1
|
Solleti SK, Matthews BE, Wu J, Rowe RK. SHIP-1 Differentially Regulates IgE-Induced IL-10 and Antiviral Responses in Human Monocytes. Eur J Immunol 2024:e202451065. [PMID: 39668409 DOI: 10.1002/eji.202451065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
IgE-mediated stimulation of monocytes regulates multiple cellular functions including cellular maturation, cytokine release, antiviral responses, and T-cell differentiation. Expression of the high-affinity IgE receptor, FcεRI, is closely linked to serum IgE levels and atopic disease. The signaling molecules regulating FcεRI effector functions have been well studied in mast cells and basophils; however, less is known about the signaling and regulatory mechanisms in monocytes. This study sought to identify regulators of IgE-mediated cytokine release in human monocytes. SHIP-1 was identified as a negative regulator of IgE-induced IL-10 production. It was also determined that IgE-mediated stimulation and SHIP-1 inhibition decreased antiviral IP-10 production after liposomal poly(I:C) stimulation, indicating differential regulation by SHIP-1 in IgE-driven and antiviral response pathways. SHIP-1 and NF-κB were activated following IgE-mediated stimulation of monocytes, and NF-κB activation was related to both SHIP-1 and FcεRIα cellular expression levels. To our knowledge, this is the first study to identify a role for SHIP-1 in regulating IgE-mediated and antiviral responses in human monocytes. Given the importance of monocytes in inflammation and immune responses, a better understanding of the signaling and regulatory mechanisms downstream of the FcεRI receptor could lead to new therapeutic targets in allergic disease.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Bailey E Matthews
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Jingyi Wu
- Biomedical Genetics and Genomics Graduate Program, University of Rochester Medical Center, Rochester, New York, USA
| | - Regina K Rowe
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
2
|
Mager LF, Krause T, McCoy KD. Interaction of microbiota, mucosal malignancies, and immunotherapy-Mechanistic insights. Mucosal Immunol 2024; 17:402-415. [PMID: 38521413 DOI: 10.1016/j.mucimm.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
The microbiome has emerged as a crucial modulator of host-immune interactions and clearly impacts tumor development and therapy efficacy. The microbiome is a double-edged sword in cancer development and therapy as both pro-tumorigenic and anti-tumorigenic bacterial taxa have been identified. The staggering number of association-based studies in various tumor types has led to an enormous amount of data that makes it difficult to identify bacteria that promote tumor development or modulate therapy efficacy from bystander bacteria. Here we aim to comprehensively summarize the current knowledge of microbiome-host immunity interactions and cancer therapy in various mucosal tissues to find commonalities and thus identify potential functionally relevant bacterial taxa. Moreover, we also review recent studies identifying specific bacteria and mechanisms through which the microbiome modulates cancer development and therapy efficacy.
Collapse
Affiliation(s)
- Lukas F Mager
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada; Department of Internal Medicine I, Faculty of Medicine, University of Tübingen, Germany; M3 Research Center for Malignom, Metabolome and Microbiome, Faculty of Medicine University Tübingen, Germany
| | - Tim Krause
- Department of Internal Medicine I, Faculty of Medicine, University of Tübingen, Germany; M3 Research Center for Malignom, Metabolome and Microbiome, Faculty of Medicine University Tübingen, Germany
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
3
|
Lin YH, Tahara-Hanaoka S, Shibuya A. Human basophils promote IgE-dependent oral allergen-induced anaphylaxis in humanized mice. Allergol Int 2024; 73:345-347. [PMID: 38042753 DOI: 10.1016/j.alit.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/04/2023] Open
Affiliation(s)
- Yu-Hsien Lin
- Department of Immunology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan; R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, Japan.
| | - Akira Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan; R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, Japan.
| |
Collapse
|
4
|
Ligan C, Ma XH, Zhao SL, Zhao W. The regulatory role and mechanism of mast cells in tumor microenvironment. Am J Cancer Res 2024; 14:1-15. [PMID: 38323271 PMCID: PMC10839313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/25/2023] [Indexed: 02/08/2024] Open
Abstract
Mast cells (MCs) have emerged as pivotal contributors to both the defensive immune response and immunomodulation. They also exhibit regulatory functions in modulating pathological processes across various allergic diseases. The impact of MC presence within tumor tissues has garnered considerable attention, yielding conflicting findings. While some studies propose that MCs within tumor tissues promote tumor initiation and progression, others advocate an opposing perspective. Notably, evidence emphasizes the dual role of MCs in cancer, both as promoters and suppressors, is crucial for optimizing cancer treatment strategies. These conflicting viewpoints have generated substantial controversy, underscoring the need for a comprehensive understanding of MC's role in tumor immune responses.
Collapse
Affiliation(s)
- Caryl Ligan
- General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical UniversityNanjing, Jiangsu, China
| | - Xin-Hua Ma
- General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical UniversityNanjing, Jiangsu, China
| | - Shu-Li Zhao
- General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical UniversityNanjing, Jiangsu, China
| | - Wei Zhao
- Department of Pathology, Nanjing First Hospital, Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
5
|
Voskamp AL, Tak T, Gerdes ML, Menafra R, Duijster E, Jochems SP, Kielbasa SM, Kormelink TG, Stam KA, van Hengel OR, de Jong NW, Hendriks RW, Kloet SL, Yazdanbakhsh M, de Jong EC, Gerth van Wijk R, Smits HH. Inflammatory and tolerogenic myeloid cells determine outcome following human allergen challenge. J Exp Med 2023; 220:e20221111. [PMID: 37428185 PMCID: PMC10333709 DOI: 10.1084/jem.20221111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/08/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Innate mononuclear phagocytic system (MPS) cells preserve mucosal immune homeostasis. We investigated their role at nasal mucosa following allergen challenge with house dust mite. We combined single-cell proteome and transcriptome profiling on nasal immune cells from nasal biopsies cells from 30 allergic rhinitis and 27 non-allergic subjects before and after repeated nasal allergen challenge. Biopsies of patients showed infiltrating inflammatory HLA-DRhi/CD14+ and CD16+ monocytes and proallergic transcriptional changes in resident CD1C+/CD1A+ conventional dendritic cells (cDC)2 following challenge. In contrast, non-allergic individuals displayed distinct innate MPS responses to allergen challenge: predominant infiltration of myeloid-derived suppressor cells (MDSC: HLA-DRlow/CD14+ monocytes) and cDC2 expressing inhibitory/tolerogenic transcripts. These divergent patterns were confirmed in ex vivo stimulated MPS nasal biopsy cells. Thus, we identified not only MPS cell clusters involved in airway allergic inflammation but also highlight novel roles for non-inflammatory innate MPS responses by MDSC to allergens in non-allergic individuals. Future therapies should address MDSC activity as treatment for inflammatory airway diseases.
Collapse
Affiliation(s)
- Astrid L. Voskamp
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Tamar Tak
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Maarten L. Gerdes
- Department of Ear, Nose and Throat, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Roberta Menafra
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Ellen Duijster
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Simon P. Jochems
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Szymon M. Kielbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Tom Groot Kormelink
- Department of Exp Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Koen A. Stam
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Nicolette W. de Jong
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Susan L. Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Esther C. de Jong
- Department of Exp Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Roy Gerth van Wijk
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
6
|
He K, Wan T, Wang D, Hu J, Zhou T, Tao W, Wei Z, Lu Q, Zhou R, Tian Z, Flavell RA, Zhu S. Gasdermin D licenses MHCII induction to maintain food tolerance in small intestine. Cell 2023; 186:3033-3048.e20. [PMID: 37327784 DOI: 10.1016/j.cell.2023.05.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/03/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
The intestinal epithelial cells (IECs) constitute the primary barrier between host cells and numerous foreign antigens; it is unclear how IECs induce the protective immunity against pathogens while maintaining the immune tolerance to food. Here, we found IECs accumulate a less recognized 13-kD N-terminal fragment of GSDMD that is cleaved by caspase-3/7 in response to dietary antigens. Unlike the 30-kD GSDMD cleavage fragment that executes pyroptosis, the IEC-accumulated GSDMD cleavage fragment translocates to the nucleus and induces the transcription of CIITA and MHCII molecules, which in turn induces the Tr1 cells in upper small intestine. Mice treated with a caspase-3/7 inhibitor, mice with GSDMD mutation resistant to caspase-3/7 cleavage, mice with MHCII deficiency in IECs, and mice with Tr1 deficiency all displayed a disrupted food tolerance phenotype. Our study supports that differential cleavage of GSDMD can be understood as a regulatory hub controlling immunity versus tolerance in the small intestine.
Collapse
Affiliation(s)
- Kaixin He
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Tingting Wan
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Decai Wang
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Ji Hu
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Tingyue Zhou
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Wanyin Tao
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Zheng Wei
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Qiao Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rongbin Zhou
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Zhigang Tian
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Shu Zhu
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China; School of Data Science, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
7
|
Bitting K, Hedgespeth B, Ehrhardt-Humbert LC, Arthur GK, Schubert AG, Bradding P, Tilley SL, Cruse G. Identification of redundancy between human FcεRIβ and MS4A6A proteins points toward additional complex mechanisms for FcεRI trafficking and signaling. Allergy 2023; 78:1204-1217. [PMID: 36424895 PMCID: PMC10159887 DOI: 10.1111/all.15595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Allergic diseases are triggered by signaling through the high-affinity IgE receptor, FcεRI. In both mast cells (MCs) and basophils, FcεRI is a tetrameric receptor complex comprising a ligand-binding α subunit (FcεRIα), a tetraspan β subunit (FcεRIβ, MS4A2) responsible for trafficking and signal amplification, and a signal transducing dimer of single transmembrane γ subunits (FcεRIγ). However, FcεRI also exists as presumed trimeric complexes that lack FcεRIβ and are expressed on several cell types outside the MC and basophil lineages. Despite known differences between humans and mice in the presence of the trimeric FcεRI complex, questions remain as to how it traffics and whether it signals in the absence of FcεRIβ. We have previously reported that targeting FcεRIβ with exon-skipping oligonucleotides eliminates IgE-mediated degranulation in mouse MCs, but equivalent targeting in human MCs was not effective at reducing degranulation. RESULTS Here, we report that the FcεRIβ-like protein MS4A6A exists in human MCs and compensates for FcεRIβ in FcεRI trafficking and signaling. Human MS4A6A promotes surface expression of FcεRI complexes and facilitates degranulation. MS4A6A and FcεRIβ are encoded by highly related genes within the MS4A gene family that cluster within the human gene loci 11q12-q13, a region linked to allergy and asthma susceptibility. CONCLUSIONS Our data suggest the presence of either FcεRIβ or MS4A6A is sufficient for degranulation, indicating that MS4A6A could be an elusive FcεRIβ-like protein in human MCs that performs compensatory functions in allergic disease.
Collapse
Affiliation(s)
- Katie Bitting
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Barry Hedgespeth
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Lauren C. Ehrhardt-Humbert
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Greer K. Arthur
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Alicia G. Schubert
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| | - Peter Bradding
- Department of Respiratory Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Stephen L. Tilley
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Glenn Cruse
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University. Raleigh, NC 27607, USA
| |
Collapse
|
8
|
Plattner K, Bachmann MF, Vogel M. On the complexity of IgE: The role of structural flexibility and glycosylation for binding its receptors. FRONTIERS IN ALLERGY 2023; 4:1117611. [PMID: 37056355 PMCID: PMC10089267 DOI: 10.3389/falgy.2023.1117611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
It is well established that immunoglobulin E (IgE) plays a crucial role in atopy by binding to two types of Fcε receptors (FcεRI and FcεRII, also known as CD23). The cross-linking of FcεRI-bound IgE on effector cells, such as basophils and mast cells, initiates the allergic response. Conversely, the binding of IgE to CD23 modulates IgE serum levels and antigen presentation. In addition to binding to FcεRs, IgE can also interact with other receptors, such as certain galectins and, in mice, some FcγRs. The binding strength of IgE to its receptors is affected by its valency and glycosylation. While FcεRI shows reduced binding to IgE immune complexes (IgE-ICs), the binding to CD23 is enhanced. There is no evidence that galectins bind IgE-ICs. On the other hand, IgE glycosylation plays a crucial role in the binding to FcεRI and galectins, whereas the binding to CD23 seems to be independent of glycosylation. In this review, we will focus on receptors that bind to IgE and examine how the glycosylation and complexation of IgE impact their binding.
Collapse
Affiliation(s)
- Kevin Plattner
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Correspondence: Monique Vogel
| |
Collapse
|
9
|
Zhang X, Wang A, Chang E, Han B, Xu J, Fu Y, Dong X, Miao S. Effects of dietary tryptophan on the antioxidant capacity and immune response associated with TOR and TLRs/MyD88/NF-κB signaling pathways in northern snakehead, Channa argus (Cantor, 1842). Front Immunol 2023; 14:1149151. [PMID: 37114056 PMCID: PMC10128191 DOI: 10.3389/fimmu.2023.1149151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Dietary tryptophan (Trp) has been shown to influence fish feed intake, growth, immunity and inflammatory responses. The purpose of this study was to investigate the effect and mechanism of Trp on immune system of juvenile northern snakehead (Channa argus Cantor, 1842). Methods A total of 540 fish (10.21 ± 0.11 g) were fed six experimental diets containing graded levels of Trp at 1.9, 3.0, 3.9, 4.8, 5.9 and 6.8 g/kg diet for 70 days, respectively. Results and Discussion The results showed that supplementation of 1.9-4.8 g/kg Trp in diets had no effect on the hepatosomatic index (HSI) and renal index (RI), while dietary 3.9 and 4.8 g/kg Trp significantly increased spleen index (SI) of fish. Dietary 3.9, 4.8, 5.9 and 6.8 g/kg Trp enhanced the total hemocyte count (THC), the activities of total antioxidant capacity (T-AOC) and superoxide dismutase (SOD). Malondinaldehyde (MDA) levels in the blood were significantly decreased by consuming 3.9 and 4.8 g/kg Trp. Fish fed with 3.0 and 3.9 g/kg Trp diets up-regulated interleukin 6 (il-6) and interleukin 8 (il-8) mRNA levels. The expression of tumor necrosis factor α (tnf-α) was highest in fish fed with 3.0 g/kg Trp diet, and the expression of interleukin 1β (il-1β) was highest in fish fed with 3.9 g/kg Trp diet. Dietary 4.8, 5.9 and 6.8 g/kg Trp significantly decreased il-6 and tnf-α mRNA levels in the intestine. Moreover, Trp supplementation was also beneficial to the mRNA expression of interleukin 22 (il-22). Additionally, the mRNA expression levels of target of rapamycin (tor), toll-like receptor-2 (tlr2), toll-like receptor-4 (tlr4), toll-like receptor-5 (tlr5) and myeloid differentiation primary response 88 (myd88) of intestine were significantly up-regulated in fish fed 1.9, 3.0 and 3.9 g/kg Trp diets, and down-regulated in fish fed 4.8, 5.9 and 6.8 g/kg Trp diets. Dietary 4.8 and 5.9 g/kg Trp significantly increased the expression of inhibitor of nuclear factor kappa B kinase beta subunit (ikkβ) and decreased the expression of inhibitor of kappa B (iκbα), but inhibited nuclear transcription factor kappa B (nf-κb) mRNA level. Collectively, these results indicated that dietary 4.8 g/kg Trp could improve antioxidant capacity and alleviate intestinal inflammation associated with TOR and TLRs/MyD88/NF-κB signaling pathways.
Collapse
|
10
|
Allergic Inflammation: Effect of Propolis and Its Flavonoids. Molecules 2022; 27:molecules27196694. [PMID: 36235230 PMCID: PMC9570745 DOI: 10.3390/molecules27196694] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The incidence of allergic diseases and their complications are increasing worldwide. Today, people increasingly use natural products, which has been termed a "return to nature". Natural products with healing properties, especially those obtained from plants and bees, have been used in the prevention and treatment of numerous chronic diseases, including allergy and/or inflammation. Propolis is a multi-component resin rich in flavonoids, collected and transformed by honeybees from buds and plant wounds for the construction and adaptation of their nests. This article describes the current views regarding the possible mechanisms and multiple benefits of flavonoids in combating allergy and allergy-related complications. These benefits arise from flavonoid anti-allergic, anti-inflammatory, antioxidative, and wound healing activities and their effects on microbe-immune system interactions in developing host responses to different allergens. Finally, this article presents various aspects of allergy pathobiology and possible molecular approaches in their treatment. Possible mechanisms regarding the antiallergic action of propolis on the microbiota of the digestive and respiratory tracts and skin diseases as a method to selectively remove allergenic molecules by the process of bacterial biotransformation are also reported.
Collapse
|
11
|
Colas L, Magnan A, Brouard S. Immunoglobulin E response in health and disease beyond allergic disorders. Allergy 2022; 77:1700-1718. [PMID: 35073421 DOI: 10.1111/all.15230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/13/2021] [Accepted: 01/16/2022] [Indexed: 12/24/2022]
Abstract
Immunoglobulin E is the latest discovered of immunoglobulin family and has been long associated with anaphylaxis and worm expulsion. Immunoglobulin E, along with mast cells, basophils, and eosinophils, is also a hallmark of type 2 immunity which is dysregulated in numerous diseases such as asthma, rhinitis, atopic dermatitis, and eosinophilic esophagitis in addition to anaphylaxis as aforementioned. However, recent advances have shed light on IgE regulation and memory explaining the low level of free IgE, the scarcity of IgE plasma cells that are mainly short live and the absence of IgE memory B cells in homeostatic conditions. Furthermore, IgE was implicated in inflammatory conditions beyond allergic disorders where IgE-mediated facilitated antigen presentation can enhance cellular and humoral response against autoantigens in systemic lupus or chronic urticaria leading to more severe disease and even against neoantigen facilitating tumor cell lysis. At last, IgE was unexpectedly associated with allograft rejection or atheromatous cardiovascular diseases where precise mechanisms remain to be deciphered. The purpose of this review is to summarize these recent advances in IgE regulation, biology, and physiopathology beyond allergic diseases opening whole new fields of IgE biology to explore.
Collapse
Affiliation(s)
- Luc Colas
- Plateforme Transversale d'Allergologie et d'immunologie Clinique PFTA Clinique dermatologique CHU de Nantes Nantes France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology Nantes France
| | - Antoine Magnan
- Hôpital Foch, Suresnes; Université de Versailles Saint‐Quentin Paris‐Saclay; INRAe Paris France
| | - Sophie Brouard
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology Nantes France
- Labex IGO Nantes France
- Centre d’Investigation Clinique en Biothérapie Centre de ressources biologiques (CRB) Nantes France
| |
Collapse
|
12
|
Araújo GRL, Marques HS, Santos MLC, da Silva FAF, da Brito BB, Correa Santos GL, de Melo FF. Helicobacter pylori infection: How does age influence the inflammatory pattern? World J Gastroenterol 2022; 28:402-411. [PMID: 35125826 PMCID: PMC8790560 DOI: 10.3748/wjg.v28.i4.402] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/21/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
The inflammatory pattern during Helicobacter pylori (H. pylori) infection is changeable and complex. During childhood, it is possible to observe a predominantly regulatory response, evidenced by high concentrations of key cytokines for the maintenance of Treg responses such as TGF-β1 and IL-10, in addition to high expression of the transcription factor FOXP3. On the other hand, there is a predominance of cytokines associated with the Th1 and Th17 responses among H. pylori-positive adults. In the last few years, the participation of the Th17 response in the gastric inflammation against H. pylori infection has been highlighted due to the high levels of TGF-β1 and IL-17 found in this infectious scenario, and growing evidence has supported a close relationship between this immune response profile and unfavorable outcomes related to the infection. Moreover, this cytokine profile might play a pivotal role in the effectiveness of anti-H. pylori vaccines. It is evident that age is one of the main factors influencing the gastric inflammatory pattern during the infection with H. pylori, and understanding the immune response against the bacterium can assist in the development of alternative prophylactic and therapeutic strategies against the infection as well as in the comprehension of the pathogenesis of the outcomes related to that microorganism.
Collapse
Affiliation(s)
- Glauber Rocha Lima Araújo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Brazil
| | - Hanna Santos Marques
- Campus Vitória da Conquista, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista 45083-900, Brazil
| | | | | | - Breno Bittencourt da Brito
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Brazil
| | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Brazil
| |
Collapse
|
13
|
Arthur GK, Cruse G. Regulation of Trafficking and Signaling of the High Affinity IgE Receptor by FcεRIβ and the Potential Impact of FcεRIβ Splicing in Allergic Inflammation. Int J Mol Sci 2022; 23:ijms23020788. [PMID: 35054974 PMCID: PMC8776166 DOI: 10.3390/ijms23020788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 12/23/2022] Open
Abstract
Mast cells are tissue-resident immune cells that function in both innate and adaptive immunity through the release of both preformed granule-stored mediators, and newly generated proinflammatory mediators that contribute to the generation of both the early and late phases of the allergic inflammatory response. Although mast cells can be activated by a vast array of mediators to contribute to homeostasis and pathophysiology in diverse settings and contexts, in this review, we will focus on the canonical setting of IgE-mediated activation and allergic inflammation. IgE-dependent activation of mast cells occurs through the high affinity IgE receptor, FcεRI, which is a multimeric receptor complex that, once crosslinked by antigen, triggers a cascade of signaling to generate a robust response in mast cells. Here, we discuss FcεRI structure and function, and describe established and emerging roles of the β subunit of FcεRI (FcεRIβ) in regulating mast cell function and FcεRI trafficking and signaling. We discuss current approaches to target IgE and FcεRI signaling and emerging approaches that could target FcεRIβ specifically. We examine how alternative splicing of FcεRIβ alters protein function and how manipulation of splicing could be employed as a therapeutic approach. Targeting FcεRI directly and/or IgE binding to FcεRI are promising approaches to therapeutics for allergic inflammation. The characteristic role of FcεRIβ in both trafficking and signaling of the FcεRI receptor complex, the specificity to IgE-mediated activation pathways, and the preferential expression in mast cells and basophils, makes FcεRIβ an excellent, but challenging, candidate for therapeutic strategies in allergy and asthma, if targeting can be realized.
Collapse
Affiliation(s)
- Greer K. Arthur
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC 27607, USA;
| | - Glenn Cruse
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC 27607, USA
- Correspondence: ; Tel.: +1-919-515-8865
| |
Collapse
|
14
|
de Sousa T, Ribeiro M, Sabença C, Igrejas G. The 10,000-Year Success Story of Wheat! Foods 2021; 10:2124. [PMID: 34574233 PMCID: PMC8467621 DOI: 10.3390/foods10092124] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Wheat is one of the most important cereal crops in the world as it is used in the production of a diverse range of traditional and modern processed foods. The ancient varieties einkorn, emmer, and spelt not only played an important role as a source of food but became the ancestors of the modern varieties currently grown worldwide. Hexaploid wheat (Triticum aestivum L.) and tetraploid wheat (Triticum durum Desf.) now account for around 95% and 5% of the world production, respectively. The success of this cereal is inextricably associated with the capacity of its grain proteins, the gluten, to form a viscoelastic dough that allows the transformation of wheat flour into a wide variety of staple forms of food in the human diet. This review aims to give a holistic view of the temporal and proteogenomic evolution of wheat from its domestication to the massively produced high-yield crop of our day.
Collapse
Affiliation(s)
- Telma de Sousa
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal; (T.d.S.); (M.R.); (C.S.)
- Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, 2825-149 Lisbon, Caparica, Portugal
| | - Miguel Ribeiro
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal; (T.d.S.); (M.R.); (C.S.)
- Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, 2825-149 Lisbon, Caparica, Portugal
| | - Carolina Sabença
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal; (T.d.S.); (M.R.); (C.S.)
- Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, 2825-149 Lisbon, Caparica, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal; (T.d.S.); (M.R.); (C.S.)
- Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, 2825-149 Lisbon, Caparica, Portugal
| |
Collapse
|
15
|
Bertho N, Meurens F. The pig as a medical model for acquired respiratory diseases and dysfunctions: An immunological perspective. Mol Immunol 2021; 135:254-267. [PMID: 33933817 DOI: 10.1016/j.molimm.2021.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
By definition no model is perfect, and this also holds for biology and health sciences. In medicine, murine models are, and will be indispensable for long, thanks to their reasonable cost and huge choice of transgenic strains and molecular tools. On the other side, non-human primates remain the best animal models although their use is limited because of financial and obvious ethical reasons. In the field of respiratory diseases, specific clinical models such as sheep and cotton rat for bronchiolitis, or ferret and Syrian hamster for influenza and Covid-19, have been successfully developed, however, in these species, the toolbox for biological analysis remains scarce. In this view the porcine medical model is appearing as the third, intermediate, choice, between murine and primate. Herein we would like to present the pros and cons of pig as a model for acquired respiratory conditions, through an immunological point of view. Indeed, important progresses have been made in pig immunology during the last decade that allowed the precise description of immune molecules and cell phenotypes and functions. These progresses might allow the use of pig as clinical model of human respiratory diseases but also as a species of interest to perform basic research explorations.
Collapse
Affiliation(s)
| | - François Meurens
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon S7N5E3, Canada
| |
Collapse
|
16
|
Cao J, Li C, Ma P, Ding Y, Gao J, Jia Q, Zhu J, Zhang T. Effect of kaempferol on IgE-mediated anaphylaxis in C57BL/6 mice and LAD2 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 79:153346. [PMID: 33002828 DOI: 10.1016/j.phymed.2020.153346] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 09/05/2020] [Accepted: 09/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Immunoglobulin E (IgE)-mediated mast cell (MC) activation is crucial in multiple allergic diseases. Parkinson disease protein 7 (DJ-1) and Lyn kinase were reported as the receptor-proximal events in IgE receptor (FcεRI) signals in human MC. Kaempferol, a natural flavonol mainly derived from the rhizome of traditional Chinese herb Kaempferia galanga L. (Zingiberaceae), has been known to inhibit allergic reactions, but it was limited to the receptor-distal signals on rat basophilic leukemia cells. A thorough investigation of the inhibitory effects of kaempferol on human MC has not been done. PURPOSE To investigate the inhibitory effects of kaempferol on IgE-mediated anaphylaxis in vivo and in human MCs, as well as the mechanism underlying its effects, especially the receptor-proximal signals. METHODS IgE-mediated passive cutaneous anaphylaxis and systemic anaphylaxis model were applied to elucidate the antiallergic activity of kaempferol in vivo. The degranulation assay, calcium imaging, the release of cytokines and chemokines on the laboratory of allergic disease 2 (LAD2) cells were used to evaluate the antiallergic effect of kaempferol in vitro. Western blot analysis was performed to investigate the DJ-1/Lyn signaling pathway and downstream molecules. Kinase activity assay, immunofluorescence, and molecular docking were conducted to confirm the influence of kaempferol on DJ-1/Lyn molecules. RESULTS Kaempferol dose-dependently attenuated ovalbumin/IgE-induced mice paw swelling, primary MC activation from paw skin, as well as rehabilitated the hypothermia, and reduced the serum concentrations of histamine, tumor necrosis factor-alpha, interleukin-8, and monocyte chemo-attractant protein-1. Additionally, kaempferol suppressed IgE-mediated LAD2 cell degranulation and calcium fluctuation. Remarkably, kaempferol was found to bind with DJ-1 protein, and initially prevented DJ-1 from translocating to the plasma membrane, thereby inhibited full activation of Lyn, and eventually restrained those receptor-distal signaling molecules, involved Syk, Btk, PLCγ, IP3R, PKC, MAPKs, Akt and NF-κB. CONCLUSION Kaempferol could be used as a DJ-1 modulator for preventing MC-mediated allergic disorders through attenuating Lyn activation.
Collapse
Affiliation(s)
- Jiao Cao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chaomei Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Pengyu Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Ding
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiapan Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qianqian Jia
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Zhu
- Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China
| | - Tao Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
17
|
Nonlethal Plasmodium yoelii Infection Drives Complex Patterns of Th2-Type Host Immunity and Mast Cell-Dependent Bacteremia. Infect Immun 2020; 88:IAI.00427-20. [PMID: 32958528 PMCID: PMC7671899 DOI: 10.1128/iai.00427-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022] Open
Abstract
Malaria strongly predisposes to bacteremia, which is associated with sequestration of parasitized red blood cells and increased gastrointestinal permeability. The mechanisms underlying this disruption are poorly understood. Here, we evaluated the expression of factors associated with mast cell activation and malaria-associated bacteremia in a rodent model. C57BL/6J mice were infected with Plasmodium yoeliiyoelli 17XNL, and blood and tissues were collected over time to assay for circulating levels of bacterial 16S DNA, IgE, mast cell protease 1 (Mcpt-1) and Mcpt-4, Th1 and Th2 cytokines, and patterns of ileal mastocytosis and intestinal permeability. Malaria strongly predisposes to bacteremia, which is associated with sequestration of parasitized red blood cells and increased gastrointestinal permeability. The mechanisms underlying this disruption are poorly understood. Here, we evaluated the expression of factors associated with mast cell activation and malaria-associated bacteremia in a rodent model. C57BL/6J mice were infected with Plasmodium yoeliiyoelli 17XNL, and blood and tissues were collected over time to assay for circulating levels of bacterial 16S DNA, IgE, mast cell protease 1 (Mcpt-1) and Mcpt-4, Th1 and Th2 cytokines, and patterns of ileal mastocytosis and intestinal permeability. The anti-inflammatory cytokines (interleukin-4 [IL-4], IL-6, and IL-10) and MCP-1/CCL2 were detected early after P. yoeliiyoelii 17XNL infection. This was followed by the appearance of IL-9 and IL-13, cytokines known for their roles in mast cell activation and growth-enhancing activity as well as IgE production. Later increases in circulating IgE, which can induce mast cell degranulation, as well as Mcpt-1 and Mcpt-4, were observed concurrently with bacteremia and increased intestinal permeability. These results suggest that P. yoeliiyoelii 17XNL infection induces the production of early cytokines that activate mast cells and drive IgE production, followed by elevated IgE, IL-9, and IL-13 that maintain and enhance mast cell activation while disrupting the protease/antiprotease balance in the intestine, contributing to epithelial damage and increased permeability.
Collapse
|
18
|
Nakamura M, Souri EA, Osborn G, Laddach R, Chauhan J, Stavraka C, Lombardi S, Black A, Khiabany A, Khair DO, Figini M, Winship A, Ghosh S, Montes A, Spicer JF, Bax HJ, Josephs DH, Lacy KE, Tsoka S, Karagiannis SN. IgE Activates Monocytes from Cancer Patients to Acquire a Pro-Inflammatory Phenotype. Cancers (Basel) 2020; 12:E3376. [PMID: 33203088 PMCID: PMC7698027 DOI: 10.3390/cancers12113376] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
IgE contributes to host-protective functions in parasitic and bacterial infections, often by monocyte and macrophage recruitment. We previously reported that monocytes contribute to tumour antigen-specific IgE-mediated tumour growth restriction in rodent models. Here, we investigate the impact of IgE stimulation on monocyte response, cellular signalling, secretory and tumour killing functions. IgE cross-linking on human monocytes with polyclonal antibodies to mimic formation of immune complexes induced upregulation of co-stimulatory (CD40, CD80, CD86), and reduced expression of regulatory (CD163, CD206, MerTK) monocyte markers. Cross-linking and tumour antigen-specific IgE antibody-dependent cellular cytotoxicity (ADCC) of cancer cells by cancer patient-derived monocytes triggered release of pro-inflammatory mediators (TNFα, MCP-1, IL-10, CXCL-10, IL-1β, IL-6, IL-23). High intratumoural gene expression of these mediators was associated with favourable five-year overall survival in ovarian cancer. IgE cross-linking of trimeric FcεRI on monocytes stimulated the phosphorylation of intracellular protein kinases widely reported to be downstream of mast cell and basophil tetrameric FcεRI signalling. These included recently-identified FcεRI pathway kinases Fgr, STAT5, Yes and Lck, which we now associate with monocytes. Overall, anti-tumour IgE can potentiate pro-inflammatory signals, and prime tumour cell killing by human monocytes. These findings will inform the development of IgE monoclonal antibody therapies for cancer.
Collapse
Affiliation(s)
- Mano Nakamura
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
| | - Elmira Amiri Souri
- Department of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, London WC2B 4BG, UK; (E.A.S.); (S.T.)
| | - Gabriel Osborn
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
| | - Roman Laddach
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- Department of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, London WC2B 4BG, UK; (E.A.S.); (S.T.)
| | - Jitesh Chauhan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Chara Stavraka
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Sara Lombardi
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Anna Black
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Atousa Khiabany
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Duaa O. Khair
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
| | - Mariangela Figini
- Biomarker Unit, Department of Applied Research and Technology Development, Fondazione, IRCCS Istituto Nazionale dei Tumouri Milano, 20133 Milan, Italy;
| | - Anna Winship
- Department of Medical Oncology and Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (A.W.); (S.G.); (A.M.)
| | - Sharmistha Ghosh
- Department of Medical Oncology and Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (A.W.); (S.G.); (A.M.)
| | - Ana Montes
- Department of Medical Oncology and Clinical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (A.W.); (S.G.); (A.M.)
| | - James F. Spicer
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Heather J. Bax
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Debra H. Josephs
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Katie E. Lacy
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, London WC2B 4BG, UK; (E.A.S.); (S.T.)
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Tower Wing, 9th Floor, Guy’s Hospital, London SE1 9RT, UK; (M.N.); (G.O.); (R.L.); (J.C.); (C.S.); (S.L.); (A.B.); (A.K.); (D.O.K.); (H.J.B.); (D.H.J.); (K.E.L.)
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| |
Collapse
|
19
|
Pellefigues C. IgE Autoreactivity in Atopic Dermatitis: Paving the Road for Autoimmune Diseases? Antibodies (Basel) 2020; 9:E47. [PMID: 32911788 PMCID: PMC7551081 DOI: 10.3390/antib9030047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Atopic dermatitis (AD) is a common skin disease affecting 20% of the population beginning usually before one year of age. It is associated with the emergence of allergen-specific IgE, but also with autoreactive IgE, whose function remain elusive. This review discusses current knowledge relevant to the mechanisms, which leads to the secretion of autoreactive IgE and to the potential function of these antibodies in AD. Multiple autoantigens have been described to elicit an IgE-dependent response in this context. This IgE autoimmunity starts in infancy and is associated with disease severity. Furthermore, the overall prevalence of autoreactive IgE to multiple auto-antigens is high in AD patients. IgE-antigen complexes can promote a facilitated antigen presentation, a skewing of the adaptive response toward type 2 immunity, and a chronic skin barrier dysfunction and inflammation in patients or AD models. In AD, skin barrier defects and the atopic immune environment facilitate allergen sensitization and the development of other IgE-mediated allergic diseases in a process called the atopic march. AD is also associated epidemiologically with several autoimmune diseases showing autoreactive IgE secretion. Thus, a potential outcome of IgE autoreactivity in AD could be the development of further autoimmune diseases.
Collapse
Affiliation(s)
- Christophe Pellefigues
- INSERM UMRS1149-CNRS ERL8252, Team «Basophils and Mast cells in Immunopathology», Centre de recherche sur l'inflammation (CRI), Inflamex, DHU Fire, Université de Paris, 75018 Paris, France
| |
Collapse
|
20
|
Monaghan KL, Farris BY, Zheng W, Wan ECK. Characterization of Immune Cells and Proinflammatory Mediators in the Pulmonary Environment. J Vis Exp 2020. [PMID: 32658184 DOI: 10.3791/61359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immune cell expansion, activation, and trafficking to the lungs, which are controlled by the expression of multiple cytokines and chemokines, may be altered by severe brain injury. This is evidenced by the fact that pneumonia is a major cause of mortality in patients who have suffered from ischemic stroke. The goal of this protocol is to describe the use of multicolor flow cytometric analysis to identify 13 types of immune cells in the lungs of mice, including alveolar macrophages, interstitial macrophages, CD103+ or CD11b+ dendritic cells (DCs), plasmacytoid DCs, eosinophils, monocytes/monocyte-derived cells, neutrophils, lymphoid-derived T and B cells, NK cells, and NKT cells, following ischemic stroke induction by transient middle cerebral artery occlusion. Moreover, we describe the preparation of lung homogenates using a bead homogenization method, to determine the expression levels of 13 different cytokines or chemokines simultaneously by multiplex bead arrays coupled with flow cytometric analysis. This protocol can also be used to investigate the pulmonary immune response in other disease settings, such as infectious lung disease or allergic disease.
Collapse
Affiliation(s)
- Kelly L Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Breanne Y Farris
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Wen Zheng
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University
| | - Edwin C K Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University; Department of Neuroscience, West Virginia University; Rockefeller Neuroscience Institute, West Virginia University;
| |
Collapse
|
21
|
Kanagaratham C, El Ansari YS, Sallis BF, Hollister BMA, Lewis OL, Minnicozzi SC, Oyoshi MK, Rosen R, Nurko S, Fiebiger E, Oettgen HC. Omeprazole inhibits IgE-mediated mast cell activation and allergic inflammation induced by ingested allergen in mice. J Allergy Clin Immunol 2020; 146:884-893.e5. [PMID: 32194041 DOI: 10.1016/j.jaci.2020.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with eosinophilic esophagitis have increased numbers of mucosal mast cells. Administration of the proton pump inhibitor omeprazole can reduce both esophageal mast cell and eosinophil numbers and attenuate type 2 inflammation in these subjects. OBJECTIVE Given that maintenance of an acidic environment within granules is important for mast cell homeostasis, we sought to evaluate the effects of omeprazole on mast cell functions including development, IgE:FcεRI-mediated activation, and responses to food allergen. METHODS Mast cell degranulation, cytokine secretion, and early signaling events in the FcεRI pathway, including protein kinase phosphorylation and Ca2+ flux, were measured after IgE crosslinking in murine bone marrow-derived mast cells and human cord blood-derived mast cells. The effects of omeprazole on these responses were investigated as was its impact on mast cell-dependent anaphylaxis and food allergy phenotypes in vivo. RESULTS Murine and human mast cells treated with omeprazole exhibited diminished degranulation and release of cytokines and histamine in response to allergen. In murine mast cells, phosphorylation of protein kinases, ERK and SYK, was decreased. Differentiation of mast cells from bone marrow progenitors was also inhibited. IgE-mediated passive anaphylaxis was blunted in mice treated with omeprazole as was allergen-induced mast cell expansion and mast cell activation in the intestine in a model of food allergy. CONCLUSIONS Our findings suggest that omeprazole targets pathways important for the differentiation and activation of murine mast cells and for the manifestations of food allergy and anaphylaxis.
Collapse
Affiliation(s)
- Cynthia Kanagaratham
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Yasmeen S El Ansari
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Institute of Laboratory Medicine, Philipps University Marburg, Germany
| | | | | | - Owen L Lewis
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Samantha C Minnicozzi
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Michiko K Oyoshi
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Rachel Rosen
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Samuel Nurko
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Edda Fiebiger
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Hans C Oettgen
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
22
|
Boukid F, Prandi B, Faccini A, Sforza S. A Complete Mass Spectrometry (MS)-Based Peptidomic Description of Gluten Peptides Generated During In Vitro Gastrointestinal Digestion of Durum Wheat: Implication for Celiac Disease. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:1481-1490. [PMID: 31049870 DOI: 10.1007/s13361-019-02212-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/02/2019] [Indexed: 05/29/2023]
Abstract
Resistance of gluten to gastrointestinal digestion is involved in immune-mediated adverse reactions to wheat, since several peptides produced by the incomplete digestion are able to trigger, in predisposed individuals, the immune response responsible, for instance, of celiac disease (CD) and other adverse reactions. Even if several peptides have been identified, an exhaustive description of the peptidome generated by wheat digestion is lacking. To this end, in the present work, durum wheat proteins were fractionated, digested, and then subjected to various proteomic techniques, including single stage and multiple stage mass spectrometry (MS) (SDS-PAGE, UPLC/ESI-MS, UPLC/ESI-MS/MS, and LTQ-Orbitrap). Based on SDS-PAGE, although proteins were severely degraded after in vitro gastrointestinal digestion, some differences were observed among protein profile of the different digests. Through untargeted UPLC techniques, 227 peptide sequences were identified, with only few sequences shared by the different digests. In particular, 9 gluten peptides involved in CD were identified. Based on target proteomic, the quantification of these peptides revealed significant (p ≤ 0.05) differences among the different extracts. Taken together, all the proteomic tools confirmed that gluten digestion is closely related to the matrix regardless of wheat genotype.
Collapse
Affiliation(s)
- Fatma Boukid
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parco Area delle Scienze 23/A, Parma, 43124, Italy
| | - Barbara Prandi
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy.
- Department of Human Sciences and Promotion of Quality of Life, Telematic University San Raffaele Roma, via Val Cannuta, 247, Rome, Italy.
| | - Andrea Faccini
- Interdepartmental Centre for Measurements, University of Parma, Parco Area Delle Scienze 23/A, 43124, Parma, Italy
| | - Stefano Sforza
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parco Area delle Scienze 23/A, Parma, 43124, Italy
| |
Collapse
|
23
|
Mouse Models for Food Allergies: Where Do We Stand? Cells 2019; 8:cells8060546. [PMID: 31174293 PMCID: PMC6627293 DOI: 10.3390/cells8060546] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Food allergies are a steadily increasing health and economic problem. Immunologically, food allergic reactions are caused by pathological, allergen-specific Th2 responses resulting in IgE-mediated mast cell degranulation and associated inflammatory reactions. Clinically, food allergies are characterized by local inflammation of the mouth mucosa, the face, the throat, the gastrointestinal tract, are frequently paralleled by skin reactions, and can result in life-threatening anaphylactic reactions. To better understand food allergies and establish novel treatment options, mouse models are indispensable. This review discusses the available mouse food allergy models, dividing them into four categories: (1) adjuvant-free mouse models, (2) mouse models relying on adjuvants to establish allergen-specific Th2 responses, (3) mouse models using genetically-modified mouse strains to allow for easier sensitization, and (4) humanized mouse models in which different immunodeficient mouse strains are reconstituted with human immune or stem cells to investigate humanized immune responses. While most of the available mouse models can reproducibly portray the immunological parameters of food allergy (Th2 immune responses, IgE production and mast cell activation/expansion), so far, the recreation of the clinical parameters has proven more difficult. Therefore, up to now none of the available mouse models can reproduce the complete human pathology.
Collapse
|
24
|
León MA, Palma C, Hernández C, Sandoval M, Cofre C, Perez-Mateluna G, Borzutzky A, Harris PR, Serrano CA. Helicobacter pylori pediatric infection changes FcεRI expression in dendritic cells and Treg profile in vivo and in vitro. Microbes Infect 2019; 21:449-455. [PMID: 31128278 DOI: 10.1016/j.micinf.2019.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/15/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
Abstract
H. pylori infection shows an inverse relationship with allergies. Dendritic cells regulate mucosal immune responses including the induction of T regulatory cells which are fundamental in Helicobacter pylori-induced dampening of allergies. In this respect expression of high-affinity IgE receptor (FcεRI) has been associated with a regulatory dendritic cell profile. Therefore we aimed to evaluate possible mechanisms by which H. pylori infection might modify atopy in pediatric patients. Here we show that H. pylori-infected children exhibited both increased expression of FcεRI on peripheral myeloid and plasmacytoid dendritic cells and higher levels of Foxp3 and Latency Associated Peptide on T regulatory cells. Moreover, exposure to H. pylori drove increased FcεRI expression and IL-10 secretion by both pediatric H. pylori-exposed monocyte derived dendritic cells and T cells. Finally, we show a positive correlation between expression of FcεRI in circulating myeloid DCs and total Treg cells, suggesting that in children, H. pylori infection may have a modulating role in atopy, mediated by both altered surface expression of FcεRI on children's DC and an increased T regulatory cell profile.
Collapse
Affiliation(s)
- Miguel A León
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Palma
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Caroll Hernández
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Sandoval
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Colomba Cofre
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Perez-Mateluna
- Department of Pediatric Infectious Diseases and Immunology, Millennium Institute on Immunology and Immunotherapy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Arturo Borzutzky
- Department of Pediatric Infectious Diseases and Immunology, Millennium Institute on Immunology and Immunotherapy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paul R Harris
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina A Serrano
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
25
|
Amison RT, Cleary SJ, Riffo-Vasquez Y, Bajwa M, Page CP, Pitchford SC. Platelets Play a Central Role in Sensitization to Allergen. Am J Respir Cell Mol Biol 2019; 59:96-103. [PMID: 29365287 DOI: 10.1165/rcmb.2017-0401oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Platelet activation occurs in patients with allergic inflammation, and platelets can be activated directly by allergen via an IgE-dependent process. Platelets have been shown to activate APCs such as CD11c+ dendritic cells in vitro. Although CD11c+ dendritic cells are a requisite for allergen sensitization, the role of platelets in this process is unknown. In this study, we investigated whether platelets were necessary for allergen sensitization. Balb/c mice sensitized to ovalbumin were exposed to subsequent aerosolized allergen (ovalbumin challenge). We analyzed lung CD11c+ cell activation, colocalization with platelets, and some other indices of inflammation. The role of platelets at the time of allergen sensitization was assessed through platelet depletion experiments restricted to the period of sensitization. Platelets colocalized with airway CD11c+ cells, and this association increased after allergen sensitization as well as after subsequent allergen exposure. Temporary platelet depletion (>95%) at the time of allergen sensitization led to a suppression of IgE and IL-4 synthesis and to a decrease in the pulmonary recruitment of eosinophils, macrophages, and lymphocytes after subsequent allergen exposure. Furthermore, in mice previously depleted of platelets at the time of sensitization, the recovered platelet population was shown to have reduced expression of FcεRI. Pulmonary CD11c+ cell recruitment was suppressed in these mice after allergen challenge, suggesting that the migration of CD11c+ cells in vivo may be dependent on direct platelet recognition of allergen. We conclude that platelets are necessary for efficient host sensitization to allergen. This propagates the subsequent inflammatory response during secondary allergen exposure and increases platelet association with airway CD11c+ cells.
Collapse
Affiliation(s)
- Richard T Amison
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Simon J Cleary
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Yanira Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Maidda Bajwa
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Simon C Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| |
Collapse
|
26
|
Moñino-Romero S, Erkert L, Schmidthaler K, Diesner SC, Sallis BF, Pennington L, Jardetzky T, Oettgen HC, Bohle B, Fiebiger E, Szépfalusi Z. The soluble isoform of human FcɛRI is an endogenous inhibitor of IgE-mediated mast cell responses. Allergy 2019; 74:236-245. [PMID: 30030936 DOI: 10.1111/all.13567] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/01/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The soluble isoform of FcɛRI, the high-affinity IgE receptor (sFcεRI), is a protein of the IgE network with poorly defined functions. OBJECTIVE To define cellular sources and signals that result in the production of human sFcεRI and study its in vivo functions. METHODS FcεRI-transfected human cell lines (MelJuso), human monocyte-derived dendritic cells (moDCs), and murine bone marrow-derived mast cells (MC) were stimulated by FcεRI cross-linking and release of sFcεRI was analyzed (ELISA, Western Blot). Lysosomal-associated membrane protein 1 degranulation assays and human basophil activation tests (BATs) were used to study IgE-dependent activation. Recombinant sFcεRI (rsFcεRI) was used to assess its role in murine models of anaphylaxis with WT (wild-type) and IgE-/- (IgE-deficient) mice. RESULTS Antigen-specific cross-linking of IgE-loaded FcɛRI on MelJuso cells that express the trimeric or tetrameric receptor isoform induced the production of sFcεRI. Using MCs and moDCs, we confirmed that IgE/FcɛRI activation induces sFcɛRI release. We demonstrated that generation of sFcɛRI requires Src phosphorylation and endo/lysosomal acidification. In experimental mouse models, sFcɛRI diminishes the severity of IgE-mediated anaphylaxis. BATs confirmed that, comparable to the anti-IgE monoclonal antibody omalizumab, sFcɛRI is an inhibitor of the human innate IgE effector axis, implying that sFcɛRI and omalizumab potentially inhibit each other in vivo. CONCLUSION sFcɛRI is produced after antigen-specific IgE/FcɛRI-mediated activation signals and functions as an endogenous inhibitor of IgE loading to FcɛRI and IgE-mediated activation. Our results imply, therefore, that sFcɛRI contributes to a negative regulatory feedback loop that aims at preventing overshooting responses after IgE-mediated immune activation.
Collapse
Affiliation(s)
- S. Moñino-Romero
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - L. Erkert
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - K. Schmidthaler
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| | - S. C. Diesner
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| | - B. F. Sallis
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| | - L. Pennington
- Department of Structural Biology; School of Medicine; Stanford University; Stanford California
| | - T. Jardetzky
- Department of Structural Biology; School of Medicine; Stanford University; Stanford California
| | - H. C. Oettgen
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Division of Immunology; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
| | - B. Bohle
- Department of Pathophysiology and Allergy Research; Medical University of Vienna; Vienna Austria
| | - E. Fiebiger
- Division of Gastroenterology, Hepatology and Nutrition; Department of Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| | - Z. Szépfalusi
- Department of Pediatrics and Adolescent Medicine; Medical University Vienna; Vienna Austria
| |
Collapse
|
27
|
Hung L, Obernolte H, Sewald K, Eiwegger T. Human ex vivo and in vitro disease models to study food allergy. Asia Pac Allergy 2019; 9:e4. [PMID: 30740352 PMCID: PMC6365658 DOI: 10.5415/apallergy.2019.9.e4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Food allergy is a growing global public health concern. As treatment strategies are currently limited to allergen avoidance and emergency interventions, there is an increasing demand for appropriate models of food allergy for the development of new therapeutics. Many models of food allergy rely heavily on the use of animals, and while useful, many are unable to accurately reflect the human system. In order to bridge the gap between in vivo animal models and clinical trials with human patients, human models of food allergy are of great importance. This review will summarize the commonly used human ex vivo and in vitro models of food allergy and highlight their advantages and limitations regarding how accurately they represent the human in vivo system. We will cover biopsy-based systems, precision cut organ slices, and coculture systems as well as organoids and organ-on-a-chip. The availability of appropriate experimental models will allow us to move forward in the field of food allergy research, to search for effective treatment options and to further explore the cause and progression of this disorder.
Collapse
Affiliation(s)
- Lisa Hung
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Helena Obernolte
- Department of Preclinical Pharmacology and In-Vitro Toxicology, Fraunhofer ITEM, Hannover, Germany
| | - Katherina Sewald
- Department of Preclinical Pharmacology and In-Vitro Toxicology, Fraunhofer ITEM, Hannover, Germany
| | - Thomas Eiwegger
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Clinical Immunology and Allergy, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
28
|
Tang XZ, Jung JB, Allen CDC. A case of mistaken identity: The MAR-1 antibody to mouse FcεRIα cross-reacts with FcγRI and FcγRIV. J Allergy Clin Immunol 2019; 143:1643-1646.e6. [PMID: 30639345 DOI: 10.1016/j.jaci.2018.11.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/10/2018] [Accepted: 11/16/2018] [Indexed: 12/01/2022]
Affiliation(s)
- Xin-Zi Tang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, Calif; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, Calif; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, Calif
| | - James B Jung
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, Calif; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, Calif; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, Calif
| | - Christopher D C Allen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, Calif; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, Calif; Department of Anatomy, University of California, San Francisco, San Francisco, Calif.
| |
Collapse
|
29
|
Baran W, Oehrl S, Ahmad F, Döbel T, Alt C, Buske-Kirschbaum A, Schmitz M, Schäkel K. Phenotype, Function, and Mobilization of 6-Sulfo LacNAc-Expressing Monocytes in Atopic Dermatitis. Front Immunol 2018; 9:1352. [PMID: 29977237 PMCID: PMC6021776 DOI: 10.3389/fimmu.2018.01352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/31/2018] [Indexed: 01/31/2023] Open
Abstract
Mononuclear phagocytes (MPs) are important immune regulatory cells in atopic dermatitis (AD). We previously identified 6-sulfo LacNAc-expressing monocytes (slanMo) as TNF-α- and IL-23-producing cells in psoriatic skin lesions and as inducers of IFN-γ-, IL-17-, and IL-22-producing T cells. These cytokines are also upregulated in AD and normalize with treatment, as recently shown for dupilumab-treated patients. We here asked for the role of slanMo in AD. Increased numbers of slanMo were found in AD skin lesions. In difference to other MPs in AD, slanMo lacked expression of FcɛRI, CD1a, CD14, and CD163. slanMo from blood of patients with AD expressed increased levels of CD86 and produced IL-12 and TNF-α at higher amounts than CD14+ monocytes and myeloid dendritic cells. While CD14+ monocytes from patients with AD revealed a reduced IL-12 production, we observed no difference in the cytokine production comparing slanMo in AD and healthy controls. Interestingly, experimentally induced mental stress, a common trigger of flares in patients with AD, rapidly mobilized slanMo which retained their high TNF-α-producing capacity. This study identifies slanMo as a distinct population of inflammatory cells in skin lesions and as proinflammatory blood cells in patients with AD. slanMo may, therefore, represent a potent future target for treatment of AD.
Collapse
Affiliation(s)
- Wojciech Baran
- Department of Dermatology, Venerology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Stephanie Oehrl
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Fareed Ahmad
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Christina Alt
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | | | - Marc Schmitz
- Institute of Immunology, Medical Faculty, Technical University of Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), Medical Faculty, Technical University of Dresden, Dresden, Germany
| | - Knut Schäkel
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
Abstract
Immunoglobulin E-mediated food allergy is rapidly developing into a global health problem. Publicly available therapeutic intervention strategies are currently restricted to allergen avoidance and emergency treatments. To gain a better understanding of the disease pathophysiology so that new therapies can be developed, major research efforts have been put into studying food allergy in mice. Animal models should reflect the human pathology as closely as possible to allow for a rapid translation of basic science observations to the bedside. In this regard, experimental models of food allergy provide significant challenges for research because of discrepancies between the presentation of disease in humans and mice. The goal of this review is to give a summary of commonly used murine disease models and to discuss how they relate to the human condition. We will focus on epicutaneous sensitization models, on mouse strains that sensitize spontaneously to food as seen in humans, and on models in humanized animals. In summary, expanding the research toolbox of experimental food allergy provides an important step toward closing gaps in our understanding of the derailing immune mechanism that underlies the human disease. The availability of additional experimental models will provide exciting opportunities to discover new intervention points for the treatment of food allergies. (Cell Mol Gastroenterol Hepatol 2018;x:x).
Collapse
Key Words
- Allergen Challenge
- Allergen Sensitization
- Anaphylaxis
- EPIT, epicutaneous immunotherapy
- Epictutaneous Sensitization
- FCER1A, high-affinity immunoglobulin epsilon receptor subunit alpha
- FCERIA
- FcεRI, high-affinity immunoglobulin E receptor
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HSC, hematopoietic stem cell
- Humanized Model
- IL, interleukin
- Ig, immunoglobulin
- IgE
- LCT, long chain triglycerides
- MCPT, mouse mast cell protease
- MCT, medium chain triglycerides
- Murine Models of Food Allergy
- OIT, oral immunotherapy
- PBMC, peripheral blood mononuclear cell
- Spontaneous Sensitization
- TSLP, thymic stromal lymphopoietin
- Th, T helper
- Treg, regulatory T cell
- WASP, Wiskott–Aldrich syndrome protein
Collapse
|
31
|
Berings M, Gevaert P, De Ruyck N, Derycke L, Holtappels G, Pilette C, Bachert C, Lambrecht BN, Dullaers M. FcεRI expression and IgE binding by dendritic cells and basophils in allergic rhinitis and upon allergen immunotherapy. Clin Exp Allergy 2018; 48:970-980. [PMID: 29682789 DOI: 10.1111/cea.13157] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 04/06/2018] [Accepted: 04/10/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND In humans, both basophils and dendritic cells (DCs) express the high-affinity IgE receptor (FcεRI). OBJECTIVE To gain more insight into the relation between serum IgE levels and FcεRI expression and IgE binding by DCs and basophils in house dust mite (HDM) allergy and during subcutaneous immunotherapy (SCIT). METHODS We measured FcεRI, IgE and HDM allergen on DCs (conventional type 2 DCs, cDC2s; plasmacytoid dendritic cells, pDCs) and basophils by flow cytometry in 22 non-allergic vs 52 allergic subjects and upon HDM SCIT in 28 allergic subjects. IgE levels were measured in serum. RESULTS Serum IgE correlated differentially with FcεRI expression and IgE binding depending on cell type and allergic status. In non-allergic subjects, FcεRI/IgE surface densities increased with serum IgE to a significantly stronger degree on basophils compared to cDC2s. By contrast, in allergic subjects FcεRI/IgE surface densities increased with serum IgE to a slightly stronger degree on cDC2s compared to basophils. In addition, the data set suggests sequential loading of IgE onto FcεRI expressed by these cells (basophils>cDC2s>pDCs). Finally, HDM SCIT induced a temporary increase in serum IgE, which was paralleled by a peak in FcεRI and IgE on DCs, but not on basophils. CONCLUSIONS & CLINICAL RELEVANCE This study provides a comprehensive insight into the relation between serum IgE and FcεRI/IgE on basophils and DC subsets. The novel finding that HDM SCIT induces a temporary increase in FcεRI expression on DCs, but not on basophils, can be an incentive for future research on the potential tolerogenic role of IgE/FcεRI signalling in DCs in the setting of allergen immunotherapy.
Collapse
Affiliation(s)
- M Berings
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium.,Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium
| | - P Gevaert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - N De Ruyck
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - L Derycke
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - G Holtappels
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - C Pilette
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - C Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - M Dullaers
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium.,Clinical Immunology Research Lab, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
32
|
Jensen-Jarolim E, Bax HJ, Bianchini R, Crescioli S, Daniels-Wells TR, Dombrowicz D, Fiebiger E, Gould HJ, Irshad S, Janda J, Josephs DH, Levi-Schaffer F, O'Mahony L, Pellizzari G, Penichet ML, Redegeld F, Roth-Walter F, Singer J, Untersmayr E, Vangelista L, Karagiannis SN. AllergoOncology: Opposite outcomes of immune tolerance in allergy and cancer. Allergy 2018; 73:328-340. [PMID: 28921585 PMCID: PMC6038916 DOI: 10.1111/all.13311] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2017] [Indexed: 12/11/2022]
Abstract
While desired for the cure of allergy, regulatory immune cell subsets and nonclassical Th2-biased inflammatory mediators in the tumour microenvironment can contribute to immune suppression and escape of tumours from immunological detection and clearance. A key aim in the cancer field is therefore to design interventions that can break immunological tolerance and halt cancer progression, whereas on the contrary allergen immunotherapy exactly aims to induce tolerance. In this position paper, we review insights on immune tolerance derived from allergy and from cancer inflammation, focusing on what is known about the roles of key immune cells and mediators. We propose that research in the field of AllergoOncology that aims to delineate these immunological mechanisms with juxtaposed clinical consequences in allergy and cancer may point to novel avenues for therapeutic interventions that stand to benefit both disciplines.
Collapse
Affiliation(s)
- E Jensen-Jarolim
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - H J Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - R Bianchini
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
| | - S Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
| | - T R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - D Dombrowicz
- INSERM, CHU Lille, European Genomic Institute of Diabetes, Institut Pasteur de Lille, U1011 - Recepteurs Nucleaires, Maladies Cardiovasculaires et Diabete, Universite de Lille, Lille, France
| | - E Fiebiger
- Division of Gastroenterology, Hepatology and Nutrition Research, Department Medicine Research, Childrens' University Hospital Boston, Boston, MA, USA
| | - H J Gould
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - S Irshad
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| | - J Janda
- Faculty of Science, Charles University, Prague, Czech Republic
| | - D H Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - F Levi-Schaffer
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Unit, The Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - L O'Mahony
- Molecular Immunology, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - G Pellizzari
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Hospital, London, UK
| | - M L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, CA, USA
| | - F Redegeld
- Faculty of Science, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - F Roth-Walter
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University Vienna, Vienna, Austria
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - J Singer
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - E Untersmayr
- Centre of Pathophysiology, Infectiology & Immunology, Institute of Pathophysiology & Allergy Research, Medical University Vienna, Vienna, Austria
| | - L Vangelista
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - S N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, Guy's Hospital, London, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
| |
Collapse
|
33
|
Lexmond WS, Goettel JA, Sallis BF, McCann K, Rings EHHM, Jensen-Jarolim E, Nurko S, Snapper SB, Fiebiger E. Spontaneous food allergy in Was -/- mice occurs independent of FcεRI-mediated mast cell activation. Allergy 2017; 72:1916-1924. [PMID: 28600891 DOI: 10.1111/all.13219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Food allergies are a growing health problem, and the development of therapies that prevent disease onset is limited by the lack of adjuvant-free experimental animal models. We compared allergic sensitization in patients with food allergy or Wiskott-Aldrich syndrome (WAS) and defined whether spontaneous disease in Was-/- mice recapitulates the pathology of a conventional disease model and/or human food allergy. METHODS Comparative ImmunoCAP ISAC microarray was performed in patients with food allergy or WAS. Spontaneous food allergy in Was-/- mice was compared to an adjuvant-based model in wild-type mice (WT-OVA/alum). Intestinal and systemic anaphylaxis was assessed, and the role of the high-affinity IgE Fc receptor (FcεRI) in allergic sensitization was evaluated using Was-/- Fcer1a-/- mice. RESULTS Polysensitization to food was detected in both WAS and food-allergic patients which was recapitulated in the Was-/- model. Oral administration of ovalbumin (OVA) in Was-/- mice induced low titers of OVA-specific IgE compared to the WT-OVA/alum model. Irrespectively, 79% of Was-/- mice developed allergic diarrhea following oral OVA challenge. Systemic anaphylaxis occurred in Was-/- mice (95%) with a mortality rate >50%. Spontaneous sensitization and intestinal allergy occurred independent of FcεRI expression on mast cells (MCs) and basophils. CONCLUSIONS Was-/- mice provide a model of food allergy with the advantage of mimicking polysensitization and low food-antigen IgE titers as observed in humans with clinical food allergy. This model will facilitate studies on aberrant immune responses during spontaneous disease development. Our results imply that therapeutic targeting of the IgE/FcεRI activation cascade will not affect sensitization to food.
Collapse
Affiliation(s)
- W. S. Lexmond
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
- Department of Medicine; Harvard Medical School; Boston MA USA
| | - J. A. Goettel
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
- Department of Medicine; Harvard Medical School; Boston MA USA
| | - B. F. Sallis
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
| | - K. McCann
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
| | - E. H. H. M. Rings
- Departments of Pediatrics; Erasmus Medical Center; Erasmus University; Rotterdam The Netherlands
- University Medical Center Leiden; Leiden University; Leiden The Netherlands
| | - E. Jensen-Jarolim
- Center of Pathophysiology, Infectiology and Immunology; Institute of Pathophysiology and Allergy Research; Medical University of Vienna; Vienna Austria
- Comparative Medicine; The Interuniversity Messerli Research Institute; University of Veterinary Medicine Vienna, Medical University of Vienna, University of Vienna; Vienna Austria
- Allergy Care; Allergy Diagnosis and Study Center; Vienna Austria
| | - S. Nurko
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
- Department of Medicine; Harvard Medical School; Boston MA USA
| | - S. B. Snapper
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
- Department of Medicine; Harvard Medical School; Boston MA USA
| | - E. Fiebiger
- Department of Pediatrics; Division of Gastroenterology, Hepatology and Nutrition; Boston Children's Hospital; Boston MA USA
- Department of Medicine; Harvard Medical School; Boston MA USA
| |
Collapse
|
34
|
Lozano-Ojalvo D, López-Fandiño R. Immunomodulating peptides for food allergy prevention and treatment. Crit Rev Food Sci Nutr 2017; 58:1629-1649. [PMID: 28102702 DOI: 10.1080/10408398.2016.1275519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Among the most promising strategies currently assayed against IgE-mediated allergic diseases stands the possibility of using immunomodulating peptides to induce oral tolerance toward offending food allergens or even to prevent allergic sensitization. This review focuses on the beneficial effects of food derived immunomodulating peptides on food allergy, which can be directly exerted in the intestinal tract or once being absorbed through the intestinal epithelial barrier to interact with immune cells. Food peptides influence intestinal homeostasis by maintaining and reinforcing barrier function or affecting intestinal cell-signalling to nearby immune cells and mucus secretion. In addition, they can stimulate cells of the innate and adaptive immune system while supressing inflammatory responses. Peptides represent an attractive alternative to whole allergens to enhance the safety and efficacy of immunotherapy treatments. The conclusions drawn from curative and preventive experiments in murine models are promising, although there is a need for more pre-clinical studies to further explore the immunomodulating strategy and its mechanisms and for a deeper knowledge of the peptide sequence and structural requirements that determine the immunoregulatory function.
Collapse
Affiliation(s)
- Daniel Lozano-Ojalvo
- a Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM) , Madrid , Spain
| | - Rosina López-Fandiño
- a Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM) , Madrid , Spain
| |
Collapse
|
35
|
Moingeon P, Mascarell L. Differences and similarities between sublingual immunotherapy of allergy and oral tolerance. Semin Immunol 2017; 30:52-60. [PMID: 28760498 DOI: 10.1016/j.smim.2017.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/13/2017] [Indexed: 12/27/2022]
Abstract
Allergen immunotherapy is the only treatment altering the natural course of IgE-mediated allergies. Whereas the subcutaneous route for immunotherapy (SCIT) has been historically considered as a reference, we discuss herein the relative advantages of the sublingual and oral routes as alternatives to SCIT in order to elicit allergen-specific tolerance. The buccal and gut immune systems are similarly organized to favor immune tolerance to antigens/allergens, due to the presence of tolerogenic dendritic cells and macrophages promoting the differentiation of CD4+ regulatory T cells. Sublingual immunotherapy (SLIT) is now established as a valid treatment option, with clinical efficacy demonstrated in allergic rhinoconjunctivitis (to either grass, tree, weed pollens or mite allergens) and encouraging results obtained in the management of mild/moderate allergic asthma. While still exploratory, oral immunotherapy (OIT) has shown promising results in the desensitization of patients with food allergies. We review at both biological and clinical levels the perspectives currently pursued for those two mucosal routes.
Collapse
Affiliation(s)
- Philippe Moingeon
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France.
| | - Laurent Mascarell
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France
| |
Collapse
|
36
|
Tordesillas L, Berin MC, Sampson HA. Immunology of Food Allergy. Immunity 2017; 47:32-50. [DOI: 10.1016/j.immuni.2017.07.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/29/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023]
|
37
|
Oettgen HC. Fifty years later: Emerging functions of IgE antibodies in host defense, immune regulation, and allergic diseases. J Allergy Clin Immunol 2017; 137:1631-1645. [PMID: 27263999 DOI: 10.1016/j.jaci.2016.04.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 01/15/2023]
Abstract
Fifty years ago, after a long search, IgE emerged as the circulating factor responsible for triggering allergic reactions. Its extremely low concentration in plasma created significant hurdles for scientists working to reveal its identity. We now know that IgE levels are invariably increased in patients affected by atopic conditions and that IgE provides the critical link between the antigen recognition role of the adaptive immune system and the effector functions of mast cells and basophils at mucosal and cutaneous sites of environmental exposure. This review discusses the established mechanisms of action of IgE in pathologic immediate hypersensitivity, as well as its multifaceted roles in protective immunity, control of mast cell homeostasis, and its more recently revealed immunomodulatory functions.
Collapse
Affiliation(s)
- Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
38
|
Jensen-Jarolim E, Pali-Schöll I, Roth-Walter F. Outstanding animal studies in allergy I. From asthma to food allergy and anaphylaxis. Curr Opin Allergy Clin Immunol 2017; 17:169-179. [PMID: 28346234 PMCID: PMC5424575 DOI: 10.1097/aci.0000000000000363] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Animal models published within the past 18 months on asthma, food allergy and anaphylaxis, all conditions of rising public health concern, were reviewed. RECENT FINDINGS While domestic animals spontaneously develop asthma, food allergy and anaphylaxis, in animal models, divergent sensitization and challenge routes, dosages, intervals and antigens are used to induce asthmatic, food allergic or anaphylactic phenotypes. This must be considered in the interpretation of results. Instead of model antigens, gradually relevant allergens such as house dust mite in asthma, and food allergens like peanut, apple and peach in food allergy research were used. Novel engineered mouse models such as a mouse with a T-cell receptor for house dust mite allergen Der p 1, or with transgenic human hFcγR genes, facilitated the investigation of single molecules of interest. Whole-body plethysmography has become a state-of-the-art in-vivo readout in asthma research. In food allergy and anaphylaxis research, novel techniques were developed allowing real-time monitoring of in-vivo effects following allergen challenge. Networks to share tissues were established as an effort to reduce animal experiments in allergy which cannot be replaced by in-vitro measures. SUMMARY Natural and artificial animal models were used to explore the pathophysiology of asthma, food allergy and anaphylaxis and to improve prophylactic and therapeutic measures. Especially the novel mouse models mimicking molecular aspects of the complex immune network in asthma, food allergy and anaphylaxis will facilitate proof-of-concept studies under controlled conditions.
Collapse
Affiliation(s)
- Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
| |
Collapse
|
39
|
Potential risk of repeated nasal vaccination that induces allergic reaction with mucosal IgE and airway eosinophilic infiltration in cynomolgus macaques infected with H5N1 highly pathogenic avian influenza virus. Vaccine 2017; 35:1008-1017. [PMID: 28109707 DOI: 10.1016/j.vaccine.2017.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 12/29/2016] [Accepted: 01/06/2017] [Indexed: 01/22/2023]
Abstract
The efficacy and detrimental effect of mucosal vaccination with an inactivated influenza vaccine were examined in a macaque model by intranasal administration with small amounts of inactivated whole virus particles and challenge by a human-derived H5N1 highly pathogenic avian influenza virus infection. Repeated nasal inoculation with the whole particle vaccine of an inactivated virus, A/duck/Hokkaido/Vac-3/2007 (H5N1) (Vac-3), induced antigen-specific IgA and IgG antibody production in nasal swabs and plasma. Vac-3-specific IgE production was also found in the nasal swabs. Nasal vaccination with Vac-3 induced broader cross-clade neutralization activity than did subcutaneous vaccination. After challenge infection, repeated nasal vaccination almost completely prevented the propagation of virus in the upper and lower airways and protected cynomolgus macaques from viral pneumonia by induction of IgA-producing B cells in the lungs. On the other hand, eosinophil clusters were observed in the lungs of vaccinated macaques. Although Vac-3-specific IgE antibody and IL-13 levels were decreased after infection compared to those before infection and no anaphylaxis in vaccinated macaques was detected after challenge infection, our results suggest that we have to pay attention to potential allergic responses at repeated nasal vaccination, especially in people who have an airway allergy.
Collapse
|
40
|
FcεRI cross-linking reduces cord blood dendritic cell responsiveness to LPS. J Allergy Clin Immunol 2016; 139:1992-1994.e3. [PMID: 27919739 DOI: 10.1016/j.jaci.2016.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/16/2016] [Accepted: 11/02/2016] [Indexed: 11/20/2022]
|
41
|
Lexmond WS, Goettel JA, Lyons JJ, Jacobse J, Deken MM, Lawrence MG, DiMaggio TH, Kotlarz D, Garabedian E, Sackstein P, Nelson CC, Jones N, Stone KD, Candotti F, Rings EH, Thrasher AJ, Milner JD, Snapper SB, Fiebiger E. FOXP3+ Tregs require WASP to restrain Th2-mediated food allergy. J Clin Invest 2016; 126:4030-4044. [PMID: 27643438 PMCID: PMC5096801 DOI: 10.1172/jci85129] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
In addition to the infectious consequences of immunodeficiency, patients with Wiskott-Aldrich syndrome (WAS) often suffer from poorly understood exaggerated immune responses that result in autoimmunity and elevated levels of serum IgE. Here, we have shown that WAS patients and mice deficient in WAS protein (WASP) frequently develop IgE-mediated reactions to common food allergens. WASP-deficient animals displayed an adjuvant-free IgE-sensitization to chow antigens that was most pronounced for wheat and soy and occurred under specific pathogen-free as well as germ-free housing conditions. Conditional deletion of Was in FOXP3+ Tregs resulted in more severe Th2-type intestinal inflammation than that observed in mice with global WASP deficiency, indicating that allergic responses to food allergens are dependent upon loss of WASP expression in this immune compartment. While WASP-deficient Tregs efficiently contained Th1- and Th17-type effector differentiation in vivo, they failed to restrain Th2 effector responses that drive allergic intestinal inflammation. Loss of WASP was phenotypically associated with increased GATA3 expression in effector memory FOXP3+ Tregs, but not in naive-like FOXP3+ Tregs, an effect that occurred independently of increased IL-4 signaling. Our results reveal a Treg-specific role for WASP that is required for prevention of Th2 effector cell differentiation and allergic sensitization to dietary antigens.
Collapse
Affiliation(s)
- Willem S. Lexmond
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeremy A. Goettel
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan J. Lyons
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Justin Jacobse
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Marion M. Deken
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Monica G. Lawrence
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Thomas H. DiMaggio
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Daniel Kotlarz
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, Munich, Germany
| | | | - Paul Sackstein
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Celeste C. Nelson
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Nina Jones
- Clinical Research Directorate/Clinical Monitoring Research Program (CMRP), Leidos Biomedical Research Inc., National Cancer Institute (NCI) Campus at Frederick, Frederick, Maryland, USA
| | - Kelly D. Stone
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Fabio Candotti
- Genetics and Molecular Biology Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Edmond H.H.M. Rings
- Departments of Pediatrics, Erasmus University, Erasmus Medical Center, Rotterdam and Leiden University, University Medical Center Leiden, Leiden, Netherlands
| | - Adrian J. Thrasher
- Great Ormond Street Hospital NHS Trust, London and Institute of Child Health, University College London, London, United Kingdom
| | - Joshua D. Milner
- Genetics and Pathogenesis of Allergy Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Scott B. Snapper
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Ponce M, Diesner SC, Szépfalusi Z, Eiwegger T. Markers of tolerance development to food allergens. Allergy 2016; 71:1393-404. [PMID: 27286276 DOI: 10.1111/all.12953] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 12/30/2022]
Abstract
IgE-mediated reactions to food allergens are the most common cause of anaphylaxis in childhood. Although allergies to cow's milk, egg, or soy proteins, in contrast to peanut and tree nut allergens, resolve within the first 6 years of life in up to 60% due to natural tolerance development, this process is not well understood. At present, there is no cure or treatment for food allergy that would result in an induction of tolerance to the symptom-eliciting food. Avoidance, providing an emergency plan and education, is the standard of treatment. Oral immunotherapeutic approaches have been proven reasonable efficacy; however, they are associated with high rates of side-effects and low numbers of patients achieving tolerance. Nevertheless, mechanisms that take place during oral immunotherapy may help to understand tolerance development. On the basis of these therapeutic interventions, events like loss of basophil activation and induction of regulatory lymphocyte subsets and of blocking antibodies have been described. Their functional importance at a clinical level, however, remains to be investigated in detail. Consequently, there is eminent need to understand the process of tolerance development to food allergens and define biomarkers to develop and monitor new treatment strategies for food allergy.
Collapse
Affiliation(s)
- M. Ponce
- Department of Pediatrics and Adolescent Medicine; Medical University of Vienna; Vienna Austria
| | - S. C. Diesner
- Department of Pediatrics and Adolescent Medicine; Medical University of Vienna; Vienna Austria
| | - Z. Szépfalusi
- Department of Pediatrics and Adolescent Medicine; Medical University of Vienna; Vienna Austria
| | - T. Eiwegger
- Department of Pediatrics and Adolescent Medicine; Medical University of Vienna; Vienna Austria
- Division of Immunology and Allergy, Food allergy and Anaphylaxis Program; The Department of Paediatrics; Hospital for Sick Children; Research Institute, Physiology and Experimental Medicine Program; The University of Toronto; Toronto ON Canada
| |
Collapse
|
43
|
Mudde ACA, Lexmond WS, Blumberg RS, Nurko S, Fiebiger E. Eosinophilic esophagitis: published evidences for disease subtypes, indications for patient subpopulations, and how to translate patient observations to murine experimental models. World Allergy Organ J 2016; 9:23. [PMID: 27458501 PMCID: PMC4947322 DOI: 10.1186/s40413-016-0114-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder of the esophagus and commonly classified as a Th2-type allergy. Major advances in our understanding of the EoE pathophysiology have recently been made, but clinicians struggle with highly unpredictable therapy responses indicative of phenotypic diversity within the patient population. Here, we summarize evidences for the existence of EoE subpopulations based on diverse inflammatory characteristics of the esophageal tissue in EoE. Additionally, clinical characteristics of EoE patients support the concept of disease subtypes. We conclude that clinical and experimental evidences indicate that EoE is an umbrella term for conditions that are unified by esophageal eosinophilia but that several disease subgroups with various inflammatory esophageal patterns and/or different clinical features exist. We further discuss strategies to study the pathophysiologic differences as observed in EoE patients in murine experimental EoE. Going forward, models of EoE that faithfully mimic EoE subentities as defined in humans will be essential because mechanistic studies on triggers which regulate the onset of diverse EoE subpopulations are not feasible in patients. Understanding how and why different EoE phenotypes develop will be a first and fundamental step to establish strategies that integrate individual variations of the EoE pathology into personalized therapy.
Collapse
Affiliation(s)
- Anne C A Mudde
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Willem S Lexmond
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| | - Samuel Nurko
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA ; Center for Motility and Functional Gastrointestinal Disorders, Boston, MA USA ; Eosinophilic Gastrointestinal Disease Center, Boston Children's Hospital, Boston, MA USA
| | - Edda Fiebiger
- Department of Medicine, Harvard Medical School, and Division of Gastroenterology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| |
Collapse
|
44
|
Maisonnasse P, Bouguyon E, Piton G, Ezquerra A, Urien C, Deloizy C, Bourge M, Leplat JJ, Simon G, Chevalier C, Vincent-Naulleau S, Crisci E, Montoya M, Schwartz-Cornil I, Bertho N. The respiratory DC/macrophage network at steady-state and upon influenza infection in the swine biomedical model. Mucosal Immunol 2016; 9:835-49. [PMID: 26530136 DOI: 10.1038/mi.2015.105] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/12/2015] [Indexed: 02/04/2023]
Abstract
Human and mouse respiratory tracts show anatomical and physiological differences, which will benefit from alternative experimental models for studying many respiratory diseases. Pig has been recognized as a valuable biomedical model, in particular for lung transplantation or pathologies such as cystic fibrosis and influenza infection. However, there is a lack of knowledge about the porcine respiratory immune system. Here we segregated and studied six populations of pig lung dendritic cells (DCs)/macrophages (Mθs) as follows: conventional DCs (cDC) 1 and cDC2, inflammatory monocyte-derived DCs (moDCs), monocyte-derived Mθs, and interstitial and alveolar Mθs. The three DC subsets present migratory and naive T-cell stimulation capacities. As observed in human and mice, porcine cDC1 and cDC2 were able to induce T-helper (Th)1 and Th2 responses, respectively. Interestingly, porcine moDCs increased in the lung upon influenza infection, as observed in the mouse model. Pig cDC2 shared some characteristics observed in human but not in mice, such as the expression of FCɛRIα and Langerin, and an intra-epithelial localization. This work, by unraveling the extended similarities of the porcine and human lung DC/Mθ networks, highlights the relevance of pig, both as an exploratory model of DC/Mθ functions and as a model for human inflammatory lung pathologies.
Collapse
Affiliation(s)
- P Maisonnasse
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - E Bouguyon
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - G Piton
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - A Ezquerra
- Dpto. de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - C Urien
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - C Deloizy
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - M Bourge
- I2BC, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | - J-J Leplat
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - G Simon
- Anses, Laboratoire de Ploufragan/Plouzané, Unité Virologie Immunologie Porcines, BP53, Ploufragan, France.,Université Européenne de Bretagne, Rennes, France
| | - C Chevalier
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - S Vincent-Naulleau
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - E Crisci
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain
| | - M Montoya
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain.,The Pirbright Institute, Surrey, UK
| | - I Schwartz-Cornil
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - N Bertho
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| |
Collapse
|
45
|
Cao PP, Shi LL, Xu K, Yao Y, Liu Z. Dendritic cells in inflammatory sinonasal diseases. Clin Exp Allergy 2016; 46:894-906. [PMID: 27159777 DOI: 10.1111/cea.12755] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are critical in linking the innate and adaptive immune responses, which have been implicated in the pathogenesis of many immune and inflammatory diseases as well as the development of tumours. The role of DCs in the pathophysiology of lung diseases has been widely studied. However, the phenotype, subset and function of DCs in upper airways under physiological or pathological conditions remain largely undefined. Allergic rhinitis (AR) and chronic rhinosinusitis (CRS) are two important upper airway diseases with a high worldwide prevalence. Aberrant innate and adaptive immune responses have been considered to play an important role in the pathogenesis of AR and CRS. To this end, understanding the function of DCs in shaping the immune responses in sinonasal mucosa is critical in exploring the pathogenic mechanisms underlying AR and CRS as well as in developing novel therapeutic strategies. This review summarizes the phenotype, subset, function and regulation of DCs in sinonasal mucosa, particularly in the setting of AR and CRS. Furthermore, this review discusses the perspectives for future research and potential clinical utility focusing on DC pathways in the context of AR and CRS.
Collapse
Affiliation(s)
- P-P Cao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L-L Shi
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - K Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Z Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Shen C, Detry B, Lecocq M, Pilette C. A novel IgA/Delta-like 4/Notch axis induces immunosuppressive activity in human dendritic cells. Clin Immunol 2016; 168:37-46. [PMID: 27117596 DOI: 10.1016/j.clim.2016.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Chong Shen
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Bruno Detry
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Marylène Lecocq
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Charles Pilette
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium.
| |
Collapse
|
47
|
IgE-tailpiece associates with α-1-antitrypsin (A1AT) to protect IgE from proteolysis without compromising its ability to interact with FcεRI. Sci Rep 2016; 6:20509. [PMID: 26842628 PMCID: PMC4740804 DOI: 10.1038/srep20509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/05/2016] [Indexed: 11/08/2022] Open
Abstract
Several splice variants of IgE exist in human plasma, including a variant called IgE-tailpiece (IgE-tp) that differs from classical IgE by the replacement of two carboxy-terminal amino acids with eight novel residues that include an ultimate cysteine. To date, the role of the secreted IgE-tp isoform in human immunity is unknown. We show that levels of IgE-tp are raised in helminth-infected donors, and that both the classical form of IgE (IgE-c) and IgE-tp interact with polymers of the serine protease inhibitor alpha-1-antitrypsin (A1AT). The association of IgE-tp with A1AT polymers in plasma protects the antibody from serine protease-mediated degradation, without affecting the functional interaction of IgE-tp with important receptors, including FcεR1. That polymers of A1AT protect IgE from degradation by helminth proteases may explain why these common and normally non-disease causing polymorphic variants of A1AT have been retained by natural selection. The observation that IgE can be complexed with polymeric forms of A1AT may therefore have important consequences for our understanding of the pathophysiology of pulmonary diseases that arise either as a consequence of A1AT-deficiency or through IgE-mediated type 1 hypersensitivity responses.
Collapse
|
48
|
Yu SJ, Liao EC, Tsai JJ. Effects of local nasal immunotherapy in allergic airway inflammation: Using urea denatured Dermatophagoides pteronyssinus. Hum Vaccin Immunother 2016; 11:915-21. [PMID: 25933184 DOI: 10.1080/21645515.2015.1012029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Despite improvements in anti-allergy medication, the prevalence of allergic airway inflammation remains high, affecting up to 40% of the population worldwide. Allergen immunotherapy is effective for inducing tolerance but has the adverse effect of severe allergic reaction. This can be avoided by denaturing with urea. In this study, we demonstrated that the serum level of allergen-specific IgE in mice sensitized with native Dermatophagoides pteronyssinus (Der p) crude extract after receiving local nasal immunotherapy (LNIT) with urea-denatured Der p crude extract (DN-Dp) significantly decreased compared to that in the normal saline (NS) treatment group. Expressions of IL-4 were significantly reduced in lung tissues after treatment. Inflammation around the bronchial epithelium improved and airway hypersensitivity was down-regulated. LNIT with DN-Dp can down-regulate IL-1b, IL-6 and TNF-a expression and then decrease Der p-induced allergic airway inflammation. This therapeutic modality may be used as an alternative treatment for airway allergic diseases.
Collapse
Affiliation(s)
- Sheng-Jie Yu
- a Institute of Biomedical Sciences; National Chung Hsing University ; Taichung , Taiwan
| | | | | |
Collapse
|
49
|
Platzer B, Stout M, Fiebiger E. Functions of dendritic-cell-bound IgE in allergy. Mol Immunol 2015; 68:116-9. [PMID: 26052071 DOI: 10.1016/j.molimm.2015.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 04/27/2015] [Accepted: 05/17/2015] [Indexed: 12/20/2022]
Abstract
Immunoglobulin E (IgE) functions as an Fc-receptor-bound antigen sensor for mast cells and basophils, the classical effector cells of allergy. A cell-bound IgE pool is formed when monomeric IgE binds to FcɛRI, the high affinity IgE Fc receptor on these cells, and minor amounts of antigen are sufficient to trigger the pro-allergic innate IgE effector axis. Additionally, FcɛRI is constitutively expressed on human dendritic cells (DCs), and thus the latter cell type also receives signals via cell-bound IgE. Notably, steady-state expression of FcɛRI on DCs is absent in SPF-housed mice. How DCs integrate IgE/FcɛRI-derived signals into their sentinel functions as gatekeepers of immunity was therefore only recently studied with transgenic mice that phenocopy human FcɛRI expression. In this review, we summarize advances in our understanding of the functions of DC-bound IgE which demonstrate that IgE-mediated activation of DCs in allergic Th2-type inflammation appears to be immune regulatory rather than pro-inflammatory.
Collapse
Affiliation(s)
- Barbara Platzer
- Division of Gastroenterology and Nutrition, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Madeleine Stout
- Division of Gastroenterology and Nutrition, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Edda Fiebiger
- Division of Gastroenterology and Nutrition, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Mechanism of the antigen-independent cytokinergic SPE-7 IgE activation of human mast cells in vitro. Sci Rep 2015; 5:9538. [PMID: 25892150 PMCID: PMC4402612 DOI: 10.1038/srep09538] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/09/2015] [Indexed: 12/27/2022] Open
Abstract
Release of pro-inflammatory mediators by mast cells is a key feature of allergic disease. The ‘dogma’ is that IgE molecules merely sensitise mast cells by binding FcεRI prior to cross-linking by multivalent allergen, receptor aggregation and mast cell activation. However, certain monoclonal IgE antibodies have been shown to elicit mast cell activation in an antigen-independent cytokinergic manner, and DNP-specific murine SPE-7 IgE is the most highly cytokinergic antibody known. We show that both monovalent hapten and recombinant SPE-7 IgE Fab inhibit its cytokinergic activity as measured by mast cell degranulation and TNF-α release. Using SPE-7 IgE, a non-cytokinergic human IgE and a poorly cytokinergic murine IgE, we reveal that interaction of the Fab region of ‘free’ SPE-7 IgE with the Fab of FcεRI-bound SPE-7 IgE is the basis of its cytokinergic activity. We rule out involvement of IgE Fc, Cε1 and Cλ/κ domains, and propose that ‘free’ SPE-7 IgE binds to FcεRI-bound SPE-7 IgE by an Fv-Fv interaction. Initial formation of a tri-molecular complex (one ‘free’ IgE molecule cross-linking two receptor-bound IgE molecules) leads to capture of further ‘free’ and receptor-bound IgEs to form larger clusters that trigger mast cell activation.
Collapse
|