51
|
Huang JY, Lyons-Cohen MR, Gerner MY. Information flow in the spatiotemporal organization of immune responses. Immunol Rev 2022; 306:93-107. [PMID: 34845729 PMCID: PMC8837692 DOI: 10.1111/imr.13046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022]
Abstract
Immune responses must be rapid, tightly orchestrated, and tailored to the encountered stimulus. Lymphatic vessels facilitate this process by continuously collecting immunological information (ie, antigens, immune cells, and soluble mediators) about the current state of peripheral tissues, and transporting these via the lymph across the lymphatic system. Lymph nodes (LNs), which are critical meeting points for innate and adaptive immune cells, are strategically located along the lymphatic network to intercept this information. Within LNs, immune cells are spatially organized, allowing them to efficiently respond to information delivered by the lymph, and to either promote immune homeostasis or mount protective immune responses. These responses involve the activation and functional cooperation of multiple distinct cell types and are tailored to the specific inflammatory conditions. The natural patterns of lymph flow can also generate spatial gradients of antigens and agonists within draining LNs, which can in turn further regulate innate cell function and localization, as well as the downstream generation of adaptive immunity. In this review, we explore how information transmitted by the lymph shapes the spatiotemporal organization of innate and adaptive immune responses in LNs, with particular focus on steady state and Type-I vs. Type-II inflammation.
Collapse
Affiliation(s)
| | | | - Michael Y Gerner
- Corresponding author: Michael Gerner, , Address: 750 Republican Street Seattle, WA 98109, Phone: 206-685-3610
| |
Collapse
|
52
|
Enterotoxigenic Escherichia coli enterotoxins regulate epithelial to immune relay of IL-33 and IL-1Ra cytokines. Infect Immun 2022; 90:e0063721. [PMID: 35191758 DOI: 10.1128/iai.00637-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) remain a major cause of diarrheal mortality and morbidity in children in low-resource settings. Few studies have explored the consequences of simultaneous intoxication with heat-stable (ST) and heat-labile (LT) enterotoxins despite the increased prevalence of wild ETEC isolates expressing both toxins. We therefore used a combination of tissue culture and murine models to explore the impact of simultaneous ST+LT intoxication of epithelial and myeloid cell responses. We report that LT induces sustained IL-33 and IL-1Ra responses in T84 intestinal epithelial cells via cAMP-production and protein kinase A activation. We demonstrate that combined ST+LT intoxication hastens epithelial transcriptional responses induced more slowly by LT alone. ST- and LT-mediated luminal fluid accumulation in vivo correlates with significant increases in IL-33 and IL-1Ra in small intestinal mucosal scrapings. Additionally, IL-33 receptor (IL-33R)-deficient mice are less susceptible to ST-mediated secretion. In the immune compartment, IL-33 is sensed by myeloid cells, and LT suppresses IL-33-induced TNFα secretion from macrophages but amplifies IL-33-mediated induction of IL-6 from bone marrow-derived dendritic cells. In conclusion, our studies suggest that enterotoxin-induced IL-33 and IL-1Ra modulate intestinal inflammation and IL-1 receptor signaling in the intestinal mucosa in response to ETEC enterotoxins.
Collapse
|
53
|
Alam A, Levanduski E, Denz P, Villavicencio HS, Bhatta M, Alhorebi L, Zhang Y, Gomez EC, Morreale B, Senchanthisai S, Li J, Turowski SG, Sexton S, Sait SJ, Singh PK, Wang J, Maitra A, Kalinski P, DePinho RA, Wang H, Liao W, Abrams SI, Segal BH, Dey P. Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell 2022; 40:153-167.e11. [PMID: 35120601 PMCID: PMC8847236 DOI: 10.1016/j.ccell.2022.01.003] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/30/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
TH2 cells and innate lymphoid cells 2 (ILC2) can stimulate tumor growth by secreting pro-tumorigenic cytokines such as interleukin-4 (IL-4), IL-5, and IL-13. However, the mechanisms by which type 2 immune cells traffic to the tumor microenvironment are unknown. Here, we show that oncogenic KrasG12D increases IL-33 expression in pancreatic ductal adenocarcinoma (PDAC) cells, which recruits and activates TH2 and ILC2 cells. Correspondingly, cancer-cell-specific deletion of IL-33 reduces TH2 and ILC2 recruitment and promotes tumor regression. Unexpectedly, IL-33 secretion is dependent on the intratumoral fungal mycobiome. Genetic deletion of IL-33 or anti-fungal treatment decreases TH2 and ILC2 infiltration and increases survival. Consistently, high IL-33 expression is observed in approximately 20% of human PDAC, and expression is mainly restricted to cancer cells. These data expand our knowledge of the mechanisms driving PDAC tumor progression and identify therapeutically targetable pathways involving intratumoral mycobiome-driven secretion of IL-33.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Eric Levanduski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Parker Denz
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Helena Solleiro Villavicencio
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Maulasri Bhatta
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Lamees Alhorebi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Yali Zhang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Brian Morreale
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Sharon Senchanthisai
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Jun Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven G Turowski
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sandra Sexton
- Department of Animal Resources, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sheila Jani Sait
- Department of Cytogenetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Prashant K Singh
- Genomics Shared Resource, Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA; Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wenting Liao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA
| | - Brahm H Segal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Prasenjit Dey
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Sts. CGP/BLSC-L5307, Buffalo, NY 14263, USA.
| |
Collapse
|
54
|
Nguyen NZN, Tran VG, Baek J, Kim Y, Youn EH, Na SW, Park SJ, Seo SK, Kwon B. IL-33 Coordinates Innate Defense to Systemic Candida albicans Infection by Regulating IL-23 and IL-10 in an Opposite Way. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:660-671. [PMID: 35022276 DOI: 10.4049/jimmunol.2100495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022]
Abstract
Invasive candidiasis has high mortality rates in immunocompromised patients, causing serious health problems. In mouse models, innate immunity protects the host by rapidly mobilizing a variety of resistance and tolerance mechanisms to systemic Candida albicans infection. We have previously demonstrated that exogenous IL-33 regulates multiple steps of innate immunity involving resistance and tolerance processes. In this study, we systematically analyzed the in vivo functions of endogenous IL-33 using Il33 -/- mice and in vitro immune cell culture. Tubular epithelial cells mainly secreted IL-33 in response to systemic C. albicans infection. Il33 -/- mice showed increased mortality and morbidity, which were due to impaired fungal clearance. IL-33 initiated an innate defense mechanism by costimulating dendritic cells to produce IL-23 after systemic C. albicans infection, which in turn promoted the phagocytosis of neutrophils through secretion of GM-CSF by NK cells. The susceptibility of Il33 -/- mice was also associated with increased levels of IL-10, and neutralization of IL-10 resulted in enhanced fungal clearance in Il33 -/- mice. However, depletion of IL-10 overrode the effect of IL-33 on fungal clearance. In Il10 -/- mouse kidneys, MHC class II+F4/80+ macrophages were massively differentiated after C. albicans infection, and these cells were superior to MHC class II-F4/80+ macrophages that were preferentially differentiated in wild-type mouse kidneys in killing of extracellular hyphal C. albicans Taken together, our results identify IL-33 as critical early regulator controlling a serial downstream signaling events of innate defense to C. albicans infection.
Collapse
Affiliation(s)
- Nu Z N Nguyen
- BK21 Integrated Immunomodulation Education and Research Team, School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Vuvi G Tran
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA
| | - Jiyeon Baek
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Younghee Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Eun H Youn
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Seung W Na
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea.,Division of Pulmonology, Department of Internal Medicine, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea; and
| | - Sang J Park
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea.,Department of Surgery, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| | - Su-Kil Seo
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan, Republic of Korea;
| | - Byungsuk Kwon
- BK21 Integrated Immunomodulation Education and Research Team, School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea; .,Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan, Republic of Korea
| |
Collapse
|
55
|
Flayer CH, Sokol CL. Sensory neurons control the functions of dendritic cells to guide allergic immunity. Curr Opin Immunol 2022; 74:85-91. [PMID: 34808584 PMCID: PMC8901476 DOI: 10.1016/j.coi.2021.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 02/03/2023]
Abstract
Dendritic cells of the innate immune system and sensory neurons of the peripheral nervous system are embedded in barrier tissues and gather information about an organisms' environment. While the mechanisms by which dendritic cells recognize and initiate adaptive immune responses to pathogens is well defined, how they sense allergens is poorly understood. Indeed, allergens induce dendritic cell maturation and migration in vivo, but not in vitro. How are adaptive immune responses to allergens initiated if dendritic cells do not directly sense allergens? Sensory neurons release neuropeptides within minutes of allergen exposure. Recent evidence demonstrated that while neuropeptides modify dendritic cell function during pathogen responses, they are required for dendritic cell function during allergic responses. These emerging studies suggest that sensory neurons do not just pass information along to the central nervous system, but also to dendritic cells, particularly during the initiation of adaptive immunity to allergens.
Collapse
Affiliation(s)
| | - Caroline L Sokol
- Corresponding author: , 149 13th St Room 8103, Charlestown, MA 02129
| |
Collapse
|
56
|
Virtanen T. Inhalant Mammal-Derived Lipocalin Allergens and the Innate Immunity. FRONTIERS IN ALLERGY 2022; 2:824736. [PMID: 35387007 PMCID: PMC8974866 DOI: 10.3389/falgy.2021.824736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 12/03/2022] Open
Abstract
A major part of important mammalian respiratory allergens belongs to the lipocalin family of proteins. By this time, 19 respiratory mammalian lipocalin allergens have been registered in the WHO/IUIS Allergen Nomenclature Database. Originally, lipocalins, small extracellular proteins (molecular mass ca. 20 kDa), were characterized as transport proteins but they are currently known to exert a variety of biological functions. The three-dimensional structure of lipocalins is well-preserved, and lipocalin allergens can exhibit high amino acid identities, in several cases more than 50%. Lipocalins contain an internal ligand-binding site where they can harbor small principally hydrophobic molecules. Another characteristic feature is their capacity to bind to specific cell-surface receptors. In all, the physicochemical properties of lipocalin allergens do not offer any straightforward explanations for their allergenicity. Allergic sensitization begins at epithelial barriers where diverse insults through pattern recognition receptors awaken innate immunity. This front-line response is manifested by epithelial barrier-associated cytokines which together with other components of immunity can initiate the sensitization process. In the following, the crucial factor in allergic sensitization is interleukin (IL)-4 which is needed for stabilizing and promoting the type 2 immune response. The source for IL-4 has been searched widely. Candidates for it may be non-professional antigen-presenting cells, such as basophils or mast cells, as well as CD4+ T cells. The synthesis of IL-4 by CD4+ T cells requires T cell receptor engagement, i.e., the recognition of allergen peptides, which also provides the specificity for sensitization. Lipocalin and innate immunity-associated cell-surface receptors are implicated in facilitating the access of lipocalin allergens into the immune system. However, the significance of this for allergic sensitization is unclear, as the recognition by these receptors has been found to produce conflicting results. As to potential adjuvants associated with mammalian lipocalin allergens, the hydrophobic ligands transported by lipocalins have not been reported to enhance sensitization while it is justified to suppose that lipopolysaccharide plays a role in it. Taken together, type 2 immunity to lipocalin allergens appears to be a harmful immune response resulting from a combination of signals involving both the innate and adaptive immunities.
Collapse
Affiliation(s)
- Tuomas Virtanen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
57
|
Temporal Quantitative Phosphoproteomics Profiling of Interleukin-33 Signaling Network Reveals Unique Modulators of Monocyte Activation. Cells 2022; 11:cells11010138. [PMID: 35011700 PMCID: PMC8749991 DOI: 10.3390/cells11010138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 superfamily cytokines, is an endogenous danger signal and a nuclear-associated cytokine. It is one of the essential mediators of both innate and adaptive immune responses. Aberrant IL-33 signaling has been demonstrated to play a defensive role against various infectious and inflammatory diseases. Although the signaling responses mediated by IL-33 have been previously reported, the temporal signaling dynamics are yet to be explored. To this end, we applied quantitative temporal phosphoproteomics analysis to elucidate pathways and proteins induced by IL-33 in THP-1 monocytes. Employing a TMT labeling-based quantitation and titanium dioxide (TiO2)-based phosphopeptide enrichment strategy followed by mass spectrometry analysis, we identified and quantified 9448 unique phosphopeptides corresponding to 3392 proteins that showed differential regulation. Of these, 171 protein kinases, 60 phosphatases and 178 transcription factors were regulated at different phases of IL-33 signaling. In addition to the confirmed activation of canonical signaling modules including MAPK, NFκB, PI3K/AKT modules, pathway analysis of the time-dependent phosphorylation dynamics revealed enrichment of several cellular processes, including leukocyte adhesion, response to reactive oxygen species, cell cycle checkpoints, DNA damage and repair pathways. The detailed quantitative phosphoproteomic map of IL-33 signaling will serve as a potentially useful resource to study its function in the context of inflammatory and pathological conditions.
Collapse
|
58
|
Ghezzi M, Pozzi E, Abbattista L, Lonoce L, Zuccotti GV, D’Auria E. Barrier Impairment and Type 2 Inflammation in Allergic Diseases: The Pediatric Perspective. CHILDREN (BASEL, SWITZERLAND) 2021; 8:1165. [PMID: 34943362 PMCID: PMC8700706 DOI: 10.3390/children8121165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 01/02/2023]
Abstract
Allergic diseases represent a global burden. Although the patho-physiological mechanisms are still poorly understood, epithelial barrier dysfunction and Th2 inflammatory response play a pivotal role. Barrier dysfunction, characterized by a loss of differentiation, reduced junctional integrity, and altered innate defence, underpins the pathogenesis of allergic diseases. Epithelial barrier impairment may be a potential therapeutic target for new treatment strategies Up now, monoclonal antibodies and new molecules targeting specific pathways of the immune response have been developed, and others are under investigation, both for adult and paediatric populations, which are affected by atopic dermatitis (AD), asthma, allergic rhinitis (AR), chronic rhinosinusitis with nasal polyps (CRSwNP), or eosinophilic esophagitis (EoE). In children affected by severe asthma biologics targeting IgE, IL-5 and against IL-4 and IL-13 receptors are already available, and they have also been applied in CRSwNP. In severe AD Dupilumab, a biologic which inhibits both IL-4 and IL-13, the most important cytokines involved in inflammation response, has been approved for treatment of patients over 12 years. While a biological approach has already shown great efficacy on the treatment of severe atopic conditions, early intervention to restore epithelial barrier integrity, and function may prevent the inflammatory response and the development of the atopic march.
Collapse
Affiliation(s)
- Michele Ghezzi
- Allergology and Pneumology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy;
| | - Elena Pozzi
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Luisa Abbattista
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Luisa Lonoce
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
- Department of Biomedical and Clinical Science “L. Sacco”, University of Milan, 20157 Milan, Italy
| | - Enza D’Auria
- Allergology and Pneumology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy;
| |
Collapse
|
59
|
Bridgewood C, Newton D, Bragazzi N, Wittmann M, McGonagle D. Unexpected connections of the IL-23/IL-17 and IL-4/IL-13 cytokine axes in inflammatory arthritis and enthesitis. Semin Immunol 2021; 58:101520. [PMID: 34799224 DOI: 10.1016/j.smim.2021.101520] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
The IL-23/IL-17 cytokine axis is related to spondyloarthropathy (SpA) pattern diseases that target the skin, eye, gut and joints. These share overlapping target tissues with Th2 type or allergic diseases, including the skin, eye and gut but SpA diseases exhibit distinct microanatomical topography, molecular characteristics, and clinical features including uveitis, psoriasis, apical pulmonary involvement, lower gastrointestinal involvement with colitis, and related arthritides including psoriatic arthritis and ankylosing spondylitis. Inflammatory arthritis is conspicuously absent from the Th2 diseases which are characterised IL-4/IL-13 dependent pathway activation including allergic rhino-conjunctivitis, atopic eczema, allergic asthma and food allergies. This traditional understanding of non-overlap of musculoskeletal territory between that atopic diseases and the IL-17 -mediated SpA diseases is undergoing a critical reappraisal with the recent demonstration of IL-4/IL-13 blockade, may be associated with the development of SpA pattern arthritis, psoriasiform skin disease and occasional anterior uveitis. Given the known plasticity within Th paradigm pathways, these findings suggest dynamic Th2 cytokine and Th17 cytokine counter regulation in vivo in humans. Unexpected, this is the case in peripheral enthesis and when the IL-4/13 immunological brake on IL-23/17 cytokines is removed, a SpA phenotype may emerge. We discuss hitherto unexpected observations in SpA, showing counter regulation between the Th17 and Th2 pathways at sites including the entheses that collectively indicate that the emergent reverse translational therapeutic data is more than coincidental and offers new insights into the "Th paradigms" in atopy and SpA.
Collapse
Affiliation(s)
- Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK.
| | - Darren Newton
- Division of Haematology and Immunology, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK
| | - Nicola Bragazzi
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK; National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK; National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, UK
| |
Collapse
|
60
|
Zhang M, Duffen JL, Nocka KH, Kasaian MT. IL-13 Controls IL-33 Activity through Modulation of ST2. THE JOURNAL OF IMMUNOLOGY 2021; 207:3070-3080. [PMID: 34789557 DOI: 10.4049/jimmunol.2100655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
IL-33 is a multifunctional cytokine that mediates local inflammation upon tissue damage. IL-33 is known to act on multiple cell types including group 2 innate lymphoid cells (ILC2s), Th2 cells, and mast cells to drive production of Th2 cytokines including IL-5 and IL-13. IL-33 signaling activity through transmembrane ST2L can be inhibited by soluble ST2 (sST2), which acts as a decoy receptor. Previous findings suggested that modulation of IL-13 levels in mice lacking decoy IL-13Rα2, or mice lacking IL-13, impacted responsiveness to IL-33. In this study, we used Il13 -/- mice to investigate whether IL-13 regulates IL-33 activity by modulating the transmembrane and soluble forms of ST2. In Il13 -/- mice, the effects of IL-33 administration were exacerbated relative to wild type (WT). Il13 -/- mice administered IL-33 i.p. had heightened splenomegaly, more immune cells in the peritoneum including an expanded ST2L+ ILC2 population, increased eosinophilia in the spleen and peritoneum, and reduced sST2 in the circulation and peritoneum. In the spleen, lung, and liver of mice given IL-33, gene expression of both isoforms of ST2 was increased in Il13 -/- mice relative to WT. We confirmed fibroblasts to be an IL-13-responsive cell type that can regulate IL-33 activity through production of sST2. This study elucidates the important regulatory activity that IL-13 exerts on IL-33 through induction of IL-33 decoy receptor sST2 and through modulation of ST2L+ ILC2s.
Collapse
Affiliation(s)
- Melvin Zhang
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Jennifer L Duffen
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Karl H Nocka
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Marion T Kasaian
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| |
Collapse
|
61
|
Agache I, Eguiluz‐Gracia I, Cojanu C, Laculiceanu A, Giacco S, Zemelka‐Wiacek M, Kosowska A, Akdis CA, Jutel M. Advances and highlights in asthma in 2021. Allergy 2021; 76:3390-3407. [PMID: 34392546 DOI: 10.1111/all.15054] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022]
Abstract
Last year brought a significant advance in asthma management, unyielding to the pressure of the pandemics. Novel key findings in asthma pathogenesis focus on the resident cell compartment, epigenetics and the innate immune system. The precision immunology unbiased approach was supplemented with novel tools and greatly facilitated by the use of artificial intelligence. Several randomised clinical trials and good quality real-world evidence shed new light on asthma treatment and supported the revision of several asthma guidelines (GINA, Expert Panel Report 3, ERS/ATS guidelines on severe asthma) and the conception of new ones (EAACI Guidelines for the use of biologicals in severe asthma). Integrating asthma management within the broader context of Planetary Health has been put forward. In this review, recently published articles and clinical trials are summarised and discussed with the goal to provide clinicians and researchers with a concise update on asthma research from a translational perspective.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Ibon Eguiluz‐Gracia
- Allergy Unit IBIMA‐Regional University Hospital of MalagaUMA, RETICS ARADyALBIONAND Malaga Spain
| | | | | | - Stefano Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | | | - Anna Kosowska
- Department of Clinical Immunology Wroclaw Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wroclaw Poland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Marek Jutel
- Department of Clinical Immunology Wroclaw Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wroclaw Poland
| |
Collapse
|
62
|
Ramsey N, Berin MC. Pathogenesis of IgE-mediated food allergy and implications for future immunotherapeutics. Pediatr Allergy Immunol 2021; 32:1416-1425. [PMID: 33715245 PMCID: PMC9096874 DOI: 10.1111/pai.13501] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/29/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
Our understanding of the immune basis of food allergy has grown rapidly in parallel with the development of new immune-targeted interventions for the treatment of food allergy. Local tissue factors, including the composition of skin and gastrointestinal microbiota and production of Th2-inducing cytokines (TSLP, IL-33, and IL-25) from barrier sites, have been shown not only to contribute to the development of food allergy, but also to act as effective targets for treatment in mice. Ongoing clinical trials are testing the targeting of these factors in human disease. There is a growing understanding of the contribution of IL-13 to the induction of high-affinity IgE and the need for continual T-cell help in the maintenance of long-lived IgE. This provides a strong rationale to test biologics targeting both IL-4 and IL-13 in the treatment of established food allergy. Various forms of allergen immunotherapy for food allergy have clearly shown that low specific IgE and elevated specific IgG4 are predictive of sustained treatment effect. Treatments that mimic that immune response, for example, lowering IgE, with monoclonal antibodies such as omalizumab, or administering allergen-specific IgG, are in various stages of investigation. As we gain more opportunities to use immune-modifying treatments for the treatment of food allergy, studies of the immune and clinical response to those interventions will continue to rapidly advance our understanding of the immune basis of food allergy and tolerance.
Collapse
Affiliation(s)
- Nicole Ramsey
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Cecilia Berin
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
63
|
Li C, Du X, Huang Q, Yang Y, Wang J, Qin X, Wang W, Liu Z, Yuan H, Liu J, Lv Z, Li Y, Chen Y, Cui Y, Corrigan CJ, Huang K, Wang W, Ying S. Repeated exposure to inactivated Streptococcus pneumoniae induces asthma-like pathological changes in mice in the presence of IL-33. Cell Immunol 2021; 369:104438. [PMID: 34530343 DOI: 10.1016/j.cellimm.2021.104438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/19/2021] [Accepted: 08/17/2021] [Indexed: 11/27/2022]
Abstract
While environmental aeroallergens and epithelial alarmins such as IL-33 are firmly implicated in asthma, the possible role of Streptococcus pneumoniae (S. pneumoniae) antigens is less clear. To explore this, wild-type BALB/c mice were repeatedly challenged per-nasally with IL-33 and inactivated S. pneumoniae, either agent alone or diluent control. Some animals were rested then later re-challenged with inactivated S. pneumoniae alone. Serum concentrations of S. pneumoniae lysates-specific IgE were measured in patients with asthma and control subjects. Interestingly, in the presence of IL-33, repeated exposure to inactivated S. pneumoniae induced asthma-like pathological changes accompanied by a systemic adaptive immune response. Subsequent re-exposure of the sensitized animals to inactivated S. pneumoniae alone was able to induce such changes. The concentration of S. pneumoniae lysates-specific IgE was significantly elevated in the asthma patients. These data suggest that antigens derived from infectious microorganisms may participate in generating the mucosal inflammation which characterizes asthma.
Collapse
Affiliation(s)
- Chenduo Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China; College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaonan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qiong Huang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yiran Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Xiaofeng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wenjun Wang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Institute of Respiratory Medicine, Beijing, China
| | - Zihan Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Otorhinolaryngology, Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Ministry of Education, Beijing Key Laboratory of Nasal Diseases, Beijing, China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Division of Asthma, Allergy & Lung Biology, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Kewu Huang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Institute of Respiratory Medicine, Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
64
|
Sasaki E, Asanuma H, Momose H, Furuhata K, Mizukami T, Hamaguchi I. Nasal alum-adjuvanted vaccine promotes IL-33 release from alveolar epithelial cells that elicits IgA production via type 2 immune responses. PLoS Pathog 2021; 17:e1009890. [PMID: 34460865 PMCID: PMC8432758 DOI: 10.1371/journal.ppat.1009890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/10/2021] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
Aluminum hydroxide salts (alum) have been added to inactivated vaccines as safe and effective adjuvants to increase the effectiveness of vaccination. However, the exact cell types and immunological factors that initiate mucosal immune responses to alum adjuvants are unclear. In this study, the mechanism of action of alum adjuvant in nasal vaccination was investigated. Alum has been shown to act as a powerful and unique adjuvant when added to a nasal influenza split vaccine in mice. Alum is cytotoxic in the alveoli and stimulates the release of damage-associated molecular patterns, such as dsDNA, interleukin (IL)-1α, and IL-33. We found that Ag-specific IgA antibody (Ab) production was markedly reduced in IL-33-deficient mice. However, no decrease was observed in Ag-specific IgA Ab production with DNase I treatment, and no decrease was observed in IL-1α/β or IL-6 production in IL-33-deficient mice. From the experimental results of primary cultured cells and immunofluorescence staining, although IL-1α was secreted by alveolar macrophage necroptosis, IL-33 release was observed in alveolar epithelial cell necroptosis but not in alveolar macrophages. Alum- or IL-33-dependent Ag uptake enhancement and elevation of OX40L expression were not observed. By stimulating the release of IL-33, alum induced Th2 immunity via IL-5 and IL-13 production in group 2 innate lymphoid cells (ILC2s) and increased MHC class II expression in antigen-presenting cells (APCs) in the lung. Our results suggest that IL-33 secretion by epithelial cell necroptosis initiates APC- and ILC2-mediated T cell activation, which is important for the enhancement of Ag-specific IgA Ab production by alum. Aluminum salts have been used as adjuvants in many vaccines. Aluminum salts induce Th2 immunity and vaccine antigen-specific antibody production aluminum salts elicit adjuvant action via cytokine production. Currently, the mechanisms underlying aluminum salt function in nasal vaccination are unknown, and elucidation of the mechanism is important for the development of particulate adjuvants. This study focused on the cytokines released from dead cells as induced by aluminum salt. This study found that aluminum adjuvant caused release of the cytokine interleukin (IL)-33 from alveolar epithelial cells by inducing necrosis. IL-33 is also crucial for antigen-specific IgA antibody production by nasal vaccination. Aluminum adjuvant also induces alveolar macrophage necrosis, which is not accompanied by IL-33 release. Aluminum salt-induced IL-33 acts as an activator for group 2 innate lymphoid cells and antigen-presenting cells in the lung. This means that by developing an adjuvant that targets the release of IL-33, it may be possible to develop a highly effective nasal vaccine. IL-33 significantly contributes to the efficacy of nasal vaccines and provides new insights into the mechanisms underlying aluminum adjuvants, showing that lung parenchymal tissue, rather than macrophages and lymphocytes, is the source of IL-33.
Collapse
Affiliation(s)
- Eita Sasaki
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
- * E-mail: (ES); (TM)
| | - Hideki Asanuma
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
| | - Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
| | - Keiko Furuhata
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
| | - Takuo Mizukami
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
- * E-mail: (ES); (TM)
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi–Murayama, Tokyo, Japan
| |
Collapse
|
65
|
Okragly AJ, Corwin KB, Elia M, He D, Schroeder O, Zhang Q, Shiyanova T, Bright S, Dicker SB, Chlewicki L, Truhlar SME, Davies J, Patel CN, Benschop RJ. Generation and Characterization of Torudokimab (LY3375880): A Monoclonal Antibody That Neutralizes Interleukin-33. J Inflamm Res 2021; 14:3823-3835. [PMID: 34408465 PMCID: PMC8364917 DOI: 10.2147/jir.s320287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Background Interleukin-33 (IL-33) is an alarmin that is released following cellular damage, mechanical injury, or necrosis. It is a member of the IL-1 family and binds to a heterodimer receptor consisting of ST2 and IL-1RAP to induce the production of a wide range of cellular mediators, including the type 2 cytokines IL-4, IL-5 and IL-13. This relationship has led to the hypothesis that the IL-33/ST2 pathway is a driver of allergic disease and inhibition of the IL-33 and ST2 association could have therapeutic benefit. Methods In this paper, we describe the selection of a phage antibody through the ability to bind human IL-33 and block IL-33/ST2 interaction. This hit antibody was then affinity matured by site-directed mutagenesis of the antibody complementarity-determining regions (CDRs). Further characterization of a fully human monoclonal antibody (mAb), torudokimab (LY3375880) included demonstration of human IL-33 neutralization activity in vitro with an NFκB reporter assay and IL-33 induced mast cell cytokine secretion assay, followed by an in vivo IL-33-induced pharmacodynamic inhibition assay in mice that used IL-5 production as the endpoint. Results Torudokimab is highly specific to IL-33 and does not bind any of the other IL-1 family members. Furthermore, torudokimab binds human and cynomolgus monkey IL-33 with higher affinity than the binding affinity of IL-33 to ST2, but does not bind mouse, rat, or rabbit IL-33. Torudokimab’s half-life in cynomolgous monkey projects monthly dosing in the clinic. Conclusion Due to torudokimab’s high affinity, its ability to completely neutralize IL-33 activity in vitro and in vivo, and the observed cynomolgus monkey pharmacokinetic properties, this molecule was selected for clinical development.
Collapse
Affiliation(s)
- Angela J Okragly
- Immunology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Marikka Elia
- BioTechnology Discovery Research Eli Lilly and Company, San Diego, CA, USA
| | - Dongmei He
- BioTechnology Discovery Research Eli Lilly and Company, San Diego, CA, USA
| | - Oliver Schroeder
- BioTechnology Discovery Research Eli Lilly and Company, San Diego, CA, USA
| | - Qing Zhang
- BioTechnology Discovery Research Eli Lilly and Company, San Diego, CA, USA
| | - Tatiyana Shiyanova
- BioTechnology Discovery Research, Eli Lilly and Company, Indianapolis, IN, USA
| | - Stuart Bright
- Immunology Research, Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | - Julian Davies
- BioTechnology Discovery Research Eli Lilly and Company, San Diego, CA, USA
| | - Chetan N Patel
- BioTechnology Discovery Research, Eli Lilly and Company, Indianapolis, IN, USA
| | | |
Collapse
|
66
|
Nakajima S, Tie D, Nomura T, Kabashima K. Novel pathogenesis of atopic dermatitis from the view of cytokines in mice and humans. Cytokine 2021; 148:155664. [PMID: 34388479 DOI: 10.1016/j.cyto.2021.155664] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/27/2022]
Abstract
Type 2 immunity and inflammation underlie allergic skin disorders, such as atopic dermatitis (AD). In type 2 inflammation, IL-4, IL-13, and IL-5, which are signature type 2 cytokines, are mainly produced by type 2 helper T (Th2) cells and form the characteristic features of AD. Epithelial cell-derived cytokines such as IL-25, IL-33, and TSLP initiate type 2 inflammation by modulating various cells, including group 2 innate lymphoid cells. Moreover, IL-31, a newly identified type 2 cytokine produced mainly by Th2 cells, induces pruritus by acting on sensory neurons in the skin. Based on both basic and clinical findings, several biologics targeting Th2 cytokines have been developed and exhibited significant efficacy as therapeutic reagents for AD. We have summarized the roles of each cytokine (IL-4, 5, 13, 25, 31, and 33, and TSLP) in the development of type 2 inflammation, especially AD, from the view of basic studies in mice and clinical trials/observation in humans.
Collapse
Affiliation(s)
- Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan; Department of Drug Discovery for Inflammatory Skin Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Duerna Tie
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan; Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
67
|
The Janus Face of IL-33 Signaling in Tumor Development and Immune Escape. Cancers (Basel) 2021; 13:cancers13133281. [PMID: 34209038 PMCID: PMC8268428 DOI: 10.3390/cancers13133281] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/06/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Interleukin-33 (IL-33) is often released from damaged cells, acting as a danger signal. IL-33 exerts its function by interacting with its receptor suppression of tumorigenicity 2 (ST2) that is constitutively expressed on most immune cells. Therefore, IL-33/ST2 signaling can modulate immune responses to participate actively in a variety of pathological conditions, such as cancer. Like a two-faced Janus, which faces opposite directions, IL-33/ST2 signaling may play contradictory roles on its impact on cancer progression through both immune and nonimmune cellular components. Accumulating evidence demonstrates both pro- and anti-tumorigenic properties of IL-33, depending on the complex nature of different tumor immune microenvironments. We summarize and discuss the most recent studies on the contradictory effects of IL-33 on cancer progression and treatment, with a goal to better understanding the various ways for IL-33 as a therapeutic target. Abstract Interleukin-33 (IL-33), a member of the IL-1 cytokine family, plays a critical role in maintaining tissue homeostasis as well as pathological conditions, such as allergy, infectious disease, and cancer, by promoting type 1 and 2 immune responses. Through its specific receptor ST2, IL-33 exerts multifaceted functions through the activation of diverse intracellular signaling pathways. ST2 is expressed in different types of immune cells, including Th2 cells, Th1 cells, CD8+ T cells, regulatory T cells (Treg), cytotoxic NK cells, group 2 innate lymphoid cells (ILC2s), and myeloid cells. During cancer initiation and progression, the aberrant regulation of the IL-33/ST2 axis in the tumor microenvironment (TME) extrinsically and intrinsically mediates immune editing via modulation of both innate and adaptive immune cell components. The summarized results in this review suggest that IL-33 exerts dual-functioning, pro- as well as anti-tumorigenic effects depending on the tumor type, expression levels, cellular context, and cytokine milieu. A better understanding of the distinct roles of IL-33 in epithelial, stromal, and immune cell compartments will benefit the development of a targeting strategy for this IL-33/ST2 axis for cancer immunotherapy.
Collapse
|
68
|
Wang W, An G, Li Y, Wang J, Lv Z, Chen Y, Corrigan CJ, Wang W, Huang K, Ying S. IL-33 amplifies airways inflammation in a murine surrogate of asthma putatively via activation of dendritic cells. Cell Immunol 2021; 366:104395. [PMID: 34198027 DOI: 10.1016/j.cellimm.2021.104395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 05/19/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022]
Abstract
Although contributions of IL-33 to pulmonary diseases, including asthma, have been well documented, the complexity of such regulation warrants additional exploration. To better understand the involvement of IL-33, we used a murine asthma surrogate based on sensitisation and challenge with dust mite extract in the presence/absence of IL-33. Murine models were established with Dermatophagoides farinae (Der f) to establish (1) the effect of co-administered rmIL-33; (2) the effect of prior glucocorticoid intervention; (3) the effect of IL-33 on challenge with sub-threshold dosage Der f. The effects of rmIL-33 on bone marrow-derived dendritic cells were explored in vitro. Mice challenged with Der f combined with IL-33 compared with diluent control evinced significantly more airways inflammation and local cytokine production which was less sensitive to inhibition by dexamethasone. IL-33 also induced airways hyperresponsiveness, eosinophilic inflammation and cytokine production in lung tissues of animals exposed to sub-threshold dosage of Der f. In vitro, IL-33-stimulated DCs showed a significantly elevated capacity to stimulate CD4+ T cell proliferation and cytokine production and were also significantly more resistant to dexamethasone-induced apoptosis. Our data suggest that IL-33 reduces the threshold for allergen-induced inflammation of the airways in acorticosteroid-resistant fashion possibly in part through acting on DCs, a phenomenon which may be relevant to the development of severe, corticosteroid-resistant airways obstruction in human asthmatic patients.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Respiratory and CriticalCare Medicine, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Institute of Respiratory Medicine, Beijing, China
| | - Gao An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Faculty of Life Sciences & Medicine, School of Immunology & Microbial Sciences, Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kewu Huang
- Department of Respiratory and CriticalCare Medicine, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Institute of Respiratory Medicine, Beijing, China.
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
69
|
Zaini A, Fulford TS, Grumont RJ, Runting J, Rodrigues G, Ng J, Gerondakis S, Zaph C, Scheer S. c-Rel Is Required for IL-33-Dependent Activation of ILC2s. Front Immunol 2021; 12:667922. [PMID: 34194431 PMCID: PMC8236704 DOI: 10.3389/fimmu.2021.667922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are emerging as important cellular regulators of homeostatic and disease-associated immune processes. The cytokine interleukin-33 (IL-33) promotes ILC2-dependent inflammation and immunity, with IL-33 having been shown to activate NF-κB in a wide variety of cell types. However, it is currently unclear which NF-κB members play an important role in IL-33-dependent ILC2 biology. Here, we identify the NF-κB family member c-Rel as a critical component of the IL-33-dependent activation of ILC2s. Although c-Rel is dispensable for ILC2 development, it is critical for ILC2 function in the lung, with c-Rel-deficient (c-Rel-/- ) mice present a significantly reduced response to papain- and IL-33-induced lung inflammation. We also show that the absence of c-Rel reduces the IL-33-dependent expansion of ILC2 precursors and lower levels of IL-5 and IL-13 cytokine production by mature ILC2s in the lung. Together, these results identify the IL-33-c-Rel axis as a central control point of ILC2 activation and function.
Collapse
Affiliation(s)
- Aidil Zaini
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Thomas S. Fulford
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Raelene J. Grumont
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jessica Runting
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Grace Rodrigues
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Judy Ng
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Steve Gerondakis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Colby Zaph
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Sebastian Scheer
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
70
|
Iwaszko M, Wielińska J, Świerkot J, Kolossa K, Sokolik R, Bugaj B, Chaszczewska-Markowska M, Jeka S, Bogunia-Kubik K. IL-33 Gene Polymorphisms as Potential Biomarkers of Disease Susceptibility and Response to TNF Inhibitors in Rheumatoid Arthritis, Ankylosing Spondylitis, and Psoriatic Arthritis Patients. Front Immunol 2021; 12:631603. [PMID: 34177886 PMCID: PMC8226138 DOI: 10.3389/fimmu.2021.631603] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
Objective Rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) belong to inflammatory rheumatic diseases, the group of conditions of unknown etiology. However, a strong genetic component in their pathogenesis has been well established. A dysregulation of cytokine networks plays an important role in the development of inflammatory arthritis. Interleukin 33 (IL-33) is a recently identified member of the IL-1 family. To date, the significance of IL-33 in inflammatory arthritis has been poorly studied. This research aimed to investigate the potential of IL-33 gene polymorphisms to serve as biomarkers for disease susceptibility and TNF inhibitor response in RA, AS, and PsA patients. Materials and Methods In total, 735 patients diagnosed with RA, AS, and PsA and 229 healthy individuals were enrolled in the study. Genotyping for three single nucleotide polymorphisms (SNPs) within the IL-33 gene, namely, rs16924159 (A/G), rs10975519 (T/C), and rs7044343 (C/T), was performed using polymerase chain reaction amplification employing LightSNiP assays. Results In the present study, the IL-33 rs10975519 CC genotype was associated with a decreased risk of developing RA in females, while the IL-33 rs16924159 polymorphism was associated with the efficacy of anti-TNF therapy and clinical parameters for RA and AS patients. The IL-33 rs16924159 AA genotype correlated with higher disease activity and worse clinical outcomes in RA patients treated with TNF inhibitors, and AS patients carrying the IL-33 rs16924159 AA genotype had higher disease activity and a worse response to anti-TNF therapy. That indicates a deleterious role of the IL-33 rs16924159 AA genotype in the context of RA, as well as AS. Conclusions The obtained results suggest that IL-33 gene polymorphisms might be potential candidate biomarkers of disease susceptibility and anti-TNF treatment response in patients with inflammatory rheumatic diseases.
Collapse
Affiliation(s)
- Milena Iwaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Joanna Wielińska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Katarzyna Kolossa
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
| | - Renata Sokolik
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Bartosz Bugaj
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Monika Chaszczewska-Markowska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sławomir Jeka
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland.,Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
71
|
Miyata J, Yokokura Y, Moro K, Arai H, Fukunaga K, Arita M. 12/15-Lipoxygenase Regulates IL-33-Induced Eosinophilic Airway Inflammation in Mice. Front Immunol 2021; 12:687192. [PMID: 34093589 PMCID: PMC8170304 DOI: 10.3389/fimmu.2021.687192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Dysregulated fatty acid metabolism is clinically associated with eosinophilic allergic diseases, including severe asthma and chronic rhinosinusitis. This study aimed to demonstrate the role of 12/15-lipoxygenase (12/15-LOX) in interleukin (IL)-33-induced eosinophilic airway inflammation; to this end, we used 12/15-LOX-deficient mice, which displayed augmented IL-33-induced lung inflammation, characterized by an increased number of infiltrated eosinophils and group 2 innate lymphoid cells (ILC2s) in the airway. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipidomics revealed that the levels of a series of 12/15-LOX-derived metabolites were significantly decreased, and application of 14(S)-hydroxy docosahexaenoic acid (HDoHE), a major 12/15-LOX-derived product, suppressed IL-33-mediated eosinophilic inflammation in 12/15-LOX-deficient mice. Using bioactive lipid screening, we found that 14(S)-HDoHE and 10(S),17(S)-diHDoHE markedly attenuated ILC2 proliferation and cytokine production at micromolar concentration in vitro. In addition, maresin 1 (MaR1) and resolvin D1 (RvD1), 12/15-LOX-derived specialized proresolving mediators (SPMs), inhibited cytokine production of ILC2s at nanomolar concentration. These findings demonstrate the protective role of endogenous 12/15-LOX-derived lipid mediators in controlling ILC2-mediated eosinophilic airway inflammation and related diseases. Thus, 12/15-LOX-derived lipid mediators may represent a potential therapeutic strategy for ameliorating airway inflammation-associated conditions.
Collapse
Affiliation(s)
- Jun Miyata
- Laboratory of Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Division of Infectious Diseases and Respiratory Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yoshiyuki Yokokura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Laboratory for Innate Immune Systems, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroyuki Arai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Arita
- Laboratory of Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Physiological Chemistry and Metabolism, Keio University Faculty of Pharmacy, Tokyo, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
72
|
Hossain MR, Ansary TM, Komine M, Ohtsuki M. Diversified Stimuli-Induced Inflammatory Pathways Cause Skin Pigmentation. Int J Mol Sci 2021; 22:3970. [PMID: 33921371 PMCID: PMC8070342 DOI: 10.3390/ijms22083970] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023] Open
Abstract
The production of melanin pigments by melanocytes and their quantity, quality, and distribution play a decisive role in determining human skin, eye, and hair color, and protect the skin from adverse effects of ultraviolet radiation (UVR) and oxidative stress from various environmental pollutants. Melanocytes reside in the basal layer of the interfollicular epidermis and are compensated by melanocyte stem cells in the follicular bulge area. Various stimuli such as eczema, microbial infection, ultraviolet light exposure, mechanical injury, and aging provoke skin inflammation. These acute or chronic inflammatory responses cause inflammatory cytokine production from epidermal keratinocytes as well as dermal fibroblasts and other cells, which in turn stimulate melanocytes, often resulting in skin pigmentation. It is confirmed by some recent studies that several interleukins (ILs) and other inflammatory mediators modulate the proliferation and differentiation of human epidermal melanocytes and also promote or inhibit expression of melanogenesis-related gene expression directly or indirectly, thereby participating in regulation of skin pigmentation. Understanding of mechanisms of skin pigmentation due to inflammation helps to elucidate the relationship between inflammation and skin pigmentation regulation and can guide development of new therapeutic pathways for treating pigmented dermatosis. This review covers the mechanistic aspects of skin pigmentation caused by inflammation.
Collapse
Affiliation(s)
| | | | - Mayumi Komine
- Department of Dermatology, Faculty of Medicine, Jichi Medical University, Tochigi 329-0498, Japan; (M.R.H.); (T.M.A.); (M.O.)
| | | |
Collapse
|
73
|
Saikumar Jayalatha AK, Hesse L, Ketelaar ME, Koppelman GH, Nawijn MC. The central role of IL-33/IL-1RL1 pathway in asthma: From pathogenesis to intervention. Pharmacol Ther 2021; 225:107847. [PMID: 33819560 DOI: 10.1016/j.pharmthera.2021.107847] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-33 (IL-33), a member of the IL-1 family, and its cognate receptor, Interleukin-1 receptor like-1 (IL-1RL1 or ST2), are susceptibility genes for childhood asthma. In response to cellular damage, IL-33 is released from barrier tissues as an 'alarmin' to activate the innate immune response. IL-33 drives type 2 responses by inducing signalling through its receptor IL-1RL1 in several immune and structural cells, thereby leading to type 2 cytokine and chemokine production. IL-1RL1 gene transcript encodes different isoforms generated through alternative splicing. Its soluble isoform, IL-1RL1-a or sST2, acts as a decoy receptor by sequestering IL-33, thereby inhibiting IL1RL1-b/IL-33 signalling. IL-33 and its receptor IL-1RL1 are therefore considered as putative biomarkers or targets for pharmacological intervention in asthma. This review will provide an overview of the genetics and biology of the IL-33/IL-1RL1 pathway in the context of asthma pathogenesis. It will discuss the potential and complexities of targeting the cytokine or its receptor, how genetics or biomarkers may inform precision medicine for asthma targeting this pathway, and the possible positioning of therapeutics targeting IL-33 or its receptor in the expanding landscape of novel biologicals applied in asthma management.
Collapse
Affiliation(s)
- A K Saikumar Jayalatha
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - L Hesse
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - M E Ketelaar
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - G H Koppelman
- University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - M C Nawijn
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands.
| |
Collapse
|
74
|
Shiratori-Hayashi M, Yamaguchi C, Eguchi K, Shiraishi Y, Kohno K, Mikoshiba K, Inoue K, Nishida M, Tsuda M. Astrocytic STAT3 activation and chronic itch require IP 3R1/TRPC-dependent Ca 2+ signals in mice. J Allergy Clin Immunol 2021; 147:1341-1353. [PMID: 32781002 DOI: 10.1016/j.jaci.2020.06.039] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Chronic itch is a debilitating symptom of inflammatory skin diseases, but the underlying mechanism is poorly understood. We have recently demonstrated that astrocytes in the spinal dorsal horn become reactive in models of atopic and contact dermatitis via activation of the transcription factor signal transducer and activator of transcription 3 (STAT3) and critically contribute to chronic itch. In general, STAT3 is transiently activated; however, STAT3 activation in reactive astrocytes of chronic itch model mice persistently occurs via an unknown mechanism. OBJECTIVE We aimed to determine the mechanisms of persistent activation of astrocytic STAT3 in chronic itch conditions. METHODS To determine the factors that are required for persistent activation of astrocytic STAT3, Western blotting and calcium imaging with cultured astrocytes or spinal cord slices were performed. Thereafter, chronic itch model mice were used for genetic and behavioral experiments to confirm the role of the factors determined to mediate persistent STAT3 activation from in vitro and ex vivo experiments in chronic itch. RESULTS IP3 receptor type 1 (IP3R1) knockdown in astrocytes suppressed IL-6-induced persistent STAT3 activation and expression of lipocalin-2 (LCN2), an astrocytic STAT3-dependent inflammatory factor that is required for chronic itch. IP3R1-dependent astrocytic Ca2+ responses involved Ca2+ influx through the cation channel transient receptor potential canonical (TRPC), which was required for persistent STAT3 activation evoked by IL-6. IL-6 expression was upregulated in dorsal root ganglion neurons in a mouse model of chronic itch. Dorsal root ganglion neuron-specific IL-6 knockdown, spinal astrocyte-specific IP3R1 knockdown, and pharmacologic spinal TRPC inhibition attenuated LCN2 expression and chronic itch. CONCLUSION Our findings suggest that IP3R1/TRPC channel-mediated Ca2+ signals elicited by IL-6 in astrocytes are necessary for persistent STAT3 activation, LCN2 expression, and chronic itch, and they may also provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Miho Shiratori-Hayashi
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Chiharu Yamaguchi
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazushi Eguchi
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuto Shiraishi
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Keita Kohno
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhiko Mikoshiba
- RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan; Faculty of Science, Toho University, Chiba, Japan; Shanghai Institute of Immunochemical Studies, Shanghai Tech University, Shanghai, China
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Motohiro Nishida
- Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences and Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Aichi, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
75
|
Peng L, Sun W, Chen L, Wen WP. The Role of Interleukin-33 in Head and Neck Squamous Cell Carcinoma Is Determined by Its Cellular Sources in the Tumor Microenvironment. Front Oncol 2021; 10:588454. [PMID: 33634017 PMCID: PMC7902021 DOI: 10.3389/fonc.2020.588454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/24/2020] [Indexed: 12/09/2022] Open
Abstract
Objectives To investigate the role of interleukin-33 (IL-33) in head and neck squamous cell carcinoma (HNSCC). Materials and Methods RNA-seq data of 520 cases of HNSCC were retrieved from The Cancer Genome Atlas. The tumor microenvironment was deconstructed by xCell using bulk RNA-seq data. The cohort was dichotomized by the median IL-33 expression level. Immune cell components and molecular markers were compared between the high and low IL-33 groups. The prognostic value of IL-33 was evaluated by the log-rank test. Differential gene expression analysis and KEGG pathway enrichment analysis were also conducted. The relationship between the IL-33 expression level and the abundance of its potential cellular sources was evaluated by Pearson’s partial correlation test. Subgroup analysis was conducted in laryngeal, oropharyngeal, and oral cavity squamous cell carcinoma (LSCC, OPSCC, and OCSCC). Results The role of IL-33 in HNSCC was heterogeneous among tumors at different sites. In LSCC, IL-33 may increase the extent of malignancy of tumor cells and act as a pro-tumor factor. In OCSCC, IL-33 may play a role in orchestrating the immune responses against tumor cells and act as an antitumor factor. The role of IL-33 in OPSCC was undetermined. IL-33 in LSCC was mainly derived from endothelial cells, while IL-33 in OCSCC was mainly derived from endothelial and epithelial cells. Conclusion According to the different sources of IL-33 in LSCC and OCSCC, we propose a hypothesis that stroma-derived IL-33 could favor tumor progression, while epithelial-derived IL-33 could favor antitumor immune responses in HNSCC.
Collapse
Affiliation(s)
- Liang Peng
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei-Ping Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
76
|
Liu EG, Yin X, Swaminathan A, Eisenbarth SC. Antigen-Presenting Cells in Food Tolerance and Allergy. Front Immunol 2021; 11:616020. [PMID: 33488627 PMCID: PMC7821622 DOI: 10.3389/fimmu.2020.616020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Food allergy now affects 6%-8% of children in the Western world; despite this, we understand little about why certain people become sensitized to food allergens. The dominant form of food allergy is mediated by food-specific immunoglobulin E (IgE) antibodies, which can cause a variety of symptoms, including life-threatening anaphylaxis. A central step in this immune response to food antigens that differentiates tolerance from allergy is the initial priming of T cells by antigen-presenting cells (APCs), primarily different types of dendritic cells (DCs). DCs, along with monocyte and macrophage populations, dictate oral tolerance versus allergy by shaping the T cell and subsequent B cell antibody response. A growing body of literature has shed light on the conditions under which antigen presentation occurs and how different types of T cell responses are induced by different APCs. We will review APC subsets in the gut and discuss mechanisms of APC-induced oral tolerance versus allergy to food identified using mouse models and patient samples.
Collapse
Affiliation(s)
- Elise G. Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
- Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT, United States
| | - Xiangyun Yin
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Anush Swaminathan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Stephanie C. Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
- Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
77
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
78
|
Hung LY, Tanaka Y, Herbine K, Pastore C, Singh B, Ferguson A, Vora N, Douglas B, Zullo K, Behrens EM, Li Hui Tan T, Kohanski MA, Bryce P, Lin C, Kambayashi T, Reed DR, Brown BL, Cohen NA, Herbert DR. Cellular context of IL-33 expression dictates impact on anti-helminth immunity. Sci Immunol 2020; 5:5/53/eabc6259. [PMID: 33188058 DOI: 10.1126/sciimmunol.abc6259] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
Interleukin-33 (IL-33) is a pleiotropic cytokine that can promote type 2 inflammation but also drives immunoregulation through Foxp3+Treg expansion. How IL-33 is exported from cells to serve this dual role in immunosuppression and inflammation remains unclear. Here, we demonstrate that the biological consequences of IL-33 activity are dictated by its cellular source. Whereas IL-33 derived from epithelial cells stimulates group 2 innate lymphoid cell (ILC2)-driven type 2 immunity and parasite clearance, we report that IL-33 derived from myeloid antigen-presenting cells (APCs) suppresses host-protective inflammatory responses. Conditional deletion of IL-33 in CD11c-expressing cells resulted in lowered numbers of intestinal Foxp3+Treg cells that express the transcription factor GATA3 and the IL-33 receptor ST2, causing elevated IL-5 and IL-13 production and accelerated anti-helminth immunity. We demonstrate that cell-intrinsic IL-33 promoted mouse dendritic cells (DCs) to express the pore-forming protein perforin-2, which may function as a conduit on the plasma membrane facilitating IL-33 export. Lack of perforin-2 in DCs blocked the proliferative expansion of the ST2+Foxp3+Treg subset. We propose that perforin-2 can provide a plasma membrane conduit in DCs that promotes the export of IL-33, contributing to mucosal immunoregulation under steady-state and infectious conditions.
Collapse
Affiliation(s)
- Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yukinori Tanaka
- Department of Dental Anesthesiology and Pain Management, Tohoku University Hospital, Sendai, Miyagi 980-8574, Japan
| | - Karl Herbine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brenal Singh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Annabel Ferguson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nisha Vora
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bonnie Douglas
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelly Zullo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward M Behrens
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Tiffany Li Hui Tan
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Kohanski
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Bryce
- Immunology and Inflammation Therapeutic Area, Sanofi US, Cambridge, MA 02319, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Breann L Brown
- Department of Biochemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Noam A Cohen
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104, USA.,Monell Chemical Senses Center, Philadelphia, PA 19104, USA.,Michael J. Crescenz Veterans Affairs Medical Center Surgical Service, Philadelphia, PA 19104, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
79
|
Hu J, Gao N, Zhang Y, Chen X, Li J, Bian F, Chi W, Liu Z, de Paiva C, Pflugfelder SC, Li DQ. IL-33/ST2/IL-9/IL-9R signaling disrupts ocular surface barrier in allergic inflammation. Mucosal Immunol 2020; 13:919-930. [PMID: 32358573 PMCID: PMC7572432 DOI: 10.1038/s41385-020-0288-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/05/2020] [Accepted: 03/24/2020] [Indexed: 02/04/2023]
Abstract
This study was to explore a novel IL-33/ST2/IL-9/IL-9R signaling pathway that disrupts ocular surface barrier and amplifies allergic inflammation. Two murine models of experimental allergic conjunctivitis (EAC) and IL-9 topical challenge in wild type Balb/c and ST2-/- mice, and two culture models of primarily human corneal epithelial cells (HCECs) and mouse CD4+ T cells were performed. Clinical manifestations, Oregon-Green Dextran (OGD) staining, the apical junction complexes (AJCs), IL-33/ST2 and IL-9/IL-9R signaling molecules were evaluated in ocular surface and its draining cervical lymph nodes (CLNs) by RT-qPCR, immunostaining and ELISA. The typical allergic signs, enhanced OGD staining intensity, disrupted morphology of AJCs, including ZO-1, claudin 1, occludin, and E-cadherin, and the stimulated signaling of IL-33/ST2 and IL-9/IL-9R were observed in ocular mucosa and draining CLNs in EAC-Balb/c mice, but significantly reduced or eliminated in EAC-ST2-/- mice. Topical challenge of IL-9 resulted in the obvious OGD staining and disrupted ocular surface AJCs in Balb/c mice and in HCECs in vitro. IL-9 production was found to be stimulated by IL-33 in CD4+ cells from Balb/c mice in vitro. Our findings uncovered a novel phenomenon and mechanism by which ocular surface barrier integrity is disrupted in allergic conjunctivitis by IL-33/ST2/IL-9/IL-9R signaling pathway, which may amplify the allergic inflammation.
Collapse
Affiliation(s)
- Jiaoyue Hu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA,Eye institute of Xiamen University and Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ning Gao
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA,Department of Ophthalmology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yun Zhang
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA,School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Xin Chen
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA,School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Jinmiao Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Fang Bian
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Wei Chi
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Zuguo Liu
- Eye institute of Xiamen University and Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Cintia de Paiva
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen C. Pflugfelder
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| | - De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA,Correspondence to: De-Quan Li, M.D., Ph.D. Associate Professor, Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin Street, NC-205, Houston, TX 77030, USA. Tel: (713) 798-1123, Fax: (713) 798-1457,
| |
Collapse
|
80
|
Substance P Release by Sensory Neurons Triggers Dendritic Cell Migration and Initiates the Type-2 Immune Response to Allergens. Immunity 2020; 53:1063-1077.e7. [PMID: 33098765 DOI: 10.1016/j.immuni.2020.10.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/21/2020] [Accepted: 09/30/2020] [Indexed: 11/20/2022]
Abstract
Dendritic cells (DCs) of the cDC2 lineage initiate allergic immunity and in the dermis are marked by their expression of CD301b. CD301b+ dermal DCs respond to allergens encountered in vivo, but not in vitro. This suggests that another cell in the dermis may sense allergens and relay that information to activate and induce the migration of CD301b+ DCs to the draining lymph node (dLN). Using a model of cutaneous allergen exposure, we show that allergens directly activated TRPV1+ sensory neurons leading to itch and pain behaviors. Allergen-activated sensory neurons released the neuropeptide Substance P, which stimulated proximally located CD301b+ DCs through the Mas-related G-protein coupled receptor member A1 (MRGPRA1). Substance P induced CD301b+ DC migration to the dLN where they initiated T helper-2 cell differentiation. Thus, sensory neurons act as primary sensors of allergens, linking exposure to activation of allergic-skewing DCs and the initiation of an allergic immune response.
Collapse
|
81
|
Xiao Q, Song Y, Chu H, Tang M, Jiang J, Meng Q, Hao W, Wei X. 1,4NQ-BC enhances the lung inflammation by mediating the secretion of IL-33 which derived from macrophages. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114729. [PMID: 32563138 DOI: 10.1016/j.envpol.2020.114729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/17/2020] [Accepted: 05/01/2020] [Indexed: 06/11/2023]
Abstract
Black carbon (BC) is a product of incomplete combustion of fossil fuels and vegetation. The compelling evidence has demonstrated that it has a close relationship with several respiratory and cardiovascular diseases. BC provides the reactive sites and surfaces to absorb various chemicals, such as polycyclic aromatic hydrocarbons (PAH). Naphthoquinone is a typical PAHs which was found in particulate matter (PM) and 1,4NQ-BC owned high oxidative potential and cytotoxicity. IL-33 is an alarmin which increases innate immunity through Th2 responses. It was reported that IL-33 was a potent inducer of pro-inflammatory cytokines, like IL-6. In our previous study, it was revealed that 1,4NQ-BC instilled intratracheally to mice could trigger the lung inflammation and stimulate the secretion of IL-33 in lung tissue. We found that IL-33 could induce inflammation in lung itself. When the macrophages were eliminated, the secretion of IL-33 was reduced and the pathological damage in the lung was relieved after exposure to 1,4NQ-BC. Both MAPK and PI3K/AKT signal pathways were involved in the process of IL-33 secretion and the lung inflammation induced by 1,4NQ-BC. The findings herein support the notion that after exposure to 1,4NQ-BC, the increased secretion of IL-33 was mainly derived from macrophages through both MAPK and PI3K/AKT signal pathways.
Collapse
Affiliation(s)
- Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Yiming Song
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Hongqian Chu
- Translational Medicine Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, PR China; Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, PR China
| | - Mengmeng Tang
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Jianjun Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Qinghe Meng
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, PR China.
| |
Collapse
|
82
|
Brandt EB, Bolcas PE, Ruff BP, Khurana Hershey GK. IL33 contributes to diesel pollution-mediated increase in experimental asthma severity. Allergy 2020; 75:2254-2266. [PMID: 31922608 DOI: 10.1111/all.14181] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 10/25/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Exposure to traffic pollution, notably diesel exhaust particles (DEP), increases risk for asthma and asthma exacerbations. The contribution of cytokines generated by stressed lung epithelial cells (IL25, IL33, TSLP) to DEP-induced asthma severity remains poorly understood. METHODS BALB/c mice were exposed intratracheally once to DEP or 9 times over 3-weeks to either saline, DEP, and/or house dust mite extract (HDM). Airway hyper-responsiveness (AHR), pulmonary inflammation, and T-cell subsets were assessed 24 hours after the last exposure in mice sufficient and deficient for the IL33 receptor ST2. RESULTS DEP exposure induces oxidative stress, IL6, neutrophils and pulmonary accumulation of IL33, but not IL25 or TSLP or other features of allergic disease. When mice are co-exposed to DEP and low doses of HDM, DEP increases IL33 lung levels and Th2 responses. ST2 deficiency partially protected mice from HDM + DEP induced AHR in association with decreased type 2 inflammation and lung levels of IL5+ IL17A+ co-producing T-cells. Upon in vitro HDM challenge of lung cells from HDM ± DEP exposed ST2-/- mice, secretion of IL5, IL13, IL6 and IL17A was abrogated by a mechanism involving IL33 signaling in both dendritic cells and T-cells. HDM + DEP exposed bone marrow derived dendritic cells and IL33 pulsed BMDC promote a mixed Th2/Th17 response that was dependent on ST2 expression by CD4+ T-cells. CONCLUSION IL33 contributes to DEP mediated increase in allergen-induced Th2 inflammation and AHR in a mouse model of severe steroid resistant asthma, potentially through the accumulation of pathogenic IL5+ IL17A+ CD4+ effector T-cells.
Collapse
Affiliation(s)
- Eric B. Brandt
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Paige E. Bolcas
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Brandy P. Ruff
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| |
Collapse
|
83
|
Paplinska-Goryca M, Misiukiewicz-Stepien P, Proboszcz M, Nejman-Gryz P, Gorska K, Krenke R. The Expressions of TSLP, IL-33, and IL-17A in Monocyte Derived Dendritic Cells from Asthma and COPD Patients are Related to Epithelial-Macrophage Interactions. Cells 2020; 9:cells9091944. [PMID: 32842623 PMCID: PMC7565129 DOI: 10.3390/cells9091944] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cross-talk between the external and internal environment in the respiratory tract involves macrophage/dendritic cell (DC) transepithelial network. Epithelium triggers dendritic cell-mediated inflammation by producing thymic stromal lymphopoietin (TSLP), IL-33, and IL-17A. The study aimed to evaluate the expression of TSLP, IL-33, and IL-17A in human monocyte derived dendritic cells (moDCs) co-cultured with respiratory epithelium and monocyte derived macrophages (moMφs) in asthma, chronic obstructive pulmonary disease (COPD) and healthy controls. METHODS The study used a triple-cell co-culture model, utilizing nasal epithelial cells, along with moMφs and moDCs. Cells were cultured in mono-, di-, and triple-co-cultures for 24 h. RESULTS Co-culture with epithelium and moMφs significantly increased TSLP in asthma and did not change IL-33 and IL-17A mRNA expression in moDCs. moDCs from asthmatics were characterized by the highest TSLP mRNA expression and the richest population of TSLPR, ST2, and IL17RA expressed cells. A high number of positive correlations between the assessed cytokines and CHI3L1, IL-12p40, IL-1β, IL-6, IL-8, TNF in moDCs was observed in asthma and COPD. CONCLUSION TSLP, IL-33, and IL-17A expression in moDCs are differently regulated by epithelium in asthma, COPD, and healthy subjects. These complex cell-cell interactions may impact airway inflammation and be an important factor in the pathobiology of asthma and COPD.
Collapse
Affiliation(s)
- Magdalena Paplinska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.P.); (P.N.-G.); (K.G.); (R.K.)
- Correspondence: ; Tel.: +48-225991241; Fax +48-225991561
| | | | - Malgorzata Proboszcz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.P.); (P.N.-G.); (K.G.); (R.K.)
| | - Patrycja Nejman-Gryz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.P.); (P.N.-G.); (K.G.); (R.K.)
| | - Katarzyna Gorska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.P.); (P.N.-G.); (K.G.); (R.K.)
| | - Rafal Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.P.); (P.N.-G.); (K.G.); (R.K.)
| |
Collapse
|
84
|
Zhang X, Chen W, Zeng P, Xu J, Diao H. The Contradictory Role of Interleukin-33 in Immune Cells and Tumor Immunity. Cancer Manag Res 2020; 12:7527-7537. [PMID: 32904627 PMCID: PMC7457384 DOI: 10.2147/cmar.s262745] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/02/2020] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-33 is a member of the IL-1 superfamily and is a crucial cytokine playing the role of a dual-function molecule. IL-33 mediates its function by interacting with its receptor suppression of tumorigenicity 2 (ST2), which is constitutively expressed on T helper (Th)1 cells, Th2 cells, and other immune cells. Previously, we summarized findings on IL-33 and performed an intensive study of the correlation between IL-33 and tumor. IL-33 enables anti-tumor immune responses through Th1 cells and natural killer (NK) cells and plays a role in tumor immune escape in cancers via Th2 cells and regulatory T cells. Herein, we discuss the contradictory role of IL-33 in immune cells in different cancer, and our summaries may be helpful for better understanding of the development of research on IL-33 and tumor immunity.
Collapse
Affiliation(s)
- Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Wenbiao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Ping Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Jia Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| |
Collapse
|
85
|
Topal FA, Zuberbier T, Makris MP, Hofmann M. The role of IL-17, IL-23 and IL-31, IL-33 in allergic skin diseases. Curr Opin Allergy Clin Immunol 2020; 20:367-373. [PMID: 32590505 DOI: 10.1097/aci.0000000000000658] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Allergic skin diseases such as urticaria, atopic dermatitis and allergic contact dermatitis are among the most common skin diseases with severe socioeconomic consequences. The pathogenesis of allergic skin diseases is complex. This review provides an overview of cytocines IL-17, IL-23, IL-31 and IL-33. RECENT FINDINGS Current research results show a variety of immunological processes in the pathogenesis of the allergic skin diseases, including the role of cytokines. In addition to the Th1 and Th2 immune response, the immune response via Th17 is becoming increasingly important in allergic skin diseases but also the cytokines IL-23, IL-31 and IL-33 have been discussed in the literature recently. Different cytokines promote in a kind of orchestra the different symptoms seen in the different allergic skin diseases, including pruritus, dermatitis, mast cell mediator release and inflammation. SUMMARY We are still in the early stages of understanding pathophysiology of allergic skin diseases and the role of various cytokines in the immune system. With the development of targeted antibodies against the proinflammatory cytokines, the variety of normal therapeutic options can be expected to evolve.
Collapse
MESH Headings
- Animals
- Clinical Trials as Topic
- Dermatitis, Allergic Contact/drug therapy
- Dermatitis, Allergic Contact/immunology
- Dermatitis, Allergic Contact/pathology
- Dermatitis, Atopic/drug therapy
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/pathology
- Disease Models, Animal
- Humans
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Interleukin-33/antagonists & inhibitors
- Interleukin-33/immunology
- Interleukin-33/metabolism
- Interleukins/antagonists & inhibitors
- Interleukins/immunology
- Interleukins/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Skin/drug effects
- Skin/immunology
- Skin/pathology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Treatment Outcome
- Urticaria/drug therapy
- Urticaria/immunology
- Urticaria/pathology
Collapse
Affiliation(s)
- Fatih A Topal
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Campus Benjamin-Franklin, Berlin, Germany
| | - Torsten Zuberbier
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Campus Benjamin-Franklin, Berlin, Germany
| | - Michael P Makris
- Allergy Unit 'D. Kalogeromitros', 2nd Department of Dermatology and Venereology, National and Kapodistrian University of Athens, Medical School, University General Hospital 'ATTIKON', Athens, Greece
| | - Maja Hofmann
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
86
|
Yang Q, Kong L, Huang W, Mohammadtursun N, Li X, Wang G, Wang L. Osthole attenuates ovalbumin‑induced lung inflammation via the inhibition of IL‑33/ST2 signaling in asthmatic mice. Int J Mol Med 2020; 46:1389-1398. [PMID: 32700747 PMCID: PMC7447319 DOI: 10.3892/ijmm.2020.4682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/03/2020] [Indexed: 01/12/2023] Open
Abstract
Asthma is a common chronic inflammatory airway disease. Recent studies have reported that interleukin (IL)-33 is a potential link between the airway epithelium and Th2-type inflammatory responses, which are closely related to the progression of asthma. The IL-33 receptor, ST2, is highly expressed in group 2 innate lymphoid cells (ILC2s), Th2 cells, mast cells, eosinophils and natural killer (NK) cells. Cnidii Fructus is a Chinese herb with a long history of use in the treatment of asthma in China. Osthole is one of the major components of Cnidii Fructus. The present study examined the anti-asthmatic effects of osthole in mice and aimed to elucidate the underlying mechanisms involving the IL-33/ST2 pathway. BALB/c mice were sensitized and challenged with ovalbumin and then treated with an intraperitoneal injection of osthole (25 and 50 mg/kg). Subsequently, the airway hyper-responsiveness (AHR) and inflammation of the lungs were evaluated. The amounts of IL-4, IL-5, IL-13, interferon (IFN)-γ and IL-33 in the bronchoalveolar lavage fluid (BALF) were measured by Luminex assay and their mRNA levels in the lungs were measured by reverse transcription-quantitative PCR. The histopathology of the lungs was performed with H&E, PAS and Masson's staining. The expression of ST2 in the lungs was evaluated by immunohistochemistry. The data demonstrated that osthole markedly reduced AHR and decreased the number of eosinophils and lymphocytes in BALF. It was also observed that osthole significantly inhibited the release of Th2-type cytokines (IL-4, IL-5 and IL-13) and upregulated the IFN-γ level in BALF. Moreover, osthole significantly attenuated the IL-33 and ST2 expression in the lungs of asthmatic mice. On the whole, osthole attenuated ovalbumin-induced lung inflammation through the inhibition of IL-33/ST2 signaling in an asthmatic mouse model. These results suggest that osthole is a promising target for the development of an asthma medication.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Respiratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Lingwen Kong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Weiling Huang
- Department of Respiratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Nabijan Mohammadtursun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiumin Li
- Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Guifang Wang
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Lixin Wang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, P.R. China
| |
Collapse
|
87
|
Giovannetti A, Straface E, Rosato E, Casciaro M, Pioggia G, Gangemi S. Role of Alarmins in the Pathogenesis of Systemic Sclerosis. Int J Mol Sci 2020; 21:ijms21144985. [PMID: 32679721 PMCID: PMC7404317 DOI: 10.3390/ijms21144985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare chronic autoimmune disease associated with significant morbidity and mortality. Two main subsets of SSc are recognized: (i) diffuse cutaneous SSc with rapidly progressive fibrosis of the skin, lungs, and other internal organs; and (ii) limited cutaneous SSc, which is dominated by vascular manifestations, with skin and organ fibrosis generally limited and slowly progressing. In spite of intense investigation, both etiology and pathogenesis of SSc are still unknown. Genetic and environmental factors, as well as abnormalities of immune functions, are strongly suggested for etiology, while microvascular abnormalities, immune system activation, and oxidative stress are suggested for the pathogenesis. Recently, it has been found that a multitude of mediators and cytokines are implicated in the fibrotic processes observed in SSc. Among these, a central role could be exerted by “alarmins”, endogenous and constitutively expressed proteins/peptides that function as an intercellular signal defense. This review describes, in a detailed manner, the role of alarmins in the pathogenesis of scleroderma.
Collapse
Affiliation(s)
- Antonello Giovannetti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: ; Tel.: +39-3476138512
| | - Elisabetta Straface
- Center for Gender-Specific Medicine, Biomarkers Unit, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Marco Casciaro
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (S.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
88
|
Jaiswal AK, Makhija S, Stahr N, Sandey M, Suryawanshi A, Mishra A. Pyruvate kinase M2 in lung APCs regulates Alternaria-induced airway inflammation. Immunobiology 2020; 225:151956. [PMID: 32747016 PMCID: PMC7403530 DOI: 10.1016/j.imbio.2020.151956] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/21/2022]
Abstract
Sensitivity to allergenic fungi (Alternaria alternata) is associated with acute, severe asthma attacks. Antigen presenting cells (APCs) in the lung sense environmental perturbations that induce cellular stress and metabolic changes and are critical for allergic airway inflammation. However, the mechanisms underlying such environmental sensing by APCs in the lung remains unclear. Here we show that acute Alternaria challenge rapidly induces neutrophil accumulation in airways, and alter expressions of Pyruvate Kinase (PKM2) and hypoxia-inducible factor -1α (Hif-1α) that correlates with proinflammatory mediator release. Blockade of IL33 signaling in vivo led to reduce oxidative stress and glycolysis in lung APCs. Lung-specific ablation of CD11c+ cells abrogates Alternaria-induced neutrophil accumulation and inflammation. Furthermore, administration of Alternaria into the airways stimulated APCs and elevate the expression of Glut-1. Mechanistically, we establish that PKM2 is a critical modulator of lung APC activation in Alternaria-induced acute inflammation. Allosteric activation of PKM2 by a small molecule ML265 or siRNA-mediated knock down correlated negatively with glycolysis and activation of APCs. These results collectively demonstrates that PKM2-mediated glycolytic reprogramming by fungal allergen Alternaria influences lung APC activation, thereby promotes acute airway inflammation. Our data support a model in which Alternaria sensitization in airways induce a circuitry of glycolysis and PKM2 regulation that confers an acute activation of APCs in the lung, whose targeting might represent a strategy for asthma treatment.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Sangeet Makhija
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Natalie Stahr
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
89
|
Serum amyloid A is a soluble pattern recognition receptor that drives type 2 immunity. Nat Immunol 2020; 21:756-765. [PMID: 32572240 DOI: 10.1038/s41590-020-0698-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
The molecular basis for the propensity of a small number of environmental proteins to provoke allergic responses is largely unknown. Herein, we report that mite group 13 allergens of the fatty acid-binding protein (FABP) family are sensed by an evolutionarily conserved acute-phase protein, serum amyloid A1 (SAA1), that promotes pulmonary type 2 immunity. Mechanistically, SAA1 interacted directly with allergenic mite FABPs (Der p 13 and Blo t 13). The interaction between mite FABPs and SAA1 activated the SAA1-binding receptor, formyl peptide receptor 2 (FPR2), which drove the epithelial release of the type-2-promoting cytokine interleukin (IL)-33 in a SAA1-dependent manner. Importantly, the SAA1-FPR2-IL-33 axis was upregulated in nasal epithelial cells from patients with chronic rhinosinusitis. These findings identify an unrecognized role for SAA1 as a soluble pattern recognition receptor for conserved FABPs found in common mite allergens that initiate type 2 immunity at mucosal surfaces.
Collapse
|
90
|
The IL-33-induced p38-/JNK1/2-TNFα axis is antagonized by activation of β-adrenergic-receptors in dendritic cells. Sci Rep 2020; 10:8152. [PMID: 32424229 PMCID: PMC7235212 DOI: 10.1038/s41598-020-65072-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/27/2020] [Indexed: 12/17/2022] Open
Abstract
IL-33, an IL-1 cytokine superfamily member, induces the activation of the canonical NF-κB signaling, and of Mitogen Activated Protein Kinases (MAPKs). In dendritic cells (DCs) IL-33 induces the production of IL-6, IL-13 and TNFα. Thereby, the production of IL-6 depends on RelA whereas the production of IL-13 depends on the p38-MK2/3 signaling module. Here, we show that in addition to p65 and the p38-MK2/3 signaling module, JNK1/2 are essential for the IL-33-induced TNFα production. The central roles of JNK1/2 and p38 in DCs are underpinned by the fact that these two MAPK pathways are controlled by activated β-adrenergic receptors resulting in a selective regulation of the IL-33-induced TNFα response in DCs.
Collapse
|
91
|
Van der Jeught K, Sun Y, Fang Y, Zhou Z, Jiang H, Yu T, Yang J, Kamocka MM, So KM, Li Y, Eyvani H, Sandusky GE, Frieden M, Braun H, Beyaert R, He X, Zhang X, Zhang C, Paczesny S, Lu X. ST2 as checkpoint target for colorectal cancer immunotherapy. JCI Insight 2020; 5:136073. [PMID: 32376804 PMCID: PMC7253019 DOI: 10.1172/jci.insight.136073] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint blockade immunotherapy delivers promising clinical results in colorectal cancer (CRC). However, only a fraction of cancer patients develop durable responses. The tumor microenvironment (TME) negatively impacts tumor immunity and subsequently clinical outcomes. Therefore, there is a need to identify other checkpoint targets associated with the TME. Early-onset factors secreted by stromal cells as well as tumor cells often help recruit immune cells to the TME, among which are alarmins such as IL-33. The only known receptor for IL-33 is stimulation 2 (ST2). Here we demonstrated that high ST2 expression is associated with poor survival and is correlated with low CD8+ T cell cytotoxicity in CRC patients. ST2 is particularly expressed in tumor-associated macrophages (TAMs). In preclinical models of CRC, we demonstrated that ST2-expressing TAMs (ST2+ TAMs) were recruited into the tumor via CXCR3 expression and exacerbated the immunosuppressive TME; and that combination of ST2 depletion using ST2-KO mice with anti-programmed death 1 treatment resulted in profound growth inhibition of CRC. Finally, using the IL-33trap fusion protein, we suppressed CRC tumor growth and decreased tumor-infiltrating ST2+ TAMs. Together, our findings suggest that ST2 could serve as a potential checkpoint target for CRC immunotherapy.
Collapse
Affiliation(s)
| | - Yifan Sun
- Department of Medical and Molecular Genetics
| | | | | | | | - Tao Yu
- Department of Medical and Molecular Genetics
| | | | | | - Ka Man So
- Center for Computational Biology and Bioinformatics
| | - Yujing Li
- Department of Medical and Molecular Genetics
| | | | - George E. Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Harald Braun
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xiaoming He
- Fischell Department of Bioengineering and
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Xinna Zhang
- Department of Medical and Molecular Genetics
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics
- Center for Computational Biology and Bioinformatics
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sophie Paczesny
- Department of Pediatrics
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
92
|
Choi J, Park SY, Moon K, Ha EH, Woo YD, Chung DH, Kwon H, Kim T, Park H, Moon H, Song W, Cho YS. Macrophage-derived progranulin promotes allergen-induced airway inflammation. Allergy 2020; 75:1133-1145. [PMID: 31758561 DOI: 10.1111/all.14129] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/22/2019] [Accepted: 10/06/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Progranulin (PGRN), mainly produced by immune and epithelial cells, has been known to be involved in the development of various inflammatory diseases. However, the function of PGRN in allergic airway inflammation has not been clearly elucidated, and we investigated the role of PGRN in allergic airway inflammation. METHODS Production of PGRN and various type 2 cytokines was evaluated in mouse airways exposed to house dust mite allergen, and main cellular sources of these molecules were investigated using macrophage, airway epithelial cell, and NKT cell lines. We elucidated the role of PGRN in allergic airway inflammation in mouse models of asthma using macrophage-derived PGRN-deficient mice and NKT cell knockout mice by evaluating cytokine levels in bronchoalveolar lavage fluids and histopathology. We also supplemented recombinant PGRN in the mouse models to confirm the role of PGRN in allergic airway inflammation. RESULTS PGRN production preceded other cytokines, mainly from macrophages, in the airway exposed to allergen. PGRN induced IL-4 and IL-13 production in NKT cells and IL-33 and TSLP in airway epithelial cells. PGRN-induced Th2 cytokine production was abolished in NKT-deficient mice. Finally, allergic inflammation was significantly attenuated in allergen-exposed PGRN-deficient mice, but inflammation was restored when recombinant PGRN was supplemented during the allergen sensitization period. CONCLUSION The presence of macrophage-derived PGRN in airways in the early sensitization period may be critical for mounting a Th2 immune response and for following an allergic airway inflammation pathway via induction of type 2 cytokine production in NKT and airway epithelial cells.
Collapse
Affiliation(s)
- Jun‐Pyo Choi
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - So Young Park
- Department of Internal Medicine Eulji University School of Medicine Seoul Korea
| | - Keun‐Ai Moon
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Eun Hee Ha
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Yeon Duk Woo
- Institute of Allergy and Clinical Immunology Seoul National University Medical Research Center Seoul Korea
| | - Doo Hyun Chung
- Institute of Allergy and Clinical Immunology Seoul National University Medical Research Center Seoul Korea
| | - Hyouk‐Soo Kwon
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Tae‐Bum Kim
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Hae‐Sim Park
- Department of Allergy and Clinical Immunology Ajou University School of Medicine Suwon Korea
| | - Hee‐Bom Moon
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Woo‐Jung Song
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - You Sook Cho
- Division of Allergy and Clinical Immunology Department of Internal Medicine Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| |
Collapse
|
93
|
Paplinska-Goryca M, Misiukiewicz-Stepien P, Nejman-Gryz P, Proboszcz M, Mlacki M, Gorska K, Krenke R. Epithelial-macrophage-dendritic cell interactions impact alarmins expression in asthma and COPD. Clin Immunol 2020; 215:108421. [PMID: 32302698 DOI: 10.1016/j.clim.2020.108421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/05/2020] [Accepted: 04/11/2020] [Indexed: 11/30/2022]
Abstract
In the respiratory system macrophages and dendritic cells collaborate as sentinels against foreign particulate antigens. The study used a triple-cell co-culture model, utilizing nasal epithelial cells, along with: monocyte derived macrophages (moMφs), and monocyte derived DCs (moDCs). Cell cultures from 15 controls, 14 asthma and 11 COPD patients were stimulated with IL-13 and poly I:C for 24 h. Co-cultivation of epithelial cells with moMφs and moDCs increased TSLP level only in asthma and the effect of IL-13 and poly I:C stimulation differed in all groups. Asthma epithelial cells expressed higher level of receptors TSLPR, ST2 and IL-17RA than controls and increased number of ST2 + ciliated and IL17RA + secretory cells. Cytokine expression in respiratory epithelium may be influenced by structural and immunological cell interaction. TSLP pathway may be associated with secretory, while IL-33 with ciliated cells. The impaired function of respiratory epithelium may impact cell-to-cell interactions in asthma.
Collapse
Affiliation(s)
| | | | - Patrycja Nejman-Gryz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland
| | - Małgorzata Proboszcz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland
| | | | - Katarzyna Gorska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland
| | - Rafal Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland
| |
Collapse
|
94
|
Zheng R, Chen Y, Shi J, Wang K, Huang X, Sun Y, Yang Q. Combinatorial IL-17RB, ST2, and TSLPR Signaling in Dendritic Cells of Patients With Allergic Rhinitis. Front Cell Dev Biol 2020; 8:207. [PMID: 32309281 PMCID: PMC7145954 DOI: 10.3389/fcell.2020.00207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Myeloid dendritic cells (DCs) in patients with allergic rhinitis (AR) express higher levels of IL-17RB, ST2, and TSLPR. However, their functional roles in DCs are much less clear. This study aimed to determine the combined effects of these three receptor signals on the T cell-polarizing function of DCs in AR patients. Methods Monocyte-derived DCs (mo-DCs) were generated and stimulated with Toll-like receptor (TLR) 1-9 ligands. Der.p1-induced mo-DCs were stimulated with different combinations of IL-25, IL-33, and TSLP to determine phenotypic characteristics and then co-cultured with CD4+ T cells to assess Th2 cytokine production. Expression levels of IL-17RB, ST2, and TSLPR on myeloid DCs (mDCs) from peripheral blood of AR and healthy subjects were detected to confirm the association of these receptors with disease severity. Results TLR ligands induced AR-derived mo-DCs to increase IL-17RB, ST2, and TSLPR expression by varying degrees; among these, Der.p1 was the strongest inducer. Der.p1-induced mo-DCs from AR showed increased OX40L expression. IL-25, IL-33, and TSLP (alone or in double combination) significantly increased OX40L expression on Der.p1-induced mo-DCs from AR, thereby increasing the production of IL-4, IL-5, and IL-13 in co-cultured CD4+ T cells; triple combination further enhanced these effects. The percentage of IL-17RB+ST2+TSLPR+ mDCs was increased in AR, higher in moderate to severe phase than in mild phase, and positively correlated with the percentages of IL-4+, IL-5+, and IL-13+ T cells. Conclusion A combination of IL-17RB, ST2, and TSLPR signals amplified the Th2-polarizing function of DCs and was associated with disease severity in AR patients.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianbo Shi
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, First People's Hospital of Foshan, Foshan, China
| | - Xuekun Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yueqi Sun
- Department of Otolaryngology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
95
|
Park SH, Jung HJ, Kim TS. IL-33 changes CD25hi Tregs to Th17 cells through a dendritic cell-mediated pathway. Immunol Lett 2020; 218:5-10. [DOI: 10.1016/j.imlet.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/30/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
|
96
|
Fu C, Chen J, Lu J, Yi L, Tong X, Kang L, Pei S, Ouyang Y, Jiang L, Ding Y, Zhao X, Li S, Yang Y, Huang J, Zeng Q. Roles of inflammation factors in melanogenesis (Review). Mol Med Rep 2020; 21:1421-1430. [PMID: 32016458 PMCID: PMC7002987 DOI: 10.3892/mmr.2020.10950] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The occurrence of hyperpigmentation or hypopigmentation after inflammation is a common condition in dermatology and cosmetology. Since the exact mechanism of its occurrence is not yet known, prevention and treatment are troublesome. Previous studies have confirmed that α-melanocyte-stimulating hormone, stem cell factor and other factors can promote melanogenesis-related gene expression through the activation of signaling pathways. Recent studies have revealed that a variety of inflammatory mediators can also participate in the regulation of melanogenesis in melanocytes. In this review, we summarized that interleukin-18, interleukin-33, granulocyte-macrophage colony stimulating factor, interferon-γ, prostaglandin E2 have the effect of promoting melanogenesis, while interleukin-1, interleukin-4, interleukin-6, interleukin-17 and tumor necrosis factor can inhibit melanogenesis. Further studies have found that these inflammatory factors may activate or inhibit melanogenesis-related signaling pathways (such as protein kinase A and mitogen activated protein kinase) by binding to corresponding receptors, thereby promoting or inhibiting the expression of melanogenesis-related genes and regulating skin pigmentation processes. This suggests that the development of drugs or treatment methods from the perspective of regulating inflammation can provide new ideas and new targets for the treatment of pigmented dermatosis. This review outlines the current understanding of the inflammation factors' roles in melanogenesis.
Collapse
Affiliation(s)
- Chuhan Fu
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jianyun Lu
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Lu Yi
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Liyang Kang
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shiyao Pei
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yujie Ouyang
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yufang Ding
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaojiao Zhao
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Si Li
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Yan Yang
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jinhua Huang
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
97
|
Hsu CL, Chhiba KD, Krier-Burris R, Hosakoppal S, Berdnikovs S, Miller ML, Bryce PJ. Allergic inflammation is initiated by IL-33-dependent crosstalk between mast cells and basophils. PLoS One 2020; 15:e0226701. [PMID: 31940364 PMCID: PMC6961911 DOI: 10.1371/journal.pone.0226701] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022] Open
Abstract
IgE-primed mast cells in peripheral tissues, including the skin, lung, and intestine, are key initiators of allergen-triggered edema and inflammation. Particularly in severe forms of allergy, this inflammation becomes strongly neutrophil dominated, and yet how mast cells coordinate this type of response is unknown. We and others have reported that activated mast cells--a hematopoietic cell type--can produce IL-33, a cytokine known to participate in allergic responses but generally considered as being of epithelial origin and driving Type 2 immune responses (e.g., ILC2 and eosinophil activation). Using models of skin anaphylaxis, our data reveal that mast cell-derived IL-33 also initiates neutrophilic inflammation. We demonstrate a cellular crosstalk mechanism whereby activated mast cells crosstalk to IL-33 receptor-bearing basophils, driving these basophils to adopt a unique response signature rich in neutrophil-associated molecules. We further establish that basophil expression of CXCL1 is necessary for IgE-driven neutrophilic inflammation. Our findings thus unearth a new mechanism by which mast cells initiate local inflammation after antigen triggering and might explain the complex inflammatory phenotypes observed in severe allergic diseases. Moreover, our findings (i) establish a functional link from IL-33 to neutrophilic inflammation that extends IL-33-mediated biology well beyond that of Type 2 immunity, and (ii) demonstrate the functional importance of hematopoietic cell-derived IL-33 in allergic pathogenesis.
Collapse
Affiliation(s)
- Chia-Lin Hsu
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Krishan D. Chhiba
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Rebecca Krier-Burris
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Shweta Hosakoppal
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Sergejs Berdnikovs
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Mendy L. Miller
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
| | - Paul J. Bryce
- Division of Allergy-Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
98
|
Pérez-Rodríguez L, Martínez-Blanco M, Lozano-Ojalvo D, Molina E, López-Fandiño R. Egg yolk augments type 2 immunity by activating innate cells. Eur J Nutr 2020; 59:3245-3256. [PMID: 31903504 DOI: 10.1007/s00394-019-02163-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022]
Abstract
PURPOSE Egg yolk (EY) may play a role during the sensitizing phase of egg allergy by exerting intestinal type 2-biasing effects. We aimed to identify the mechanism and role of EY in the induction of allergy to egg white (EW). METHODS BALB/c mice were exposed intragastrically to EW, EY, or the mixture of EW:EY. In addition in vitro experiments were conducted with intestinal epithelial cells (IECs), dendritic cells (DCs), and T cells from naïve mice. Inflammatory and type 2 responses were evaluated. RESULTS Administration of EW upregulated duodenal expression of factors that influence epithelial barrier integrity and function, such as Muc2 and Cldn2, type 2-promoting epithelial cytokines Il33 and Il25, DC genes Irf4 and Tnfsf4, and Th2-cytokines Il14 and Il13. EW:EY further increased the expression of Il25 and Tslp in the duodenum, Il33 and Tslp in the jejunum, and the proportion of lamina propria group 2 innate immune cells (ILC2s) over EW alone. Moreover, it distinctively enhanced the expression of Irf4 and Cd1d1 in the Peyer's patches (PPs), and of Il6, Il33, Gata3, and Il13, both in PPs and mesenteric lymph nodes. In co-cultures of DCs and T cells, EW:EY induced a higher expression of Gata3, Il4, and Il13, secretion of IL-13 and expansion of CD4+ T cells expressing ST2, the IL-33 receptor, than EW or EY added individually. CONCLUSION Co-administration of EY may promote sensitization to EW through activation of innate immune cells, such as IECs, DCs and ILC2s, that are central to the progress of allergies.
Collapse
Affiliation(s)
- Leticia Pérez-Rodríguez
- Instituto de Investigación en Ciencias de La Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Mónica Martínez-Blanco
- Instituto de Investigación en Ciencias de La Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Daniel Lozano-Ojalvo
- Instituto de Investigación en Ciencias de La Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Elena Molina
- Instituto de Investigación en Ciencias de La Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049, Madrid, Spain
| | - Rosina López-Fandiño
- Instituto de Investigación en Ciencias de La Alimentación (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049, Madrid, Spain.
| |
Collapse
|
99
|
Miki H, Tahara-Hanaoka S, Almeida MS, Hitomi K, Shibagaki S, Kanemaru K, Lin YH, Iwata K, Miyake S, Shibayama S, Sumida T, Shibuya K, Shibuya A. Allergin-1 Immunoreceptor Suppresses House Dust Mite-Induced Allergic Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 204:753-762. [PMID: 31900344 DOI: 10.4049/jimmunol.1900180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/02/2019] [Indexed: 01/04/2023]
Abstract
House dust mite (HDM) allergens are leading causes of allergic asthma characterized by Th2 responses. The lung-resident CD11b+ dendritic cells (DCs) play a key role in Th2 cell development in HDM-induced allergic asthma. However, the regulatory mechanism of HDM-induced CD11b+ DC activation remains incompletely understood. In this study, we demonstrate that mice deficient in an inhibitory immunoreceptor, Allergin-1, showed exacerbated HDM-induced airway eosinophilia and serum IgE elevation. By using bone marrow-chimeric mice that were sensitized with adoptively transferred HDM-stimulated wild-type or Allergin-1-deficient CD11b+ bone marrow-derived cultured DCs (BMDCs), followed by challenge with HDM, we show that Allergin-1 on the BMDCs suppressed HDM-induced allergic airway inflammation. We also show that Allergin-1 suppressed HDM-induced PGE2 production from CD11b+ BMDCs by inhibiting Syk tyrosine kinase activation through recruitment of SHP-1, subsequently leading to negative regulation of Th2 responses. These results suggest that Allergin-1 plays an important role in regulation of HDM-induced allergic airway inflammation.
Collapse
Affiliation(s)
- Haruka Miki
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Department of Internal Medicine, Faculty of Medicine, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; .,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Mariana Silva Almeida
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kaori Hitomi
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shohei Shibagaki
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazumasa Kanemaru
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yu-Hsien Lin
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; and
| | - Kanako Iwata
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shota Miyake
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shiro Shibayama
- Research Center of Immunology, Tsukuba Institute, Ono Pharmaceutical Co., Ltd., Tsukuba, Ibaraki 300-4247, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; .,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
100
|
Hasan A, Kochumon S, Al-Ozairi E, Tuomilehto J, Al-Mulla F, Ahmad R. Correlation Profile of Suppression of Tumorigenicity 2 and/or Interleukin-33 with Biomarkers in the Adipose Tissue of Individuals with Different Metabolic States. Diabetes Metab Syndr Obes 2020; 13:3839-3859. [PMID: 33116731 PMCID: PMC7586022 DOI: 10.2147/dmso.s251978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/01/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The suppression of tumorigenicity 2 (ST2) has two main splice variants including a membrane bound (ST2) form, which activates the myeloid differentiation primary response 88 (MyD88)/nuclear factor-kappa B (NF-κB) signaling pathway, and a secreted soluble form (sST2), which acts as a decoy receptor for ST2 ligand, interleukin (IL)-33. The IL-33/ST2 axis is protective against obesity, insulin resistance, and type 2 diabetes (T2D). In humans, adipose tissue IL-33 displays distinct correlation profiles with glycated hemoglobin, ST2, and other immunometabolic mediators, depending on the glycemic health of the individuals. We determined whether adipose tissue ST2 displays distinct correlation profiles with immunometabolic mediators and whether ST2 and/or IL-33 are correlated with intracellular signaling molecules. PATIENTS AND METHODS A total of 91 adults with normal glycemia, prediabetes, and T2D were included. After measuring their anthropometric and biochemical parameters, subcutaneous adipose tissues were isolated and mRNA expression of biomarkers was measured. RESULTS In individuals with normal glycemia, adipose tissue ST2 was directly correlated with chemokine (C-C motif) ligand (CCL)-2, CCL5, IL-12, fibrinogen-like protein 2 (FGL2) and interferon regulatory factor (IRF)-4, but inversely correlated with cytochrome C oxidase subunit 7A1. IL-33 and ST2 were directly correlated with tumor necrosis factor receptor-associated factor 6 (TRAF6), NF-κB, and nuclear factor of activated T-cells 5 (NFAT5). In individuals with prediabetes, ST2 was inversely correlated with IL-5, whereas IL-33 but not ST2 was directly correlated with MyD88 and NF-κB. In individuals with T2D, ST2 was directly correlated with CCL2, IL-1β, and IRF5. IL-33 and ST2 were directly correlated with MyD88, TRAF6, and NF-κB. CONCLUSION Adipose tissue ST2 and IL-33 show different correlation profiles with various immunometabolic biomarkers depending on the metabolic state of the individuals. Therefore, targeting the IL-33/ST2 axis might form the basis for novel therapies to combat metabolic disorders.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Kuwait City, Kuwait
- Correspondence: Amal Hasan Email
| | - Shihab Kochumon
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Medical Division, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Medicine, Faculty of Medicine, Kuwait City, Kuwait
| | - Jaakko Tuomilehto
- Research Division, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Public Health, University of Helsinki, Helsinki, Finland
- National School of Public Health, Madrid, Spain
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Functional Genomics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|