Ligation of the low affinity receptor Fc gamma RIII was specifically required for the enhanced Th2 responses, as Fc gamma RIII(-/-) DCs failed to augment Th2-mediated airway inflammation in vivo or induce Th2 differentiation in vitro. Further, Fc gamma RIII(-/-) mice had impaired Th2 cytokine production and exhibited reduced airway inflammation, whereas no defect was found in Fc gamma RI(-/-) mice. The augmentation of Th2 immunity was regulated by interleukin 10 production from the DCs but was distinct and independent of the well-established role of Fc gamma RIII in augmenting antigen presentation. Thus, our studies reveal a novel and specific role for Fc gamma RIII signaling in the regulation of Th cell responses and suggest that in addition to immunoglobulin (Ig)E, antigen-specific IgG also contributes to the pathogenesis of Th2-mediated diseases such as asthma and allergies.
Collapse
Affiliation(s)
- Hozefa S Bandukwala
- Committee on Immunology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Desai DD, Harbers SO, Flores M, Colonna L, Downie MP, Bergtold A, Jung S, Clynes R. Fc gamma receptor IIB on dendritic cells enforces peripheral tolerance by inhibiting effector T cell responses. THE JOURNAL OF IMMUNOLOGY 2007; 178:6217-26. [PMID: 17475849 DOI: 10.4049/jimmunol.178.10.6217] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The uptake of immune complexes by FcRs on APCs augments humoral and cellular responses to exogenous Ag. In this study, CD11c+ dendritic cells are shown to be responsible in vivo for immune complex-triggered priming of T cells. We examine the consequence of Ab-mediated uptake of self Ag by dendritic cells in the rat insulin promoter-membrane OVA model and identify a role for the inhibitory FcgammaRIIB in the maintenance of peripheral CD8 T cell tolerance. Effector differentiation of diabetogenic OT-I CD8+ T cells is enhanced in rat insulin promoter-membrane OVA mice lacking FcgammaRIIB, resulting in a high incidence of diabetes. FcgammaRIIB-mediated inhibition of CD8 T cell priming results from suppression of both DC activation and cross-presentation through activating FcgammaRs. Further FcgammaRIIB on DCs inhibited the induction of OVA-specific Th1 effectors, limiting Th1-type differentiation and memory T cell accumulation. In these MHC II-restricted responses, the presence of FcgammaRIIB only modestly affected initial CD4 T cell proliferative responses, suggesting that FcgammaRIIB limited effector cell differentiation primarily by inhibiting DC activation. Thus, FcgammaRIIB can contribute to peripheral tolerance maintenance by inhibiting DC activation alone or by also limiting processing of exogenously acquired Ag.
Collapse
Affiliation(s)
- Dharmesh D Desai
- Department of Medicine and Microbiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Mast cells are effector cells of the innate immune system, but because they express Fc receptors (FcRs), they can be engaged in adaptive immunity by antibodies. Mast cell FcRs include immunoglobulin E (IgE) and IgG receptors and, among these, activating and inhibitory receptors. The engagement of mast cell IgG receptors by immune complexes may or may not trigger cell activation, depending on the type of mast cell. The coengagement of IgG and IgE receptors results in inhibition of mast cell activation. The Src homology-2 domain-containing inositol 5-phosphatase-1 is a major effector of negative regulation. Biological responses of mast cells depend on the balance between positive and negative signals that are generated in FcR complexes. The contribution of human mast cell IgG receptors in allergies remains to be clarified. Increasing evidence indicates that mast cells play critical roles in IgG-dependent tissue-specific autoimmune diseases. Convincing evidence was obtained in murine models of multiple sclerosis, rheumatoid arthritis, bullous pemphigoid, and glomerulonephritis. In these models, the intensity of lesions depended on the relative engagement of activating and inhibitory IgG receptors. In vitro models of mature tissue-specific murine mast cells are needed to investigate the roles of mast cells in these diseases. One such model unraveled unique differentiation/maturation-dependent biological responses of serosal-type mast cells.
Collapse
Affiliation(s)
- Odile Malbec
- Unité d'Allergologie Moléculaire et Cellulaire, Département d'Immunologie, Institut Pasteur, Paris, France
| | | |
Collapse
|
41
|
Verbeek R, van der Mark K, Wawrousek EF, Plomp AC, van Noort JM. Tolerization of an established alphaB-crystallin-reactive T-cell response by intravenous antigen. Immunology 2007; 121:416-26. [PMID: 17386078 PMCID: PMC2265960 DOI: 10.1111/j.1365-2567.2007.02592.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Tolerance induction to prevent activation of a naïve T-cell repertoire has been well documented in rodents and can be readily achieved by intravenous, oral or intranasal administration of antigen in the absence of adjuvants. In autoimmune diseases such as multiple sclerosis (MS) the presence of an established memory/effector T-cell repertoire against self-antigens is likely to be more relevant than the potential reactivity of naive T cells. Methods to eliminate such an established T-cell response are less well understood. In this study, we explored the effectiveness of intravenous soluble antigen to eliminate a pre-existing T-cell response against alphaB-crystallin, a candidate autoantigen in MS. We used mice that are deficient for the target antigen. This condition allowed for a vigourous T-cell and antibody response to develop upon immunization, and eliminated all possible endogenous mechanisms of tolerance for alphaB-crystallin that are found in normal rodents. When applied 3 weeks after priming with alphaB-crystallin, intravenous administration of soluble antigen almost completely abrogated the established T-cell response in a dose-dependent manner as evidenced by T-cell non-responsiveness in tolerized animals to a re-challenge with antigen in complete Freund's adjuvant. Evaluating delayed-type hypersensitivity responses after tolerance induction revealed that the tolerizing effect was achieved within 24 hr. Furthermore, the tolerizing effect was found to be antigen-specific and long lasting. In contrast, serum antibody levels were markedly increased. Our data clarify that in the absence of any natural form of immune regulation, antigen-specific memory/effector T cells can be effectively silenced by intravenous antigen.
Collapse
Affiliation(s)
- Richard Verbeek
- Department of Biosciences, TNO Quality of Life, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
42
|
Su K, Yang H, Li X, Li X, Gibson AW, Cafardi JM, Zhou T, Edberg JC, Kimberly RP. Expression profile of FcgammaRIIb on leukocytes and its dysregulation in systemic lupus erythematosus. THE JOURNAL OF IMMUNOLOGY 2007; 178:3272-80. [PMID: 17312177 PMCID: PMC2824439 DOI: 10.4049/jimmunol.178.5.3272] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FcgammaRIIb (CD32B, Online Mendelian Inheritance in Man 604590), an IgG FcR with a tyrosine-based inhibitory motif, plays a critical role in the balance of tolerance and autoimmunity in murine models. However, the high degree of homology between FcgammaRIIb and FcgammaRIIa in humans and the lack of specific Abs to differentiate them have hampered study of the normal expression profile of FcgammaRIIb and its potential dysregulation in autoimmune diseases such as systemic lupus erythematosus (SLE). Using our newly developed anti-FcgammaRIIb mAb 4F5 which does not react with FcgammaRIIa, we found that FcgammaRIIb is expressed on the cell surface of circulating B lymphocytes, monocytes, neutrophils, myeloid dendritic cells (DCs), and at very low levels on plasmacytoid DCs from some donors. Normal donors with the less frequent 2B.4 promoter haplotype have higher FcgammaRIIb expression on monocytes, neutrophils, and myeloid DCs similar to that reported for B lymphocytes, indicating that FcgammaRIIb expression on both myeloid and lymphoid cells is regulated by the naturally occurring regulatory single nucleotide polymorphisms in the FCGR2B promoter. FcgammaRIIb expression in normal controls is up-regulated on memory B lymphocytes compared with naive B lymphocytes. In contrast, in active SLE, FcgammaRIIb is significantly down-regulated on both memory and plasma B lymphocytes compared with naive and memory/plasma B lymphocytes from normals. Similar down-regulation of FcgammaRIIb on myeloid-lineage cells in SLE was not seen. Our studies demonstrate the constitutive regulation of FcgammaRIIb by natural gene polymorphisms and the acquired dysregulation in SLE autoimmunity, which may identify opportunities for using this receptor as a therapeutic target.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Autoimmunity/genetics
- Female
- Gene Expression Regulation/immunology
- Humans
- Leukocytes/immunology
- Leukocytes/metabolism
- Leukocytes/pathology
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/pathology
- Lupus Erythematosus, Systemic/therapy
- Male
- Mice
- Polymorphism, Single Nucleotide/immunology
- Promoter Regions, Genetic
- Receptors, IgG/biosynthesis
- Receptors, IgG/genetics
- Receptors, IgG/immunology
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Robert P. Kimberly
- Address correspondence and reprint requests to Dr. Robert P. Kimberly, University of Alabama at Birmingham, 1530 Third Avenue South, Shelby Interdisciplinary Biomedical Research Building 172D, Birmingham, AL 35294-2182.
| |
Collapse
|
43
|
Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol 2007; 119:780-91. [PMID: 17321578 DOI: 10.1016/j.jaci.2007.01.022] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 01/14/2007] [Accepted: 01/16/2007] [Indexed: 10/23/2022]
Abstract
Allergen-specific immunotherapy (SIT) has been used for almost a century as a desensitizing therapy for allergic diseases and represents the only curative and specific method of treatment. Administration of appropriate concentrations of allergen extracts has been shown to be reproducibly effective when patients are carefully selected. The mechanisms by which allergen-SIT has its effects include the modulation of T-cell and B-cell responses and related antibody isotypes as well as effector cells of allergic inflammation, such as eosinophils, basophils, and mast cells. The balance between allergen-specific T-regulatory (Treg) and T(H)2 cells appears to be decisive in the development of allergic and healthy immune responses against allergens. Treg cells consistently represent the dominant subset specific for common environmental allergens in sensitized healthy individuals. In contrast, there is a high frequency of allergen-specific T(H)2 cells in patients with allergy. The induction of a tolerant state in peripheral T cells represents an essential step in allergen-SIT. Peripheral T-cell tolerance is characterized mainly by generation of allergen-specific Treg cells leading to suppressed T-cell proliferation and T(H)1 and T(H)2 cytokine responses against the allergen. This is accompanied by a significant increase in allergen-specific IgG(4), and also IgG(1) and IgA, and a decrease in IgE in the late stage of the disease. In addition, decreased tissue infiltration of mast cells and eosinophils and their mediator release including circulating basophils takes place. Current understanding of mechanisms of allergen-SIT, particularly the role of Treg cells in peripheral tolerance, may enable novel treatment strategies.
Collapse
Affiliation(s)
- Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | | |
Collapse
|
44
|
Bros M, Jährling F, Renzing A, Wiechmann N, Dang NA, Sutter A, Ross R, Knop J, Sudowe S, Reske-Kunz AB. A newly established murine immature dendritic cell line can be differentiated into a mature state, but exerts tolerogenic function upon maturation in the presence of glucocorticoid. Blood 2007; 109:3820-9. [PMID: 17209058 DOI: 10.1182/blood-2006-07-035576] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract
The phenotype and function of murine dendritic cells (DCs) are primarily studied using bone-marrow–derived DCs (BM-DCs), but may be hampered by the heterogenous phenotype of BM-DCs due to their differential state of maturation. Here we characterize a newly established murine DC line (SP37A3) of myeloid origin. During maintainance in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and M-CSF, SP37A3 cells resemble immature DCs characterized by low expression of major histocompatibility complex (MHC) II and costimulatory molecules and low T-cell stimulatory capacity. Upon stimulation, SP37A3 cells acquire a mature phenotype and activate naive T cells as potently as BM-DCs. Similar to BM-DCs, SP37A3 cells activated in the presence of dexamethasone-induced regulatory T cells, which were anergic upon restimulation and suppressed proliferation of naive T cells. This tolerogenic state was reflected by lower expression levels of costimulatory molecules and proinflammatory cytokines compared with mature cells, as well as up-regulated expression of FcγRIIB and interleukin-1RA (IL-1RA). SP37A3 cells were responsive to dexamethasone even when applied at later time points during activation, suggesting functional plasticity. Thus, DC line SP37A3 represents a suitable model to study functions of immature and mature as well as tolerogenic myeloid DCs, circumventing restrictions associated with the use of primary DCs and BM-DCs.
Collapse
Affiliation(s)
- Matthias Bros
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg-University, Obere Zahlbacher Strasse 63, D-55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The immune system is organized as a number of distinct lymphoid organs interconnected by recirculating lymphocytes. These organs, such as lymph nodes, spleen, and gut-associated Peyer's patches, are compartmentalized, providing separate niches for T and B cells. In addition, regional compartmentalization of lymphoid organs themselves exists, leading to the distinction between the mucosal and the systemic immune systems. This distinction not only reflects the anatomical localization but also is based on functional differences, with predominant tolerance induction via mucosal routes and immunity seen after systemic antigen exposure. These differences are associated with regional differences in the lymphoid organs and with environmental conditions of the tissues in which the immune system functions. Recirculation patterns of lymphocytes differ between mucosal and systemic lymphoid organs, and more insight into the mechanisms that imprint this behavior has been generated recently. Differences in dendritic cells have been observed between mucosal and systemic sites, and knowledge on how local factors contribute to the immune system is emerging. From our studies on mucosal tolerance in mouse models, it has become evident that regional lymph nodes draining the mucosa are important sites to direct immune responses. Here, we discuss the way regional lymph nodes contribute to the direction of immune responses and what is known about the local factors and cell behavior that form the basis for these differences.
Collapse
Affiliation(s)
- Georg Kraal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
46
|
Nimmerjahn F. Activating and inhibitory FcγRs in autoimmune disorders. ACTA ACUST UNITED AC 2006; 28:305-19. [PMID: 17115158 DOI: 10.1007/s00281-006-0052-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 09/29/2006] [Indexed: 10/24/2022]
Abstract
Autoimmune disorders are characterized by the destruction of self-tissues by the immune system. Multiple checkpoints are in place to prevent autoreactivity under normal circumstances. Coexpression of activating and inhibitory Fc receptors (FcR) represents such a checkpoint by establishing a threshold for immune cell activation. In many human autoimmune diseases, however, balanced FcR expression is disturbed. Analysis of murine model systems provides strong evidence that aberrant FcR expression can result in uncontrolled immune responses and the initiation of autoimmune disease. This review will summarize this data and explain how this information might be used to better understand human autoimmune diseases and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Laboratory of Molecular Genetics and Immunology, 1230 York Avenue, New York, NY, 10021, USA,
| |
Collapse
|
47
|
Abstract
The only disease-modifying treatment that is available for allergic patients is allergen-specific immunotherapy. Two competing application forms are used: subcutaneous immunotherapy, which has been used for > 90 years, and a relatively new immunotherapy where the allergen is applied sublingually. Numerous studies have shown efficacy for subcutaneous immunotherapy and have identified possible mechanisms that are responsible for the observed reduction in allergic responses. In contrast, the efficacy of sublingual immunotherapy has not been documented to the same degree and the responsible immunological mechanisms have not yet been clearly defined. This review focuses on the published clinical and experimental data on sublingual immunotherapy and points at possible mechanisms of how sublingual immunotherapy may differ from subcutaneous immunotherapy in its mode of action, and also discusses the potential advantages and pit falls of both therapies.
Collapse
Affiliation(s)
- Melanie Werner-Klein
- Department of Pulmonary Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | |
Collapse
|
48
|
Abstract
Allergen immunotherapy is a well-established strategy for treating allergic diseases with the goal of inducing allergen-specific tolerance. Identified mechanisms contributing to the therapeutic effect of immunotherapy include a shift of T helper 2 (Th2)-type immune responses to a modified Th2 immune response, a change of the balance of IgE-producing B cells to the production of IgG subtypes, in addition to increased IL-10 and TGF-beta secretion and activation of the suppressive functions of regulatory T-cells. Dendritic cells (DCs), which as outposts of the immune system are capable of T-cell priming through efficient allergen uptake by IgE receptors expressed on their cell surface. Most of the hypotheses concerning the function of DCs as facilitators of allergen-specific tolerance in allergen immunotherapy remain speculative. Therefore, studies must focus on the functional changes of DCs under immunotherapy to close the gap of knowledge about their exact role. These experimental data should help confirm the hypothesis of DCs as efficient silencers and potential target cells and take advantage of the bivalent character and tolerogenic properties of DCs.
Collapse
Affiliation(s)
- Natalija Novak
- Department of Dermatology, University of Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany.
| |
Collapse
|
49
|
Ohnuki H, Mizutani A, Otani H. Oral ingestion of cow's milk immunoglobulin G stimulates some cellular immune systems and suppresses humoral immune responses in mouse. Int Immunopharmacol 2006; 6:1315-22. [PMID: 16782545 DOI: 10.1016/j.intimp.2006.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Revised: 03/10/2006] [Accepted: 04/08/2006] [Indexed: 11/16/2022]
Abstract
Four-week-old male C3H/HeN mice were bred with diets consisting of ovalbumin alone (OVA, control diet) or mixtures of OVA and cow's milk immunoglobulin G (IgG-added diets) as a protein source for 4 or 5 weeks, and both the cellular and humoral immune properties of the mice were investigated. The number of interleukin (IL)-12+CD11b+ cells in spleens and the formation of superoxide by peritoneal macrophages were higher in mice given the IgG-added diet than in those given the control diet. The number of natural killer cells in Peyer's patches or spleens and the cytotoxic activity of spleen cells toward an erythroleukemia cell line, K562, were also higher in mice given the IgG-added diet. In contrast, the numbers of interferon-gamma+CD4+ and IL-4+CD4+ cells in Peyer's patches or spleens and the levels of total or OVA-specific intestinal IgA and serum IgG were significantly lower in mice given the IgG-added diet than in those given the control diet. In addition, the number of cells expressing CD19 in spleens was significantly higher in mice given the IgG-added diet. These results indicate that oral ingestion of cow's milk IgG may stimulate some innate cellular immune systems, while suppressing humoral adaptive immune responses in the mouse.
Collapse
Affiliation(s)
- Hidetaka Ohnuki
- Integrated Department of Development of Functional Foods, Graduate School of Agriculture, Shinshu University, Minamiminowa-mura 8304, Kamiina-gun, Nagano-ken 399-4598, Japan
| | | | | |
Collapse
|
50
|
Bibliography. Current world literature. Outcome measures. Curr Opin Allergy Clin Immunol 2006; 6:241-4. [PMID: 16670521 DOI: 10.1097/01.all.0000225167.72842.fd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|