101
|
Satoh JI, Tabunoki H. Molecular network of chromatin immunoprecipitation followed by deep sequencing-based vitamin D receptor target genes. Mult Scler 2013; 19:1035-45. [PMID: 23401126 DOI: 10.1177/1352458512471873] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Vitamin D is a liposoluble vitamin essential for calcium metabolism. The ligand-bound vitamin D receptor (VDR), heterodimerized with retinoid X receptor, interacts with vitamin D response elements (VDREs) to regulate gene expression. Vitamin D deficiency due to insufficient sunlight exposure confers an increased risk for multiple sclerosis (MS). OBJECTIVE To study a protective role of vitamin D in multiple sclerosis (MS), it is important to characterize the global molecular network of VDR target genes (VDRTGs) in immune cells. METHODS We identified genome-wide VDRTGs collectively from two distinct chromatin immunoprecipitation followed by deep sequencing (ChIP-Seq) datasets of VDR-binding sites derived from calcitriol-treated human cells of B cell and monocyte origins. We mapped short reads of next generation sequencing (NGS) data on hg19 with Bowtie, detected the peaks with Model-based Analysis of ChIP-Seq (MACS), and identified genomic locations by GenomeJack, a novel genome viewer for NGS platforms. RESULTS We found 2997 stringent peaks distributed on protein-coding genes, chiefly located in the promoter and the intron on VDRE DR3 sequences. However, the corresponding transcriptome data verified calcitriol-induced upregulation of only a small set of VDRTGs. The molecular network of 1541 calcitriol-responsive VDRTGs showed a significant relationship with leukocyte transendothelial migration, Fcγ receptor-mediated phagocytosis, and transcriptional regulation by VDR, suggesting a pivotal role of genome-wide VDRTGs in immune regulation. CONCLUSION These results suggest the working hypothesis that persistent deficiency of vitamin D might perturb the complex network of VDRTGs in immune cells, being responsible for induction of an autoimmune response causative for MS.
Collapse
Affiliation(s)
- Jun-ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan.
| | | |
Collapse
|
102
|
Soyer OU, Akdis M, Ring J, Behrendt H, Crameri R, Lauener R, Akdis CA. Mechanisms of peripheral tolerance to allergens. Allergy 2013; 68:161-70. [PMID: 23253293 DOI: 10.1111/all.12085] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2012] [Indexed: 12/24/2022]
Abstract
The immune system is regulated to protect the host from exaggerated stimulatory signals establishing a state of tolerance in healthy individuals. The disequilibrium in immune regulatory vs effector mechanisms results in allergic or autoimmune disorders in genetically predisposed subjects under certain environmental conditions. As demonstrated in allergen-specific immunotherapy and in the healthy immune response to high-dose allergen exposure models in humans, T regulatory cells are essential in the suppression of Th2-mediated inflammation, maintenance of immune tolerance, induction of the two suppressive cytokines interleukin-10 and transforming growth factor-β, inhibition of allergen-specific IgE, and enhancement of IgG4 and IgA. Also, suppression of dendritic cells, mast cells, and eosinophils contributes to the construction of peripheral tolerance to allergens. This review focuses on mechanisms of peripheral tolerance to allergens with special emphasis on recent developments in the area of immune regulation.
Collapse
Affiliation(s)
| | | | | | | | - R. Crameri
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos; Switzerland
| | | | | |
Collapse
|
103
|
Akdis M, Palomares O, van de Veen W, van Splunter M, Akdis CA. TH17 and TH22 cells: a confusion of antimicrobial response with tissue inflammation versus protection. J Allergy Clin Immunol 2013; 129:1438-49; quiz1450-1. [PMID: 22657405 DOI: 10.1016/j.jaci.2012.05.003] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/02/2012] [Accepted: 05/02/2012] [Indexed: 12/30/2022]
Abstract
Substantial progress in understanding mechanisms of immune regulation in allergy, asthma, autoimmune diseases, tumors, organ transplantation, chronic infections, and pregnancy is in an exciting developmental phase that might lead to a variety of targeted therapeutic approaches. Recent progress in the interaction between immune/inflammatory cell subsets through cytokines, particularly the extension of the knowledge on reciprocal regulation and counterbalance between subsets of T(H)1, T(H)2, T(H)9, T(H)17, T(H)22, T follicular helper cells and different subsets of regulatory T cells, as well as corresponding and co-orchestrating B-cell, natural killer cell, dendritic cell, and innate lymphoid cell subsets, offers new possibilities for immune intervention. Studies on new subsets confirm the important role of T cells in the instruction of tissue cells and also demonstrate the important role of feedback regulation for the polarization toward distinct T-cell subsets. T(H)17 and T(H)22 cells are 2 emerging T(H) cell subsets that link the immune response to tissue inflammation; IL-17A and IL-17F and IL-22 are their respective prototype cytokines. Although both cytokines play roles in immune defense to extracellular bacteria, IL-17 augments inflammation, whereas IL-22 plays a tissue-protective role. This review focuses on current knowledge on T(H)17 and T(H)22 cells and their role in inflammation, with special focus on the mechanisms of their generation and driving and effector cytokines, as well as their role in host defense, autoimmunity, and allergic diseases.
Collapse
Affiliation(s)
- Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | | | | | | |
Collapse
|
104
|
Hultquist JF, McDougle RM, Anderson BD, Harris RS. HIV type 1 viral infectivity factor and the RUNX transcription factors interact with core binding factor β on genetically distinct surfaces. AIDS Res Hum Retroviruses 2012; 28:1543-51. [PMID: 22725134 DOI: 10.1089/aid.2012.0142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) requires the cellular transcription factor core binding factor subunit β (CBFβ) to stabilize its viral infectivity factor (Vif) protein and neutralize the APOBEC3 restriction factors. CBFβ normally heterodimerizes with the RUNX family of transcription factors, enhancing their stability and DNA-binding affinity. To test the hypothesis that Vif may act as a RUNX mimic to bind CBFβ, we generated a series of CBFβ mutants at the RUNX/CBFβ interface and tested their ability to stabilize Vif and impact transcription at a RUNX-dependent promoter. While several CBFβ amino acid substitutions disrupted promoter activity, none of these impacted the ability of CBFβ to stabilize Vif or enhance degradation of APOBEC3G. A mutagenesis screen of CBFβ surface residues identified a single amino acid change, F68D, that disrupted Vif binding and its ability to degrade APOBEC3G. This mutant still bound RUNX and stimulated RUNX-dependent transcription. These separation-of-function mutants demonstrate that HIV-1 Vif and the RUNX transcription factors interact with cellular CBFβ on genetically distinct surfaces.
Collapse
Affiliation(s)
- Judd F. Hultquist
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, Minnesota
| | - Rebecca M. McDougle
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Brett D. Anderson
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Reuben S. Harris
- Department of Biochemistry, Molecular Biology and Biophysics, Institute for Molecular Virology, Center for Genome Engineering, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Department of Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
105
|
Li L, Patsoukis N, Petkova V, Boussiotis VA. Runx1 and Runx3 are involved in the generation and function of highly suppressive IL-17-producing T regulatory cells. PLoS One 2012; 7:e45115. [PMID: 22984619 PMCID: PMC3440330 DOI: 10.1371/journal.pone.0045115] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 08/14/2012] [Indexed: 11/21/2022] Open
Abstract
CD4+Foxp3+ T regulatory cells (Tregs) display phenotypic and functional plasticity that is regulated by cytokines and other immune cells. Previously, we determined that during co-culture with CD4+CD25− T cells and antigen presenting cells, Tregs produced IL-17. Here, we investigated the mechanisms underlying the differentiation of IL-17-producing Treg (Tr17) cells and their molecular and functional properties. We determined that during stimulation via TCR/CD3 and CD28, the combination of IL-1β and IL-2 was necessary and sufficient for the generation of Tr17 cells. Tr17 cells expressed Runx1 transcription factor, which was required for sustained expression of Foxp3 and RORγt and for production of IL-17. Surprisingly, Tr17 cells also expressed Runx3, which regulated transcription of perforin and granzyme B thereby mediating cytotoxic activity. Our studies indicate that Tr17 cells concomitantly express Foxp3, RORγt, Runx1 and Runx3 and are capable of producing IL-17 while mediating potent suppressive and cytotoxic function.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Core Binding Factor Alpha 2 Subunit/genetics
- Core Binding Factor Alpha 2 Subunit/immunology
- Core Binding Factor Alpha 2 Subunit/metabolism
- Core Binding Factor Alpha 3 Subunit/genetics
- Core Binding Factor Alpha 3 Subunit/immunology
- Core Binding Factor Alpha 3 Subunit/metabolism
- Flow Cytometry
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Gene Expression
- Granzymes/genetics
- Granzymes/immunology
- Granzymes/metabolism
- Immunophenotyping
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-1beta/pharmacology
- Interleukin-2/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Perforin/genetics
- Perforin/immunology
- Perforin/metabolism
- RNA Interference
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Lequn Li
- Department of Medicine, Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nikolaos Patsoukis
- Department of Medicine, Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Victoria Petkova
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vassiliki A. Boussiotis
- Department of Medicine, Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
106
|
Soyka MB, Holzmann D, Akdis CA. Regulatory cells in allergen-specific immunotherapy. Immunotherapy 2012; 4:389-96. [PMID: 22512633 DOI: 10.2217/imt.12.10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Allergen-specific immunotherapy (SIT) is currently the best available curative treatment in allergies and has been used for the treatment of patients for the past 100 years. The formation of a Th2 cell predominant inflammation in addition to production of allergen-specific IgE, the attraction of proinflammatory cells and the degranulation of effector cells, such as mast cells, are essential mechanisms in allergy development. Tregs aim to diminish these effects by IL-10- and TGF-β-mediated anti-inflammatory reactions and therefore are one of the main targets in SIT. The induction of allergen tolerance is the key to successful SIT. With a special focus on Tregs, this review aims to clarify what is currently known about allergy development and the mode of action in allergen-SIT, which helps to develop further therapeutic strategies in the fight against allergic diseases.
Collapse
Affiliation(s)
- Michael B Soyka
- Swiss Institute of Allergy & Asthma Research (SIAF), University of Zurich, Davos, Obere Strasse 22, 7270 Davos Platz, Switzerland
| | | | | |
Collapse
|
107
|
Transcription factor Foxp3 and its protein partners form a complex regulatory network. Nat Immunol 2012; 13:1010-9. [PMID: 22922362 PMCID: PMC3448012 DOI: 10.1038/ni.2402] [Citation(s) in RCA: 356] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 07/19/2012] [Indexed: 02/07/2023]
Abstract
The transcription factor Foxp3 is indispensible for the differentiation and function of regulatory T cells (Treg cells). To gain insights into the molecular mechanisms of Foxp-mediated gene expression we purified Foxp3 complexes and explored their composition. Biochemical and mass-spectrometric analyses revealed that Foxp3 forms multi-protein complexes of 400–800 kDa or larger and identified 361 associated proteins, ~30% of which are transcription-related. Foxp3 directly regulated expression of a large proportion of the genes encoding its co-factors. Reciprocally, some transcription factor partners of Foxp3 facilitated its expression. Functional analysis of Foxp3 cooperation with one such partner, GATA-3, provided further evidence for a network of transcriptional regulation afforded by Foxp3 and its associates to control distinct aspects of Treg cell biology.
Collapse
|
108
|
Soyka MB, Wawrzyniak P, Eiwegger T, Holzmann D, Treis A, Wanke K, Kast JI, Akdis CA. Defective epithelial barrier in chronic rhinosinusitis: the regulation of tight junctions by IFN-γ and IL-4. J Allergy Clin Immunol 2012; 130:1087-1096.e10. [PMID: 22840853 DOI: 10.1016/j.jaci.2012.05.052] [Citation(s) in RCA: 363] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 05/25/2012] [Accepted: 05/30/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a common disease with still unclear pathophysiologic mechanisms. Epithelial tight junctions (TJs) have been shown to be involved in different chronic disorders, including bronchial asthma, inflammatory bowel diseases, and skin disorders. The regulation of epithelial barrier function and TJ expression has not been extensively studied in patients with CRS and in the paranasal sinus epithelium thus far. OBJECTIVE We sought to elucidate the TJ expression pattern in the epithelium of the sinonasal mucosa and its regulation in patients with CRS. METHODS Trans-tissue resistance was measured in biopsy specimens from healthy control subjects and patients with CRS with and without nasal polyps. TJ protein expression was determined by using immunofluorescence, Western blotting, and real-time PCR. Primary epithelial cell cultures from patients with CRS and control subjects were used in air-liquid interface (ALI) cultures for the measurement of transepithelial resistance (TER) and TJ expression. The effect of IFN-γ, IL-4, and IL-17 on ALI cultures was assessed. RESULTS A decreased trans-tissue resistance was found in biopsy specimens from patients with CRS with nasal polyps along with an irregular, patchy, and decreased expression of the TJ molecules occludin and zonula occludens 1. TER was reduced in ALI cultures from patients with CRS with nasal polyps. The cytokines IFN-γ and IL-4 decreased TER, whereas IL-17 did not have any influence on epithelial integrity. CONCLUSION A defective epithelial barrier was found in patients with CRS with nasal polyps along with a decreased expression of TJ proteins. The disruption of epithelial integrity by IFN-γ and IL-4 in vitro indicates a possible role for these proinflammatory cytokines in the pathogenesis of patients with CRS.
Collapse
Affiliation(s)
- Michael B Soyka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Is RUNX3 a new player in the pathogenesis of ankylosing spondylitis? Rheumatol Int 2012; 33:2449-50. [PMID: 22814793 DOI: 10.1007/s00296-012-2477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 07/07/2012] [Indexed: 10/28/2022]
|
110
|
Abstract
Regulatory T cells (Tregs) are essential for controlling peripheral tolerance by the active suppression of various immune cells including conventional T effector cells (Teffs). Downstream of the T cell receptor (TCR), more than 500 protein kinases encoded by the human genome have to be considered in signaling cascades regulating the activation of Tregs and Teffs, respectively. Following TCR engagement, Tregs posses a number of unique attributes, such as constitutive expression of Foxp3, hyporesponsiveness and poor cytokine production. Furthermore, recent studies showed that altered regulation of protein kinases is important for Treg function. These data indicate that signaling pathways in Tregs are distinctly organized and alterations at the level of protein kinases contribute to the unique Treg phenotype. However, kinase-based signaling networks in Tregs are poorly understood and necessitate further systematic characterization. In this study, we analyzed the differential expression of kinases in Tregs and Teffs by using a kinase-selective proteome strategy. In total, we revealed quantitative information on 185 kinases expressed in the human CD4(+) T cell subsets. The majority of kinases was equally abundant in both T cell subsets, but 11 kinases were differentially expressed in Tregs. Most strikingly, Tregs showed an altered expression of cell cycle kinases including CDK6. Quantitative proteomics generates first comparative insight into the kinase complements of the CD4(+) Teff and Treg subset. Treg-specific expression pattern of 11 protein kinases substantiate the current opinion that TCR-mediated signaling cascades are altered in Tregs and further suggests that Tregs exhibit significant specificities in cell-cycle control and progression.
Collapse
|
111
|
Park EJ, Kim MJ, Lee W, Park SY. Ets-2 is involved in transcriptional regulation of C1qTNF-related protein 5 in muscle cells. Mol Biol Rep 2012; 39:9445-51. [DOI: 10.1007/s11033-012-1809-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/09/2012] [Indexed: 12/22/2022]
|
112
|
Kim MJ, Park EJ, Lee W, Kim JE, Park SY. Regulation of the transcriptional activation of CTRP3 in chondrocytes by c-Jun. Mol Cell Biochem 2012; 368:111-7. [DOI: 10.1007/s11010-012-1349-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/16/2012] [Indexed: 11/25/2022]
|
113
|
Akdis CA. Therapies for allergic inflammation: refining strategies to induce tolerance. Nat Med 2012; 18:736-49. [PMID: 22561837 DOI: 10.1038/nm.2754] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Current therapies for asthma and allergy are relatively safe and effective at controlling symptoms but do not change the chronic course of disease. There is no established method to prevent asthma and allergy, and major unmet needs in this area include the better control of the severe forms of these diseases and the developments of curative therapies. Two major therapeutic strategies for asthma and allergy are currently being developed, and I here discuss the advances and challenges for future therapeutic development in these two areas. The first approach, allergen-specific immunotherapy, aims to induce specific immune tolerance and has a long-term disease-modifying effect. The second approach is the use of biological immune response modifiers to decrease pathological immune responses. Combination strategies using both of these approaches may also provide a route for addressing the unmet clinical needs in allergic diseases.
Collapse
Affiliation(s)
- Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Switzerland.
| |
Collapse
|
114
|
Meyer N, Christoph J, Makrinioti H, Indermitte P, Rhyner C, Soyka M, Eiwegger T, Chalubinski M, Wanke K, Fujita H, Wawrzyniak P, Bürgler S, Zhang S, Akdis M, Menz G, Akdis C. Inhibition of angiogenesis by IL-32: Possible role in asthma. J Allergy Clin Immunol 2012; 129:964-73.e7. [DOI: 10.1016/j.jaci.2011.12.1002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 12/08/2011] [Accepted: 12/21/2011] [Indexed: 11/29/2022]
|
115
|
Antó JM, Pinart M, Akdis M, Auffray C, Bachert C, Basagaña X, Carlsen KH, Guerra S, von Hertzen L, Illi S, Kauffmann F, Keil T, Kiley JP, Koppelman GH, Lupinek C, Martinez FD, Nawijn MC, Postma DS, Siroux V, Smit HA, Sterk PJ, Sunyer J, Valenta R, Valverde S, Akdis CA, Annesi-Maesano I, Ballester F, Benet M, Cambon-Thomsen A, Chatzi L, Coquet J, Demoly P, Gan W, Garcia-Aymerich J, Gimeno-Santos E, Guihenneuc-Jouyaux C, Haahtela T, Heinrich J, Herr M, Hohmann C, Jacquemin B, Just J, Kerkhof M, Kogevinas M, Kowalski ML, Lambrecht BN, Lau S, Lødrup Carlsen KC, Maier D, Momas I, Noel P, Oddie S, Palkonen S, Pin I, Porta D, Punturieri A, Rancière F, Smith RA, Stanic B, Stein RT, van de Veen W, van Oosterhout AJM, Varraso R, Wickman M, Wijmenga C, Wright J, Yaman G, Zuberbier T, Bousquet J. Understanding the complexity of IgE-related phenotypes from childhood to young adulthood: a Mechanisms of the Development of Allergy (MeDALL) seminar. J Allergy Clin Immunol 2012; 129:943-54.e4. [PMID: 22386796 DOI: 10.1016/j.jaci.2012.01.047] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/22/2011] [Accepted: 01/12/2012] [Indexed: 12/18/2022]
Abstract
Mechanisms of the Development of Allergy (MeDALL), a Seventh Framework Program European Union project, aims to generate novel knowledge on the mechanisms of initiation of allergy. Precise phenotypes of IgE-mediated allergic diseases will be defined in MeDALL. As part of MeDALL, a scientific seminar was held on January 24, 2011, to review current knowledge on the IgE-related phenotypes and to explore how a multidisciplinary effort could result in a new integrative translational approach. This article provides a summary of the meeting. It develops challenges in IgE-related phenotypes and new clinical and epidemiologic approaches to the investigation of allergic phenotypes, including cluster analysis, scale-free models, candidate biomarkers, and IgE microarrays; the particular case of severe asthma was reviewed. Then novel approaches to the IgE-associated phenotypes are reviewed from the individual mechanisms to the systems, including epigenetics, human in vitro immunology, systems biology, and animal models. The last chapter deals with the understanding of the population-based IgE-associated phenotypes in children and adolescents, including age effect in terms of maturation, observed effects of early-life exposures and shift of focus from early life to pregnancy, gene-environment interactions, cohort effects, and time trends in patients with allergic diseases. This review helps to define phenotypes of allergic diseases in MeDALL.
Collapse
Affiliation(s)
- Josep M Antó
- Centre for Research in Environmental Epidemiology, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Sakaguchi S, Benham H, Cope AP, Thomas R. T‐cell receptor signaling and the pathogenesis of autoimmune arthritis: insights from mouse and man. Immunol Cell Biol 2012; 90:277-87. [DOI: 10.1038/icb.2012.4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Shimon Sakaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University Suita Japan
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University Kyoto Japan
| | - Helen Benham
- The University of Queensland, Diamantina Institute, Princess Alexandra Hospital Brisbane Queensland Australia
| | - Andrew P Cope
- Academic Department of Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, Division of Immunology, Infection and Inflammatory Diseases, School of Medicine, King's College London London UK
| | - Ranjeny Thomas
- The University of Queensland, Diamantina Institute, Princess Alexandra Hospital Brisbane Queensland Australia
| |
Collapse
|
117
|
Sonobe Y, Li H, Jin S, Kishida S, Kadomatsu K, Takeuchi H, Mizuno T, Suzumura A. Midkine inhibits inducible regulatory T cell differentiation by suppressing the development of tolerogenic dendritic cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2602-11. [PMID: 22323540 DOI: 10.4049/jimmunol.1102346] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Midkine (MK), a heparin-binding growth factor, reportedly contributes to inflammatory diseases, including Crohn's disease and rheumatoid arthritis. We previously showed that MK aggravates experimental autoimmune encephalomyelitis (EAE) by decreasing regulatory CD4(+)CD25(+)Foxp3(+) T cells (Tregs), a population that regulates the development of autoimmune responses, although the precise mechanism remains uncertain. In this article, we show that MK produced in inflammatory conditions suppresses the development of tolerogenic dendritic cells (DCregs), which drive the development of inducible Treg. MK suppressed DCreg-mediated expansion of the CD4(+)CD25(+)Foxp3(+) Treg population. DCregs expressed significantly higher levels of CD45RB and produced significantly less IL-12 compared with conventional dendritic cells. However, MK downregulated CD45RB expression and induced IL-12 production by reducing phosphorylated STAT3 levels via src homology region 2 domain-containing phosphatase-2 in DCreg. Inhibiting MK activity with anti-MK RNA aptamers, which bind to the targeted protein to suppress the function of the protein, increased the numbers of CD11c(low)CD45RB(+) dendritic cells and Tregs in the draining lymph nodes and suppressed the severity of EAE, an animal model of multiple sclerosis. Our results also demonstrated that MK was produced by inflammatory cells, in particular, CD4(+) T cells under inflammatory conditions. Taken together, these results suggest that MK aggravates EAE by suppressing DCreg development, thereby impairing the Treg population. Thus, MK is a promising therapeutic target for various autoimmune diseases.
Collapse
Affiliation(s)
- Yoshifumi Sonobe
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
118
|
Palomares O, Rückert B, Jartti T, Kücüksezer UC, Puhakka T, Gomez E, Fahrner HB, Speiser A, Jung A, Kwok WW, Kalogjera L, Akdis M, Akdis CA. Induction and maintenance of allergen-specific FOXP3+ Treg cells in human tonsils as potential first-line organs of oral tolerance. J Allergy Clin Immunol 2012; 129:510-20, 520.e1-9. [DOI: 10.1016/j.jaci.2011.09.031] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 09/20/2011] [Accepted: 09/28/2011] [Indexed: 01/31/2023]
|
119
|
Luzina IG, Lockatell V, Lavania S, Pickering EM, Kang PH, Bashkatova YN, Andreev SM, Atamas SP. Natural production and functional effects of alternatively spliced interleukin-4 protein in asthma. Cytokine 2012; 58:20-6. [PMID: 22249152 DOI: 10.1016/j.cyto.2011.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 11/13/2011] [Accepted: 12/22/2011] [Indexed: 11/29/2022]
Abstract
We have previously described an alternatively spliced isoform of IL-4 mRNA that omits exon 2 and is termed IL-4δ2. However, the natural production of IL-4δ2 protein and its association with disease have not been previously assessed due to unavailability of an antibody that interacts with IL-4δ2 without cross-reactivity with full length IL-4. We used a unique monoclonal antibody (mAb) that reacts with IL-4δ2, but not with IL-4, and observed that IL-4δ2 is naturally produced by T cells from patients with asthma, but not from healthy controls. The kinetics of IL-4δ2 and IL-4 production by phorbol myristate acetate (PMA)/ionomycin-activated cells differed, with IL-4δ2 increasing at 48-72h and IL-4 peaking at 24h. The steady-state levels of IL-4δ2 mRNA varied significantly among the donors and were discordant with the corresponding protein levels, suggesting post-transcriptional regulation of protein production. Polarized Th1 or Th2 lymphocytes were not a major source of IL-4δ2. Stimulation of cultured T lymphocytes with IL-4δ2 caused elevated production of IFN-γ, IL-10, IL-6, MCP-1, and TNF-α, with notable differences between patients and controls in the production of IFN-γ, IL-10, and IL-6. Thus, IL-4δ2 is natively produced not only as mRNA but also as a protein by cells other than Th1 or Th2. It is regulated post-transcriptionally, is associated with allergic asthma, and regulates production of other cytokines by primary T lymphocytes. Alternatively spliced interleukin-4 may be a new biomarker, a pathophysiological player, and possibly a molecular target for future therapies in asthma.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
120
|
Abstract
The immune system has evolved to mount an effective defense against pathogens and to minimize deleterious immune-mediated inflammation caused by commensal microorganisms, immune responses against self and environmental antigens, and metabolic inflammatory disorders. Regulatory T (Treg) cell-mediated suppression serves as a vital mechanism of negative regulation of immune-mediated inflammation and features prominently in autoimmune and autoinflammatory disorders, allergy, acute and chronic infections, cancer, and metabolic inflammation. The discovery that Foxp3 is the transcription factor that specifies the Treg cell lineage facilitated recent progress in understanding the biology of regulatory T cells. In this review, we discuss cellular and molecular mechanisms in the differentiation and function of these cells.
Collapse
Affiliation(s)
- Steven Z Josefowicz
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, NY 10021, USA
| | | | | |
Collapse
|
121
|
Fujita H, Soyka MB, Akdis M, Akdis CA. Mechanisms of allergen-specific immunotherapy. Clin Transl Allergy 2012; 2:2. [PMID: 22409879 PMCID: PMC3395833 DOI: 10.1186/2045-7022-2-2] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 01/05/2012] [Indexed: 01/25/2023] Open
Abstract
Allergen-specific immunotherapy (allergen-SIT) is a potentially curative treatment approach in allergic diseases. It has been used for almost 100 years as a desensitizing therapy. The induction of peripheral T cell tolerance and promotion of the formation of regulatory T-cells are key mechanisms in allergen-SIT. Both FOXP3+CD4+CD25+ regulatory T (Treg) cells and inducible IL-10- and TGF-β-producing type 1 Treg (Tr1) cells may prevent the development of allergic diseases and play a role in successful allergen-SIT and healthy immune response via several mechanisms. The mechanisms of suppression of different pro-inflammatory cells, such as eosinophils, mast cells and basophils and the development of allergen tolerance also directly or indirectly involves Treg cells. Furthermore, the formation of non-inflammatory antibodies particularly IgG4 is induced by IL-10. Knowledge of these molecular basis is crucial in the understanding the regulation of immune responses and their possible therapeutic targets in allergic diseases.
Collapse
Affiliation(s)
- Hiroyuki Fujita
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| | | | | | | |
Collapse
|
122
|
Maruyama T, Konkel JE, Zamarron BF, Chen W. The molecular mechanisms of Foxp3 gene regulation. Semin Immunol 2011; 23:418-23. [PMID: 21752667 PMCID: PMC3205321 DOI: 10.1016/j.smim.2011.06.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/07/2011] [Indexed: 01/03/2023]
Abstract
Induction of Foxp3 gene expression and acquisition of regulatory T cell fate is, understandably, a highly controlled process and one which many investigators want to illuminate. In studying the regulation of Foxp3 gene expression, several conserved non-coding regions have been identified and the role of various transcription factors at these sites has been explored. What emerges is that many factors, some positive, some negative, interact to collectively drive Foxp3 gene expression and then maintain its expression in Foxp3(+) regulatory T cells. TCR signaling is imperative for Foxp3 gene expression and TGF-β is a key cytokine for initiating Foxp3 gene expression in naïve T cells. But other signaling pathways are also known to play a role in properly orchestrating Foxp3 gene expression and regulatory T cell expansion. Here we review the recent progress in understanding the complex molecular events that drive Foxp3 gene expression and allow functional regulatory T cells to develop.
Collapse
Affiliation(s)
- Takashi Maruyama
- Mucosal Immunology Section, OIIB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
123
|
Molecular mechanisms underlying the regulation and functional plasticity of FOXP3(+) regulatory T cells. Genes Immun 2011; 13:1-13. [PMID: 22048454 DOI: 10.1038/gene.2011.77] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD4(+) CD25(+) regulatory T (Treg) cells engage in the maintenance of immunological self-tolerance and homeostasis by limiting aberrant or excessive inflammation. The transcription factor forkhead box P3 (FOXP3) is critical for the development and function of Treg cells. The differentiation of the Treg cell lineage is not terminal, as developmental and functional plasticity occur through the sensing of inflammatory signals in the periphery. Here, we review the recent progress in our understanding of the molecular mechanisms underlying the regulation and functional plasticity of CD4(+) CD25(+) FOXP3(+) Treg cells, through the perturbation of FOXP3 and its complex at a transcriptional, translational and post-translational level.
Collapse
|
124
|
Balancing acts: the role of TGF-β in the mucosal immune system. Trends Mol Med 2011; 17:668-76. [PMID: 21890412 DOI: 10.1016/j.molmed.2011.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/30/2011] [Accepted: 07/07/2011] [Indexed: 12/23/2022]
Abstract
The gastrointestinal mucosal immune system faces unique challenges in dealing not only with fed antigens but also both commensal and pathogenic bacteria. It is tasked with digesting, transporting and using nutritional antigens while protecting the host from pathogenic organisms. As such, mechanisms that mediate effective immunity and immune tolerance are active within the gut environment. To accomplish this, the mucosal immune system has evolved sophisticated mechanisms that safeguard the integrity of the mucosal barrier. Transforming growth factor-β (TGF-β) emerges as a key mediator, balancing the tolerogenic and immunogenic forces at play in the gut. In this review, we discuss the role of TGF-β in the generation and functioning of gut lymphocyte populations. We highlight recent findings, summarize controversies, outline remaining questions and provide our personal perspectives.
Collapse
|
125
|
Eiwegger T, Akdis CA. IL-33 links tissue cells, dendritic cells and Th2 cell development in a mouse model of asthma. Eur J Immunol 2011; 41:1535-8. [PMID: 21618506 DOI: 10.1002/eji.201141668] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IL-33 is becoming a central molecule in allergic asthma that addresses various cascades of innate and adaptive immune responses that lead to inflammation in the lung. Its effects are exerted via its heterodimeric receptor that consists of ST2 and the ubiquitously expressed IL-1 receptor accessory protein (ILRAcP). IL-33 integrates both innate and adaptive immunity in a unique fashion via basophils, mast cells, eosinophils, innate lymphoid cells, NK and NKT cells, nuocytes, Th2 lymphocytes and a CD34(pos) precursor cell population. These actions of IL-33 seem to be particularly strong and dominant in models with mucosal inflammation. A study in this issue of the European Journal of Immunology demonstrates that IL-33 acts, in an ST2-dependent manner, as a maturation factor for BM-derived DCs via up-regulation of CD80, CD40 and OX40L. This process is accompanied by the release of pro-inflammatory cytokines, such as IL-6, IL-1β, TNF-α and TARC/CCL17. IL-33-pre-treated DCs were significantly more potent for the generation of allergen-specific Th2-type cells with IL-5 and IL-13 production. Intratracheal administration of OVA-pulsed DCs with IL-33 significantly enhances eosinophil numbers and mucous secretion. In conclusion, IL-33 affects both the development of allergic sensitization and the development of lung inflammation in allergic asthma.
Collapse
Affiliation(s)
- Thomas Eiwegger
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | | |
Collapse
|
126
|
Fujita H, Chalubinski M, Rhyner C, Indermitte P, Meyer N, Ferstl R, Treis A, Gomez E, Akkaya A, O'Mahony L, Akdis M, Akdis CA. Claudin-1 expression in airway smooth muscle exacerbates airway remodeling in asthmatic subjects. J Allergy Clin Immunol 2011; 127:1612-21.e8. [PMID: 21624620 DOI: 10.1016/j.jaci.2011.03.039] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 03/04/2011] [Accepted: 03/08/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Increased airway smooth muscle (ASM) mass is an essential component of airway remodeling and asthma development, and there is no medication specifically against it. Tight junction (TJ) proteins, which are expressed in endothelial and epithelial cells and affect tissue integrity, might exist in other types of cells and display additional functions in the asthmatic lung. OBJECTIVE The aim of this study was to investigate the existence, regulation, and function of TJ proteins in ASM in asthmatic patients. METHODS The expression and function of TJ proteins in primary ASM cell lines, human bronchial biopsy specimens, and a murine model of asthma were analyzed by means of RT-PCR, multispectral imaging flow cytometry, immunohistochemistry, Western blotting, 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester staining, tritiated thymidine incorporation, wound-healing assay, and luminometric bead array. RESULTS Increased claudin-1 expression was observed in ASM of asthmatic patients, as well as in a murine model of asthma-like airway inflammation. Whereas IL-1β and TNF-α upregulated claudin-1 expression, it was downregulated by the T(H)2 cytokines IL-4 and IL-13 in primary human ASM cells. Claudin-1 was localized to the nucleus and cytoplasm but not to the cell surface in ASM cells. Claudin-1 played a central role in ASM cell proliferation, as demonstrated by increased ASM cell proliferation seen with overexpression and decreased proliferation seen with small interfering RNA knockdown of claudin-1. Overexpression of claudin-1 induced vascular endothelial growth factor and downregulated IL-6, IL-8, and IFN-γ-induced protein 10 production by ASM cells. Claudin-1 upregulation by IL-1β or TNF-α was suppressed by dexamethasone but not by rapamycin, FK506, or salbutamol. CONCLUSION These results demonstrate that claudin-1 might play a role in airway remodeling in asthmatic patients by means of regulation of ASM cell proliferation, angiogenesis, and inflammation.
Collapse
Affiliation(s)
- Hiroyuki Fujita
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Soyer OU, Akdis M, Akdis CA. Mechanisms of subcutaneous allergen immunotherapy. Immunol Allergy Clin North Am 2011; 31:175-90, vii-viii. [PMID: 21530813 DOI: 10.1016/j.iac.2011.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Allergen-specific immunotherapy (SIT) is the only curative approach in the treatment of allergic diseases defined up-to-date. Peripheral T-cell tolerance to allergens, the goal of successful allergen-SIT, is the primary mechanism in healthy immune responses to allergens. By repeated administration of increased doses of the causative allergen, allergen-SIT induces a state of immune tolerance to allergens through the constitution of T regulatory (Treg) cells, including allergen-specific interleukin (IL)-10-secreting Treg type 1 cells and CD4(+)CD25(+)Treg cells; induction of suppressive cytokines, such as IL-10 and transforming growth factor β; suppression of allergen-specific IgE and induction of IgG4 and IgA; and suppression of mast cells, basophils, eosinophils, and inflammatory dendritic cells. This review summarizes the current knowledge on the mechanisms of allergen-SIT with emphasis on the roles of Treg cells in allergen-SIT.
Collapse
Affiliation(s)
- Ozge U Soyer
- Department of Pediatric Allergy, Ankara Education and Research Hospital, Ministry of Health, Ulucanlar Street, No 11, Ankara, Turkey 06080
| | | | | |
Collapse
|
128
|
Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Mechanisms of immunotherapy to wasp and bee venom. Clin Exp Allergy 2011; 41:1226-34. [PMID: 21729181 DOI: 10.1111/j.1365-2222.2011.03812.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hymenoptera venoms are important allergens that can elicit both local and systemic allergic reactions, including life-threatening anaphylaxis. Venom immunotherapy (VIT) remains the most effective treatment, reducing the risk of systemic reactions in individuals with Hymenoptera venom allergy. VIT can restore normal immunity against venom allergens and provide patients with a lifetime of tolerance to venoms. During VIT, peripheral tolerance is induced by the generation of allergen-specific regulatory T (Treg) cells, which suppress proliferative and cytokine responses against the venom allergens. Treg cells are characterized by IL-10 secretion that directly or indirectly influence effector cells of allergic inflammation, such as mast cells, basophils and eosinophils. Treg cells also have influence on B cells, suppressing IgE production and inducing the production of blocking type IgG4 antibodies against venom allergens. An accumulating body of evidence suggests that Treg cells may affect allergen sensitization and methods for enhancing this cell population may eventually improve the efficacy of VIT. In this article, immune mechanisms enrolled in bee and wasp VIT are reviewed.
Collapse
Affiliation(s)
- C Ozdemir
- Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | | | | | | |
Collapse
|
129
|
Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Specific immunotherapy and turning off the T cell: how does it work? Ann Allergy Asthma Immunol 2011; 107:381-92. [PMID: 22018608 DOI: 10.1016/j.anai.2011.05.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/08/2011] [Accepted: 05/17/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To examine T-regulatory (Treg) cell functions in allergic immune responses and their roles during allergen specific immunotherapy based on recent developments and current understanding of immune regulation. DATA SOURCES PubMed search of English-language articles regarding Treg cells and allergen specific immunotherapy. STUDY SELECTION Articles on the subject matter were selected and reviewed. RESULTS Allergen specific immunotherapy is the ultimate treatment modality targeting the immunopathogenic mechanisms of allergic disorders. A diminished allergen-specific T-cell proliferation and suppressed secretion of T(H)1- and T(H)2-type cytokines are the characteristic hallmarks. In addition, Treg cells inhibit the development of allergen-specific T(H)2 and T(H)1 cell responses and therefore exert key roles in healthy immune response to allergens. Treg cells potently suppress IgE production and directly or indirectly control the activity of effector cells of allergic inflammation, such as eosinophils, basophils, and mast cells. CONCLUSION As advancements in the field of allergen specific immunotherapy ensue, they may provide novel progression of more rational and safer approaches for the prevention and treatment of allergic disorders. Currently, the Treg cell field is an open research area to increase our understanding in mechanisms of peripheral tolerance to allergens.
Collapse
Affiliation(s)
- Cevdet Ozdemir
- Division of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
| | | | | | | |
Collapse
|
130
|
Abstract
The studies on the mechanisms of specific immunotherapy (SIT) point out its targets that decide on the efficacy of SIT and hence might be used for its further improvement. Several mechanisms have been proposed to explain the beneficial effects of immunotherapy. The knowledge of the mechanisms underlying allergic diseases and curative treatment possibilities has experienced exciting advances over the last three decades. Studies in several clinical trials in allergen-SIT have demonstrated that the induction of a tolerant state against allergens in many ways represents a key step in the development of a healthy immune response against allergens. Several cellular and molecular mechanisms have been demonstrated: allergen-specific suppressive capacities of both inducible subsets of CD4(+) CD25(+) forkhead box P3(+) T-regulatory and IL-10-secreting type 1 T-regulatory cells increase in peripheral blood; suppression of eosinophils, mast cells, and basophils; Ab isotype change from IgE to IgG4. This review aims at the better understanding of the observed immunological changes associated with allergen SIT.
Collapse
Affiliation(s)
- Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wrocław, Poland.
| | | |
Collapse
|
131
|
Palomares O, O'Mahony L, Akdis CA. The many routes of dendritic cells to ensure immune regulation. J Allergy Clin Immunol 2011; 127:1541-2. [DOI: 10.1016/j.jaci.2011.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 11/29/2022]
|
132
|
Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, Klunker S, Meyer N, O'Mahony L, Palomares O, Rhyner C, Ouaked N, Quaked N, Schaffartzik A, Van De Veen W, Zeller S, Zimmermann M, Akdis CA. Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases. J Allergy Clin Immunol 2011; 127:701-21.e1-70. [PMID: 21377040 DOI: 10.1016/j.jaci.2010.11.050] [Citation(s) in RCA: 573] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 12/17/2022]
Abstract
Advancing our understanding of mechanisms of immune regulation in allergy, asthma, autoimmune diseases, tumor development, organ transplantation, and chronic infections could lead to effective and targeted therapies. Subsets of immune and inflammatory cells interact via ILs and IFNs; reciprocal regulation and counter balance among T(h) and regulatory T cells, as well as subsets of B cells, offer opportunities for immune interventions. Here, we review current knowledge about ILs 1 to 37 and IFN-γ. Our understanding of the effects of ILs has greatly increased since the discoveries of monocyte IL (called IL-1) and lymphocyte IL (called IL-2); more than 40 cytokines are now designated as ILs. Studies of transgenic or knockout mice with altered expression of these cytokines or their receptors and analyses of mutations and polymorphisms in human genes that encode these products have provided important information about IL and IFN functions. We discuss their signaling pathways, cellular sources, targets, roles in immune regulation and cellular networks, roles in allergy and asthma, and roles in defense against infections.
Collapse
Affiliation(s)
- Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev 2011; 238:247-62. [PMID: 20969597 DOI: 10.1111/j.1600-065x.2010.00951.x] [Citation(s) in RCA: 425] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD4(+) T cells, also known as T-helper (Th) cells, play an important role in orchestrating adaptive immune responses to various infectious agents. They are also involved in the induction of autoimmune and allergic diseases. Upon T-cell receptor (TCR)-mediated cell activation, naive CD4(+) T cells can differentiate into at least four major lineages, Th1, Th2, Th17, and iTreg cells, that participate in different types of immune responses. Networks of cytokines and transcription factors are critical for determining CD4(+) T-cell fates and effector cytokine production. Here, we review collaboration and cross-regulation between various essential cytokines in the activation/induction of key transcription factors during the process of Th cell differentiation towards these distinct lineages. We also discuss the interactions of key transcription factors at both genetic and protein levels and the function of the resulting network(s) in regulating the expression of effector cytokines.
Collapse
Affiliation(s)
- Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA.
| | | |
Collapse
|
134
|
Yin G, Li J, Wan Y, Hou R, Li X, Zhang J, Cheng T, Zhang K. Abnormality of RUNX1 signal transduction in psoriatic CD34+ bone marrow cells. Br J Dermatol 2011; 164:1043-51. [DOI: 10.1111/j.1365-2133.2010.10192.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
135
|
Bandukwala HS, Wu Y, Feurer M, Chen Y, Barbosa B, Ghosh S, Stroud JC, Benoist C, Mathis D, Rao A, Chen L. Structure of a domain-swapped FOXP3 dimer on DNA and its function in regulatory T cells. Immunity 2011; 34:479-91. [PMID: 21458306 PMCID: PMC3085397 DOI: 10.1016/j.immuni.2011.02.017] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 12/24/2010] [Accepted: 02/23/2011] [Indexed: 12/18/2022]
Abstract
The transcription factor FOXP3 is essential for the suppressive function of regulatory T cells that are required for maintaining self-tolerance. We have solved the crystal structure of the FOXP3 forkhead domain as a ternary complex with the DNA-binding domain of the transcription factor NFAT1 and a DNA oligonucleotide from the interleukin-2 promoter. A striking feature of this structure is that FOXP3 forms a domain-swapped dimer that bridges two molecules of DNA. Structure-guided or autoimmune disease (IPEX)-associated mutations in the domain-swap interface diminished dimer formation by the FOXP3 forkhead domain without compromising FOXP3 DNA binding. These mutations also eliminated T cell-suppressive activity conferred by FOXP3, both in vitro and in a murine model of autoimmune diabetes in vivo. We conclude that FOXP3-mediated suppressor function requires dimerization through the forkhead domain and that mutations in the dimer interface can lead to the systemic autoimmunity observed in IPEX patients.
Collapse
Affiliation(s)
- Hozefa S. Bandukwala
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children’s Hospital, Boston, MA 02115
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Department of Pediatrics, Children’s Hospital Boston and Harvard Medical School, Boston, MA 02115
| | - Yongqing Wu
- Department of Biological Sciences, Department of Chemistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089
| | - Markus Feurer
- Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Yongheng Chen
- Department of Biological Sciences, Department of Chemistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089
| | - Bianca Barbosa
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children’s Hospital, Boston, MA 02115
| | - Srimoyee Ghosh
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children’s Hospital, Boston, MA 02115
- Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - James C. Stroud
- Department of Biological Sciences, Department of Chemistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089
| | | | - Diane Mathis
- Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Anjana Rao
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children’s Hospital, Boston, MA 02115
- Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Lin Chen
- Department of Biological Sciences, Department of Chemistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
136
|
Sugai M, Aoki K, Osato M, Nambu Y, Ito K, Taketo MM, Shimizu A. Runx3 is required for full activation of regulatory T cells to prevent colitis-associated tumor formation. THE JOURNAL OF IMMUNOLOGY 2011; 186:6515-20. [PMID: 21515792 DOI: 10.4049/jimmunol.1001671] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation is increasingly recognized as an essential component of tumorigenesis, which is promoted and suppressed by various T cell subsets acting in different ways. It was shown previously in Runx3-deficient mice that differentiation of CD8 T and NK cells is perturbed. In this study, we show that Runx3 is also required for proper differentiation and function of regulatory T cells. In Runx3-deficient mice, T cells were unable to inhibit inflammation and to suppress tumor development. As expected, recombination activating gene 2-deficient mice bearing Runx3-deficient lymphocytes spontaneously developed colon tumors. However, tumor formation was completely blocked by transfer of either regulatory T cells or CD8 T cells derived from wild-type mice to mutant mice or by housing mutant mice in a specific pathogen-free condition. These results indicate that Runx3-deficient lymphocytes and microorganisms act together to induce inflammation and consequently induce the development of colon tumors.
Collapse
Affiliation(s)
- Manabu Sugai
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
137
|
Martino D, Prescott S. Epigenetics and prenatal influences on asthma and allergic airways disease. Chest 2011; 139:640-647. [PMID: 21362650 DOI: 10.1378/chest.10-1800] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Uterine life is arguably the most critical time in developmental programming, when environmental exposures may have the greatest potential to influence evolving fetal structure and function. There has been substantial progress in understanding the epigenetic mechanisms through which environmental exposures can permanently alter the expression of fetal genes and contribute to the increasing propensity for many complex diseases. These concepts of "developmental origins" of disease are being applied across virtually all fields of medicine, and emerging epigenetic paradigms are the likely mechanism behind the environment-driven epidemic of asthma and allergic disease. Here, we examine the epigenetic regulation of immune development and the early immune profiles that contribute to allergic risk. In particular we review new evidence that key environmental exposures, such as microbial exposure, dietary changes, tobacco smoke, and pollutants, can induce epigenetic changes in gene expression and alter disease risk. Although most of these factors have already been clearly implicated in epidemiologic studies of asthma and allergic disease, new studies investigating the mechanisms of these effects may provide new avenues for using these pathways for disease prevention.
Collapse
Affiliation(s)
- David Martino
- School of Paediatrics and Child Health Research, University of Western Australia, Perth, WA, Australia
| | - Susan Prescott
- School of Paediatrics and Child Health Research, University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
138
|
The role of epigenetic dysregulation in the epidemic of allergic disease. Clin Epigenetics 2011; 2:223-232. [PMID: 21949548 PMCID: PMC3156327 DOI: 10.1007/s13148-011-0028-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 03/13/2011] [Indexed: 12/14/2022] Open
Abstract
The epidemic of allergic disease in early life is one of the clearest indicators that the developing immune system is vulnerable to modern environmental changes. A range of environmental exposures epidemiologically associated with allergic disease have been shown to have effects on the foetal immune function in pregnancy, including microbial burden, dietary changes and environmental pollutants. Preliminary studies now suggest that these early effects on immune development may be mediated epigenetically through a variety of processes that collectively modify gene expression and allergic susceptibility and that these effects are potentially heritable across generations. It is also possible that rising rates of maternal allergy, a recognised direct risk factor for infant allergic disease, may be further amplifying the effects of environmental changes. Whilst effective prevention strategies are the ultimate goal in reversing the allergy epidemic, the specific environmental drivers, target genes, and intracellular pathways and mechanisms of early life immune programming are still unclear. It is hoped that identifying genes that are differentially regulated in association with subsequent allergic disease will assist in identifying causal pathways and upstream contributing environmental factors. In this way, epigenetic paradigms are likely to provide valuable insights into how the early environment can be modified to more favourably drive immune development and reverse the allergic epidemic.
Collapse
|
139
|
Schmidl C, Hansmann L, Andreesen R, Edinger M, Hoffmann P, Rehli M. Epigenetic reprogramming of the RORC locus during in vitro expansion is a distinctive feature of human memory but not naïve Treg. Eur J Immunol 2011; 41:1491-8. [PMID: 21469109 DOI: 10.1002/eji.201041067] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 01/03/2011] [Accepted: 01/28/2011] [Indexed: 12/17/2022]
Abstract
The adoptive transfer of in vitro expanded Treg is a promising treatment option for autoimmune as well as alloantigen-induced diseases. Yet, concerns about the phenotypic and functional stability of Tregs upon in vitro culture command both careful selection of the starting population and thorough characterization of the final cell product. Recently, a high degree of developmental plasticity has been described for murine Treg and Th17 cells. Similarly, IL-17-producing FOXP3(+) cells have been detected among the CD45RA(-) memory-type subpopulation of human Tregs ex vivo. This prompted us to investigate the predisposition of human naïve and memory Tregs to develop into Th17 cells during polyclonal in vitro expansion. Here, we show that stimulation-induced DNA demethylation of RORC, which encodes the lineage-defining transcription factor for Th17 cells, occurs selectively in CD45RA(-) memory-type Tregs, irrespective of their FOXP3 expression level. On the contrary, naïve CD45RA(+) Tregs retain stable CpG methylation across the RORC locus even upon prolonged ex vivo expansion and in consequence show only a marginal tendency to express RORC and develop into IL-17-producing cells. These findings are highly relevant for the generation of therapeutic Treg products.
Collapse
Affiliation(s)
- Christian Schmidl
- Department of Hematology & Oncology, University Hospital Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
140
|
The cellular orchestra in skin allergy; are differences to lung and nose relevant? Curr Opin Allergy Clin Immunol 2011; 10:443-51. [PMID: 20736733 DOI: 10.1097/aci.0b013e32833d7d48] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW It has been a long lasting question that although a similar peripheral allergen-specific immune response has been observed, why some patients show only atopic dermatitis, rhinitis and asthma alone or their combinations. The answer resides in the propensity of resident tissue cells and local antigen-presenting cells and T cells for developing an allergic inflammatory immune response. Antigen-presenting cells introduce processed allergens to T helper lymphocytes, where a decision of developing different types of T cell immunity is given under the influence of several cytokines, chemokines, costimulatory signals and regulatory T cells. RECENT FINDINGS We focused in this review article on effector T cell subsets, which have been recently described such as Th9, Th17 cells and Th22 cells, which are characterized by their IL-9 and IL-10, IL-17 (or IL-17A) and IL-22 expression, respectively together with other proinflammatory cytokines, which coordinate local tissue inflammation. Both naturally occurring CD4+CD25+ regulatory T (Treg) cells and inducible populations of allergen-specific, IL-10-secreting Treg type 1 cells inhibit allergen-specific effector cells and have been shown to play a central role in the maintenance of peripheral homeostasis and the establishment of controlled immune responses in allergic inflammatory tissues. SUMMARY Better understanding and characterization of newly described effector cell subsets and their interaction between antigen presenting cells and resident tissue cells will enlighten our knowledge on the mechanisms of allergic diseases.
Collapse
|
141
|
Addey C, White M, Dou L, Coe D, Dyson J, Chai JG. Functional Plasticity of Antigen-Specific Regulatory T Cells in Context of Tumor. THE JOURNAL OF IMMUNOLOGY 2011; 186:4557-64. [DOI: 10.4049/jimmunol.1003797] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
142
|
Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy. J Allergy Clin Immunol 2011; 127:18-27; quiz 28-9. [PMID: 21211639 DOI: 10.1016/j.jaci.2010.11.030] [Citation(s) in RCA: 309] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 11/22/2010] [Accepted: 11/22/2010] [Indexed: 01/05/2023]
Abstract
Allergen-specific immunotherapy has been used for 100 years as a desensitizing therapy for allergic diseases and represents the potentially curative and specific method of treatment. The mechanisms of action of allergen-specific immunotherapy include the very early desensitization effects, modulation of T-and B-cell responses and related antibody isotypes, and migration of eosinophils, basophils, and mast cells to tissues, as well as release of their mediators. Regulatory T (Treg) cells have been identified as key regulators of immunologic processes in peripheral tolerance to allergens. Skewing of allergen-specific effector T cells to a regulatory phenotype appears as a key event in the development of healthy immune response to allergens and successful outcome in patients undergoing allergen-specific immunotherapy. Naturally occurring forkhead box protein 3-positive CD4(+)CD25(+) Treg cells and inducible T(R)1 cells contribute to the control of allergen-specific immune responses in several major ways, which can be summarized as suppression of dendritic cells that support the generation of effector T cells; suppression of effector T(H)1, T(H)2, and T(H)17 cells; suppression of allergen-specific IgE and induction of IgG4; suppression of mast cells, basophils, and eosinophils; and suppression of effector T-cell migration to tissues. New strategies for immune intervention will likely include targeting of the molecular mechanisms of allergen tolerance and reciprocal regulation of effector and Treg cell subsets.
Collapse
Affiliation(s)
- Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| | | |
Collapse
|
143
|
Control of the development of CD8αα+ intestinal intraepithelial lymphocytes by TGF-β. Nat Immunol 2011; 12:312-9. [PMID: 21297643 PMCID: PMC3062738 DOI: 10.1038/ni.1997] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 01/11/2011] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms directing the development of TCRαβ+CD8αα+ intestinal intraepithelial lymphocytes (IEL) are not thoroughly understood. Here we show that transforming growth factor-β (TGF-β) controls the development of TCRαβ+CD8αα+ IEL. Mice with either a TGF-β1 null mutation or a T cell-specific deletion of the TGF-β receptor I lacked TCRαβ+CD8αα+ IEL, whereas transgenic mice that over-expressed TGF-β1 had an increased population of TCRαβ+CD8αα+ IEL. Defective development of the TCRαβ+CD8αα+ IEL thymic precursors (CD4-CD8-TCRαβ+CD5+) was observed in the absence of TGF-β. In addition, we showed that TGF-β signaling induced CD8α expression in TCRαβ+CD8αα+ IEL thymic precursors and induced and maintained CD8α expression in peripheral populations of T cells. These data demonstrate a previously unrecognized role for TGF-β in the development of TCRαβ+CD8αα+ IEL and the expression of CD8 in T cells.
Collapse
|
144
|
Wong WF, Kurokawa M, Satake M, Kohu K. Down-regulation of Runx1 expression by TCR signal involves an autoregulatory mechanism and contributes to IL-2 production. J Biol Chem 2011; 286:11110-8. [PMID: 21292764 DOI: 10.1074/jbc.m110.166694] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Runx1 transcription factor plays multiple roles in T cell development, differentiation, and function. However, the regulatory mechanisms and functional significance of high Runx1 protein expression in resting peripheral CD4+ T cells is not well understood. Here, we demonstrate that T-cell receptor (TCR) activation down-regulates distal Runx1 transcription, resulting in a significant reduction of Runx1 protein. Interestingly, this down-regulation of distal Runx1 transcription appears to be mediated through a negative auto-regulatory mechanism, whereby Runx1 protein binds to a Runx consensus site in the distal promoter. Through the use of Runx1-overexpressing cells from transgenic mice, we demonstrate that interference with TCR-mediated Runx1 down-regulation inhibits IL-2 production and proliferation in activated CD4+ T cells. In contrast, using Runx1-deficient cells prepared from targeted mice, we show that the absence of Runx1 in unstimulated CD4+ T cells results in IL-2 derepression. In summary, we propose that high levels of Runx1 in resting CD4+ T cells functions negatively in the regulation of IL-2 transcription, and that TCR activation-mediated down-regulation of Runx1 involves negative auto-regulation of the distal Runx1 promoter and contributes to IL-2 production.
Collapse
Affiliation(s)
- Won Fen Wong
- Department of Molecular Immunology, Institute of Development, Aging, and Cancer, Tohoku University, Aoba-ku, Sendai, Japan
| | | | | | | |
Collapse
|
145
|
Mantel PY, Schmidt-Weber CB. Transforming growth factor-beta: recent advances on its role in immune tolerance. Methods Mol Biol 2011; 677:303-38. [PMID: 20941619 DOI: 10.1007/978-1-60761-869-0_21] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor (TGF-β1) is a pleiotropic cytokine, secreted by immune and nonhematopoietic cells. TGF-β is involved in many different critical processes, such as embryonal development, cellular maturation and differentiation, wound healing, and immune regulation. It maintains immune homeostasis by acting as a potent immune suppressor through inhibition of proliferation, differentiation, activation, and effector function of immune cells. Paradoxically, depending on the context, it displays proinflammatory properties by being a potent chemoattractant for neutrophils and promoting inflammation. In addition, it does not only induce differentiation into the anti-inflammatory Treg cells, but also into the proinflammatory Th17 and Th9 cells and inhibits Th22 differentiation. TGF-β has been demonstrated to be involved in multiple pathologies. In infections, it protects against collateral damages caused by the immune system, but it also promotes immune evasion and chronic infections. In autoimmune diseases, a TGF-β dysfunction leads to the loss of tolerance to self-antigens. In cancer, TGF-β is a potent inhibitor of cell proliferation and acts as a tumor suppressor at the beginning of tumorogenesis. However, once the cells become resistant to TGF-β, it mainly supports tumor growth and metastasis by promoting immune evasion and angiogenesis. In asthma, it is assumed to promote allergen tolerance, but plays a detrimental role in irreversible remodeling of the airways. Despite the high numbers of TGF-β-targeted pathways, it is a promising drug target for treatment of autoimmunity, cancer, fibrosis, if cell specificity can be achieved.This review summarizes the progresses that have been accomplished on the understanding of TGF-β's signaling in the immune homeostasis and its role in pathogenesis.
Collapse
Affiliation(s)
- Pierre-Yves Mantel
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA, USA.
| | | |
Collapse
|
146
|
O'Mahony L, Akdis M, Crameri R, Akdis CA. Novel immunotherapeutic approaches for allergy and asthma. Autoimmunity 2011; 43:493-503. [PMID: 20380589 DOI: 10.3109/08916931003674725] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The immune response is a tightly regulated process, which normally results in protection from infection and tolerance of innocuous environmental antigens. However, in allergic disease, the activated immune response results in a chronic pro-inflammatory state characterized by antibody secretion (IgE) and T cell activation to normally well-tolerated antigens. Currently, the treatment of allergic disease is focused on the suppression of key inflammatory mediators or inflammatory cell populations and include anti-histamines, anti-leukotrienes, β2 adrenergic receptor agonists and corticosteroids. However, these approaches only provide a temporary suppression of disease symptoms. Successful long-term treatment can only be provided by allergen-specific immunotherapy (allergen-SIT), which restores normal immunity against allergens. This review will discuss novel approaches to the management of allergy and asthma by targeting the T regulatory cell via modulation of the commensal microbiota and allergen-SIT.
Collapse
Affiliation(s)
- Liam O'Mahony
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.
| | | | | | | |
Collapse
|
147
|
Akkoc T, Akdis M, Akdis CA. Update in the mechanisms of allergen-specific immunotheraphy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2011; 3:11-20. [PMID: 21217920 PMCID: PMC3005313 DOI: 10.4168/aair.2011.3.1.11] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 09/07/2010] [Indexed: 12/26/2022]
Abstract
Allergic diseases represent a complex innate and adoptive immune response to natural environmental allergens with Th2-type T cells and allergen-specific IgE predominance. Allergen-specific immunotherapy is the most effective therapeutic approach for disregulated immune response towards allergens by enhancing immune tolerance mechanisms. The main aim of immunotherapy is the generation of allergen nonresponsive or tolerant T cells in sensitized patients and downregulation of predominant T cell- and IgE-mediated immune responses. During allergen-specific immunotherapy, T regulatory cells are generated, which secrete IL-10 and induce allergen-specific B cells for the production of IgG4 antibodies. These mechanisms induce tolerance to antigens that reduces allergic symptoms. Although current knowledge highlights the role of T regulatory cell-mediated immunetolerance, definite mechanisms that lead to a successful clinical outcomes of allergen-specific immunotherapy still remains an open area of research.
Collapse
Affiliation(s)
- Tunc Akkoc
- Division of Pediatric Allergy and Immunology, Marmara University, Istanbul, Turkey
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| |
Collapse
|
148
|
Vale-Pereira S, Todo-Bom A, Geraldes L, Schmidt-Weber C, Akdis CA, Mota-Pinto A. FoxP3, GATA-3 and T-bet expression in elderly asthma. Clin Exp Allergy 2010; 41:490-6. [PMID: 21114556 DOI: 10.1111/j.1365-2222.2010.03640.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Asthma is a chronic inflammatory disorder in which Th2, Th1 and suppressive T cells (Tregs) play a role. The transcription factor FoxP3 plays a role in Treg differentiation while T-bet is important for Th1 and GATA-3 for Th2 differentiation from naïve T cells. Recent data show that age-related deregulation of Treg cells is a mechanism of senescence affecting several chronic diseases. It is crucial to understand the behaviour of these cell populations in asthma for elderly patients. OBJECTIVE To evaluate FoxP3, GATA-3 and T-bet gene expression under basal conditions and after in vitro activation in a group of elderly asthmatic compared with age-matched healthy individuals. METHODS Thirty-two elderly asthmatics and 17 healthy elderly individuals were selected. Serum total IgE was measured, and peripheral blood mononuclear cells (PBMCs) were isolated and stimulated in vitro with anti-CD3/anti-CD28, followed by mRNA isolation. After reverse transcription, real-time quantitative PCR was performed and relative quantification was determined 2(-ΔΔCt)(2(-ΔΔCt) method). RESULTS The mean values and standard deviation of FoxP3, GATA-3 and T-bet relative expression for control vs. asthma were 10.2±6.8 vs. 4.8±3.8, 2.4±2.9 vs. 1.7±0.9 and 3.3±2.1 vs. 2.1±1.5, respectively. Healthy individuals showed significantly higher expression of FoxP3 and T-bet; asthmatics had a lower T-bet/GATA-3 ratio, higher serum IgE and a positive significant correlation between total IgE and GATA-3 expression. CONCLUSION AND CLINICAL RELEVANCE Elderly asthmatic patients have lower FoxP3 mRNA expression in PBMC, which can be associated with the sustained inflammatory process and with the decreased immune tolerance by Treg cells. The T-bet deficiency and the correlation of GATA-3 expression with the increase of IgE are characteristics of long-lasting asthma. Changes related to the immunosenescence process could provide an explanation for the minor differences observed between the groups. It is important to clarify persistent modifications in long-lasting asthma in the elderly and adequate future therapeutic approaches.
Collapse
Affiliation(s)
- S Vale-Pereira
- General Pathology Laboratory, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | | | | | | | | | | |
Collapse
|
149
|
Wong WF, Kohu K, Chiba T, Sato T, Satake M. Interplay of transcription factors in T-cell differentiation and function: the role of Runx. Immunology 2010; 132:157-64. [PMID: 21091910 DOI: 10.1111/j.1365-2567.2010.03381.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Over the past years, increasing numbers of distinct subsets have been discovered and identified for a T lymphocytes' entity. Differentiation and function of each T cell subset are controlled by a specific master transcription factor. Importantly, Runt-related transcription factors, particularly Runx1 and Runx3, interplay with these master regulators in various aspects of T cells' immunity. In this review article, we first explain roles of Th-Pok and Runx3 in differentiation of CD4 versus CD8 single positive cells, and later focus on cross-regulation of Th-Pok and Runx3 and their relationship with other factors such as TCR strength. Next, we provide evidences for the direct interplay of Runx1/3 with T-bet and GATA3 during Th1 versus Th2 commitment to activate or silence transcription of signature cytokine genes, IFNγ and IL4. Lastly, we explain feed-forward relationship between Runx1 and Foxp3 and discuss roles of Runx1 in regulatory T cells' suppressive activity. This review highlights an essential importance of Runx molecules in controlling various T cell subsets' differentiation and functions through molecular interplay with the master transcription factors in terms of protein-protein interaction as well as regulation of gene expression.
Collapse
Affiliation(s)
- Won Fen Wong
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Isehara, Japan
| | | | | | | | | |
Collapse
|
150
|
Transcriptional regulation of Foxp3 in regulatory T cells. Immunobiology 2010; 216:678-85. [PMID: 21122941 DOI: 10.1016/j.imbio.2010.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 11/05/2010] [Accepted: 11/05/2010] [Indexed: 12/30/2022]
Abstract
Regulatory T (Treg) cells constitute a unique T-cell lineage that plays a pivotal role in the maintenance of the peripheral tolerance. The transcription factor Foxp3 (Forkhead box P3) was identified as a master regulator for the development and function of Treg cells. It is well defined that Foxp3 expression is critical to program CD4+CD25+ Treg cell development and function; however, the molecular mechanisms that are involved in the regulation of the Foxp3 expression remain unclear. Recent studies have showed an indication that this process is influenced by a number of transcription factors. In this review, we summarize the current knowledge of how Foxp3 expression is controlled at molecular level by focusing on these factors.
Collapse
|