1
|
Liang C, Shen Y, Xu Y, Liang Y, Qiu S, Tang H, Zhong X. Dendritic Cells Promote the Differentiation of ILCs into NCR -ILC3s in the Lungs of Mice Exposed to Cigarette Smoke. COPD 2024; 21:2389909. [PMID: 39143749 DOI: 10.1080/15412555.2024.2389909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/07/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
The involvement of Group 3 innate lymphoid cells (ILC3s) and dendritic cells (DCs) in chronic lung inflammation has been increasingly regarded as the key to understand the inflammatory mechanisms of smoke-related chronic obstructive pulmonary disease (COPD). However, the mechanism underlying the engagement of both remains unclear. Our study aimed to explore NCR-ILC3 differentiation in the lungs of mice exposed to cigarette smoke (CS) and to further investigate whether DCs activated by CS exposure contribute to the differentiation of ILCs into NCR-ILC3s. The study involved both in vivo and in vitro experiments. In the former, the frequencies of lung NCR-ILC3s and NKp46-IL-17A+ ILCs and the expression of DCs, CD40, CD86, IL-23, and IL-1β quantified by flow cytometry were compared between CS-exposed mice and air-exposed mice. In the latter, NKp46-IL-17A+ ILC frequencies quantified by flow cytometry were compared after two cocultures, one involving lung CD45+Lin-CD127+ ILCs sorted from air-exposed mice and DCs sifted by CD11c magnetic beads from CS-exposed mice and another including identical CD45+Lin-CD127+ ILCs and DCs from air-exposed mice. The results indicated significant increases in the frequencies of NCR-ILC3s and NKp46-IL-17A+ ILCs; in the expression of DCs, CD40, CD86, IL-23, and IL-1β in CS-exposed mice; and in the frequency of NKp46-IL-17A+ ILCs after the coculture with DCs from CS-exposed mice. In conclusion, CS exposure increases the frequency of lung ILCs and NCR-ILC3s. CS-induced DC activation enhances the differentiation of ILCs into NCR-ILC3s, which likely acts as a mediating step in the involvement of NCR-ILC3s in chronic lung inflammation.
Collapse
Affiliation(s)
- Caixia Liang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Ying Shen
- General Practice School, Guangxi Medical University, Nanning, P.R. China
| | - Yifang Xu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Yi Liang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Shilin Qiu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Haijuan Tang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Xiaoning Zhong
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| |
Collapse
|
2
|
Roy-Dorval A, Deagle RC, Roth F, Raybaud M, Ismailova N, Krisna SS, Aboud DGK, Stegen C, Leconte J, Berberi G, Esomojumi A, Fritz JH. Analysis of lipid uptake, storage, and fatty acid oxidation by group 2 innate lymphoid cells. Front Immunol 2024; 15:1493848. [PMID: 39497825 PMCID: PMC11532145 DOI: 10.3389/fimmu.2024.1493848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 11/07/2024] Open
Abstract
Group 2 Innate Lymphoid Cells (ILC2) are critical drivers of both innate and adaptive type 2 immune responses, known to orchestrate processes involved in tissue restoration and wound healing. In addition, ILC2 have been implicated in chronic inflammatory barrier disorders in type 2 immunopathologies such as allergic rhinitis and asthma. ILC2 in the context of allergen-driven airway inflammation have recently been shown to influence local and systemic metabolism, as well as being rich in lipid-storing organelles called lipid droplets. However, mechanisms of ILC2 lipid anabolism and catabolism remain largely unknown and the impact of these metabolic processes in regulating ILC2 phenotypes and effector functions has not been extensively characterized. ILC2 phenotypes and effector functions are shaped by their metabolic status, and determining the metabolic requirements of ILC2 is critical in understanding their role in type 2 immune responses and their associated pathophysiology. We detail here a novel experimental method of implementing flow cytometry for large scale analysis of fatty acid uptake, storage of neutral lipids, and fatty acid oxidation in primary murine ILC2 with complementary morphological analysis of lipid storage using confocal microscopy. By combining flow cytometry and confocal microscopy, we can identify the metabolic lipid requirements for ILC2 functions as well as characterize the phenotype of lipid storage in ILC2. Linking lipid metabolism pathways to ILC2 phenotypes and effector functions is critical for the assessment of novel pharmaceutical strategies to regulate ILC2 functions in type 2 immunopathologies.
Collapse
Affiliation(s)
- Audrey Roy-Dorval
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Rebecca C. Deagle
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Frederik Roth
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Mathilde Raybaud
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Nailya Ismailova
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Sai Sakktee Krisna
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Damon G. K. Aboud
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Camille Stegen
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Julien Leconte
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Gabriel Berberi
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Ademola Esomojumi
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Jörg H. Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| |
Collapse
|
3
|
Yoon J, Lee J, Park A, Yoon J, Kim JR, Moon GJ, Yu J. Type 2 Innate Lymphoid Cells and Skin Fibrosis in a Murine Model of Atopic Dermatitis-Like Skin Inflammation. J Korean Med Sci 2024; 39:e221. [PMID: 39106888 PMCID: PMC11301010 DOI: 10.3346/jkms.2024.39.e221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/24/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disease. Although murine studies have demonstrated that type 2 innate lymphoid cells (ILCs) mediate type 2 skin inflammation, their role in skin fibrosis in AD remains unclear. This study investigated whether type 2 ILCs are involved in skin fibrosis using an AD-like murine model. METHODS C57BL/6 mice were treated epicutaneously with Aspergillus fumigatus (Af) for 5 consecutive days per week for 5 weeks to induce skin fibrosis. Mature lymphocyte deficient Rag1-/- mice were also used to investigate the role of type 2 ILCs in skin fibrosis. RESULTS The clinical score and transepidermal water loss (TEWL) were significantly higher in the AD group than in the control group. The AD group also showed significantly increased epidermal and dermal thicknesses and significantly higher numbers of eosinophils, neutrophils, mast cells, and lymphocytes in the lesional skin than the control group. The lesional skin of the AD group showed increased stain of collagen and significantly higher levels of collagen than the control group (10.4 ± 2.2 µg/mg vs. 1.6 ± 0.1 µg/mg, P < 0.05). The AD group showed significantly higher populations of type 2 ILCs in the lesional skin compared to the control group (0.08 ± 0.01% vs. 0.03 ± 0.01%, P < 0.05). These findings were also similar with the AD group of Rag1-/- mice compared to their control group. Depletion of type 2 ILCs with anti-CD90.2 monoclonal antibodies significantly improved clinical symptom score, TEWL, and infiltration of inflammatory cells, and significantly decreased levels of collagen were observed in the AD group of Rag1-/- mice (1.6 ± 0.0 μg/mg vs. 4.5 ± 0.3 μg/mg, P < 0.001). CONCLUSION In the Af-induced AD-like murine model, type 2 ILCs were elevated, with increased levels of collagen. Additionally, removal of type 2 ILCs resulted in decreased collagen levels and improved AD-like pathological findings. These findings suggest that type 2 ILCs play a role in the mechanism of skin fibrosis in AD.
Collapse
Affiliation(s)
- Jisun Yoon
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul, Korea
- Clinical Trial Support Team, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea
| | - Jiho Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Arum Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Jin Yoon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Jeong Ryun Kim
- Department of Cell and Genetic Engineering, University of Ulsan College of Medicine, Seoul, Korea
| | - Gyeong Joon Moon
- Department of Cell and Genetic Engineering, University of Ulsan College of Medicine, Seoul, Korea
- Center for Cell Therapy, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea.
| | - Jinho Yu
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
4
|
Miyamoto S, Takayama Y, Kondo T, Maruyama K. Senso-immunology: the hidden relationship between sensory system and immune system. J Bone Miner Metab 2024; 42:413-420. [PMID: 39060499 DOI: 10.1007/s00774-024-01538-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
The primary sensory neurons involved in pain perception express various types of receptor-type ion channels at their nerve endings. These molecules are responsible for triggering neuronal excitation, translating environmental stimuli into pain signals. Recent studies have shown that acute nociception, induced by neuronal excitation, not only serves as a sensor for signaling life-threatening situations but also modulates our pathophysiological conditions. This modulation occurs through the release of neuropeptides by primary sensory neurons excited by nociceptive stimuli, which directly or indirectly affect peripheral systems, including immune function. Senso-immunology, an emerging research field, integrates interdisciplinary studies of pain and immunology and has garnered increasing attention in recent years. This review provides an overview of the systemic pathophysiological functions regulated by receptor-type ion channels, such as transient receptor potential (TRP) channels in primary sensory neurons, from the perspective of senso-immunology.
Collapse
Affiliation(s)
- Satoshi Miyamoto
- Department of Pharmacology, Aichi Medical University School of Medicine, Aichi, 480-1195, Japan
| | - Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan.
| | - Takeshi Kondo
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Ibaraki, 305-8565, Japan
| | - Kenta Maruyama
- Department of Pharmacology, Aichi Medical University School of Medicine, Aichi, 480-1195, Japan.
| |
Collapse
|
5
|
Lahire S, Fichel C, Rubaszewski O, Lerévérend C, Audonnet S, Visneux V, Perotin JM, Deslée G, Le Jan S, Potteaux S, Le Naour R, Pommier A. Elastin-derived peptides favor type 2 innate lymphoid cells in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2024; 326:L812-L820. [PMID: 38712445 DOI: 10.1152/ajplung.00306.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a condition characterized by chronic airway inflammation and obstruction, primarily caused by tobacco smoking. Although the involvement of immune cells in COPD pathogenesis is well established, the contribution of innate lymphoid cells (ILCs) remains poorly understood. ILCs are a type of innate immune cells that participate in tissue remodeling processes, but their specific role in COPD has not been fully elucidated. During COPD, the breakdown of pulmonary elastin generates elastin peptides that elicit biological activities on immune cells. This study aimed to investigate the presence of ILC in patients with COPD and examine the impact of elastin peptides on their functionality. Our findings revealed an elevated proportion of ILC2 in the peripheral blood of patients with COPD, and a general activation of ILC as indicated by an increase in their cytokine secretion capacity. Notably, our study demonstrated that serum from patients with COPD promotes ILC2 phenotype, likely due to the elevated concentration of IL-5, a cytokine known to favor ILC2 activation. Furthermore, we uncovered that this increase in IL-5 secretion is partially attributed to its secretion by macrophages upon stimulation by elastin peptides, suggesting an indirect role of elastin peptides on ILC in COPD. These findings shed light on the involvement of ILC in COPD and provide insights into the potential interplay between elastin breakdown, immune cells, and disease progression. Further understanding of the mechanisms underlying ILC activation and their interaction with elastin peptides could contribute to the development of novel therapeutic strategies for COPD management.NEW & NOTEWORTHY Elastin-derived peptides, generated following alveolar degradation during emphysema in patients with COPD, are able to influence the response of type 2 innate lymphoid cells. We show that the orientation of innate lymphoid cells in patients with COPD is shifted toward a type 2 profile and that elastin peptides are indirectly participating in that shift through their influence of macrophages, which in turn impact innate lymphoid cells.
Collapse
Affiliation(s)
- Sarah Lahire
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Caroline Fichel
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Océane Rubaszewski
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Cédric Lerévérend
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
| | - Sandra Audonnet
- Université de Reims Champagne Ardenne, Plateforme de cytométrie en flux, URCACyt, Reims, France
| | - Vincent Visneux
- CHU de Reims, Service des maladies respiratoires, Reims, France
| | - Jeanne-Marie Perotin
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Gaëtan Deslée
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Sébastien Le Jan
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Stéphane Potteaux
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
- Délégation régionale Inserm Paris Ile-de France Centre Nord, Paris, France
| | - Richard Le Naour
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Arnaud Pommier
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| |
Collapse
|
6
|
Tang M, Da X, Xu Z, Zhao X, Zhou H. UHPLC/MS-based metabolomics of asthmatic mice reveals metabolic changes in group 2 innate lymphoid cells. Int Immunopharmacol 2024; 130:111775. [PMID: 38430805 DOI: 10.1016/j.intimp.2024.111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Helper Th2-type immune responses are essential in allergic airway diseases, including asthma and allergic rhinitis. Recent studies have indicated that group 2 innate lymphoid cells (ILC2s) play a crucial role in the occurrence and development of asthma. However, the metabolic profile of ILC2s and their regulatory mechanisms in asthma remain unclear. Therefore, we established two asthma mouse models: an ovalbumin (OVA)-induced asthma model and an IL-33-induced asthma model. We then used ultra-high-performance liquid chromatography/mass spectrometry (UHPLC/MS) to conduct high-throughput untargeted metabolic analysis of ILC2s in the lung tissues of the asthma models. The identified metabolites primarily consisted of lipids, lipid-like molecules, benzene, organic acids, derivatives, and organic oxidation compounds. Specifically, 34 differentially accumulated metabolites influenced the metabolic profiles of the control and OVA-induced asthma model groups. Moreover, the accumulation of 39 metabolites significantly differed between the Interleukin 33 (IL-33) and control groups. These differentially accumulated metabolites were mainly involved in pathways such as sphingolipid, oxidative phosphorylation, and fatty acid metabolism. This metabolomic study revealed, for the first time, the key metabolites and metabolic pathways of ILC2s, revealing new aspects of cellular metabolism in the context of airway inflammation. These findings not only contribute to unraveling the pathogenesis of asthma but also provide a crucial theoretical foundation for the future development of therapeutic strategies targeting ILC2s.
Collapse
Affiliation(s)
- Min Tang
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Xianzong Da
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China
| | - Zhiwei Xu
- Department of Pediatrics, Bengbu Medical College, Bengbu, China
| | - Xiaoman Zhao
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Haoquan Zhou
- Department of Pediatrics, Provincial Hospital affiliated to Anhui Medical University, Hefei, China; Department of Pediatrics, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
7
|
Yoshinaga M, Takeuchi O. Regulation of inflammatory diseases via the control of mRNA decay. Inflamm Regen 2024; 44:14. [PMID: 38491500 PMCID: PMC10941436 DOI: 10.1186/s41232-024-00326-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
Inflammation orchestrates a finely balanced process crucial for microorganism elimination and tissue injury protection. A multitude of immune and non-immune cells, alongside various proinflammatory cytokines and chemokines, collectively regulate this response. Central to this regulation is post-transcriptional control, governing gene expression at the mRNA level. RNA-binding proteins such as tristetraprolin, Roquin, and the Regnase family, along with RNA modifications, intricately dictate the mRNA decay of pivotal mediators and regulators in the inflammatory response. Dysregulated activity of these factors has been implicated in numerous human inflammatory diseases, underscoring the significance of post-transcriptional regulation. The increasing focus on targeting these mechanisms presents a promising therapeutic strategy for inflammatory and autoimmune diseases. This review offers an extensive overview of post-transcriptional regulation mechanisms during inflammatory responses, delving into recent advancements, their implications in human diseases, and the strides made in therapeutic exploitation.
Collapse
Affiliation(s)
- Masanori Yoshinaga
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan.
| | - Osamu Takeuchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
8
|
Jutel M, Agache I, Zemelka-Wiacek M, Akdis M, Chivato T, Del Giacco S, Gajdanowicz P, Gracia IE, Klimek L, Lauerma A, Ollert M, O'Mahony L, Schwarze J, Shamji MH, Skypala I, Palomares O, Pfaar O, Torres MJ, Bernstein JA, Cruz AA, Durham SR, Galli SJ, Gómez RM, Guttman-Yassky E, Haahtela T, Holgate ST, Izuhara K, Kabashima K, Larenas-Linnemann DE, von Mutius E, Nadeau KC, Pawankar R, Platts-Mills TAE, Sicherer SH, Park HS, Vieths S, Wong G, Zhang L, Bilò MB, Akdis CA. Nomenclature of allergic diseases and hypersensitivity reactions: Adapted to modern needs: An EAACI position paper. Allergy 2023; 78:2851-2874. [PMID: 37814905 DOI: 10.1111/all.15889] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/11/2023]
Abstract
The exponential growth of precision diagnostic tools, including omic technologies, molecular diagnostics, sophisticated genetic and epigenetic editing, imaging and nano-technologies and patient access to extensive health care, has resulted in vast amounts of unbiased data enabling in-depth disease characterization. New disease endotypes have been identified for various allergic diseases and triggered the gradual transition from a disease description focused on symptoms to identifying biomarkers and intricate pathogenetic and metabolic pathways. Consequently, the current disease taxonomy has to be revised for better categorization. This European Academy of Allergy and Clinical Immunology Position Paper responds to this challenge and provides a modern nomenclature for allergic diseases, which respects the earlier classifications back to the early 20th century. Hypersensitivity reactions originally described by Gell and Coombs have been extended into nine different types comprising antibody- (I-III), cell-mediated (IVa-c), tissue-driven mechanisms (V-VI) and direct response to chemicals (VII). Types I-III are linked to classical and newly described clinical conditions. Type IVa-c are specified and detailed according to the current understanding of T1, T2 and T3 responses. Types V-VI involve epithelial barrier defects and metabolic-induced immune dysregulation, while direct cellular and inflammatory responses to chemicals are covered in type VII. It is notable that several combinations of mixed types may appear in the clinical setting. The clinical relevance of the current approach for allergy practice will be conferred in another article that will follow this year, aiming at showing the relevance in clinical practice where various endotypes can overlap and evolve over the lifetime.
Collapse
Affiliation(s)
- Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania
| | | | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Tomás Chivato
- School of Medicine, University CEU San Pablo, Madrid, Spain
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Allergy and Clinical Immunology, University Hospital "Duilio Casula", Monserrato, Italy
| | - Pawel Gajdanowicz
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
| | - Ibon Eguiluz Gracia
- Allergy Unit, UMA-Regional University Hospital of Malaga, IBIMA-BIONAND, Malaga, Spain
| | - Ludger Klimek
- Department of Otolaryngology, Head and Neck Surgery, Universitätsmedizin Mainz, Mainz, Germany
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Centre, Odense University Hospital, Odense Research Center for Anaphylaxis (ORCA), Odense, Denmark
| | - Liam O'Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland, National University of Ireland, Cork, Ireland
| | - Jürgen Schwarze
- Child Life and Health, Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, London, UK
| | - Isabel Skypala
- Department of Inflammation and Repair, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Part of Guys and St Thomas' NHS Foundation Trust, London, UK
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Maria Jose Torres
- Allergy Unit, UMA-Regional University Hospital of Malaga, IBIMA-BIONAND, Malaga, Spain
| | - Jonathan A Bernstein
- Department of Internal Medicine, Division of Rheumatology, Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alvaro A Cruz
- Fundaçao ProAR, Federal University of Bahia and GARD/WHO Planning Group, Salvador, Bahia, Brazil
| | - Stephen R Durham
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Stephen J Galli
- Department of Pathology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | | | - Emma Guttman-Yassky
- Department of Dermatology and the Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Stephen T Holgate
- Academic Unit of Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Kenji Izuhara
- Department of Biomolecular Sciences, Division of Medical Biochemistry, Saga Medical School, Saga, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Désirée E Larenas-Linnemann
- Center of Excellence in Asthma and Allergy, Médica Sur Clinical Foundation and Hospital, Mexico City, Mexico
| | - Erica von Mutius
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
- Institute of Asthma and Allergy Prevention, Helmholtz Centre Munich, Munich, Germany
- German Center for Lung Research (DZL), Giesen, Germany
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ruby Pawankar
- Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Tomas A E Platts-Mills
- Department of Medicine, Division of Allergy and Clinical Immunology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott H Sicherer
- Division of Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | | | - Gary Wong
- Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - M Beatrice Bilò
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona and Allergy Unit, Department of Internal Medicine, University Hospital of Marche, Ancona, Italy
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
9
|
Miyamoto S, Kondo T, Maruyama K. Senso-immunology: the past, present, and future. J Biochem 2023; 174:305-315. [PMID: 37461198 DOI: 10.1093/jb/mvad052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 09/29/2023] Open
Abstract
Pain and mechanical stimulation are thought to be alarm systems that alert the brain to physical abnormalities. When we experience unpleasant feelings in infected or traumatized tissues, our awareness is directed to the afflicted region, prompting activities such as resting or licking the tissue. Despite extensive research into the molecular biology of nociceptors, it was unclear whether their role was limited to the generation and transmission of unpleasant feelings or whether they actively modulate the pathogenesis of infected or traumatized tissues. Recently, it has become clear how the sensory and immune systems interact with one another and share similar receptors and ligands to modify the pathogenesis of various diseases. In this paper, we summarize the mechanisms of crosstalk between the sensory and immune systems and the impact of this new interdisciplinary field, which should be dubbed 'senso-immunology,' on medical science.
Collapse
Affiliation(s)
- Satoshi Miyamoto
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 3N7, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Kondo
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido 060-8636, Japan
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8787, Japan
| |
Collapse
|
10
|
Srinivasan A, Giri A, Duraisamy SK, Alsup A, Castro M, Sundar IK. Chronic HDM exposure shows time-of-day and sex-based differences in inflammatory response associated with lung circadian clock disruption. iScience 2023; 26:107580. [PMID: 37664635 PMCID: PMC10470299 DOI: 10.1016/j.isci.2023.107580] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 07/13/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Circadian rhythms and sex differences are involved in the pathophysiology of asthma. Yet, there are no reports that simultaneously address the role of the circadian clock and sex-based differences in chronic house dust mite (HDM)-induced asthma. Here, we sought to determine if chronic HDM exposure during the resting phase (zeitgeber time: ZT0/6:00 a.m.) versus the active phase (ZT12/6:00 p.m.) differentially affects the circadian clock and alters asthma pathobiology in female and male mice. HDM exposure at ZT12 exaggerated infiltration of eosinophil subtypes and associated chemokines in females compared to males. Furthermore, HDM exposure augmented eosinophil chemokines, Th2 gene expression and cytokine release, and humoral immune response in females compared to males at ZT12. Concurrently, histopathological evaluation confirmed increased airway inflammation at ZT12 in both females and males. Overall, we showed a time-of-day response and sex-based differences in HDM-induced exaggerated asthmatic phenotypes (inflammation/remodeling) and circadian clock disruption in females compared to males.
Collapse
Affiliation(s)
- Ashokkumar Srinivasan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Allan Giri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Santhosh Kumar Duraisamy
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Alexander Alsup
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Isaac Kirubakaran Sundar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
11
|
Sudduth ER, Trautmann-Rodriguez M, Gill N, Bomb K, Fromen CA. Aerosol pulmonary immune engineering. Adv Drug Deliv Rev 2023; 199:114831. [PMID: 37100206 PMCID: PMC10527166 DOI: 10.1016/j.addr.2023.114831] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/23/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
Aerosolization of immunotherapies poses incredible potential for manipulating the local mucosal-specific microenvironment, engaging specialized pulmonary cellular defenders, and accessing mucosal associated lymphoid tissue to redirect systemic adaptive and memory responses. In this review, we breakdown key inhalable immunoengineering strategies for chronic, genetic, and infection-based inflammatory pulmonary disorders, encompassing the historic use of immunomodulatory agents, the transition to biological inspired or derived treatments, and novel approaches of complexing these materials into drug delivery vehicles for enhanced release outcomes. Alongside a brief description of key immune targets, fundamentals of aerosol drug delivery, and preclinical pulmonary models for immune response, we survey recent advances of inhaled immunotherapy platforms, ranging from small molecules and biologics to particulates and cell therapies, as well as prophylactic vaccines. In each section, we address the formulation design constraints for aerosol delivery as well as advantages for each platform in driving desirable immune modifications. Finally, prospects of clinical translation and outlook for inhaled immune engineering are discussed.
Collapse
Affiliation(s)
- Emma R Sudduth
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - Nicole Gill
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kartik Bomb
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
12
|
Chen L, Sun R, Lei C, Xu Z, Song Y, Deng Z. Alcohol-mediated susceptibility to lung fibrosis is associated with group 2 innate lymphoid cells in mice. Front Immunol 2023; 14:1178498. [PMID: 37457733 PMCID: PMC10343460 DOI: 10.3389/fimmu.2023.1178498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Chronic alcohol ingestion promotes acute lung injury and impairs immune function. However, the mechanisms involved are incompletely understood. Here, we show that alcohol feeding enhances bleomycin-induced lung fibrosis and inflammation via the regulation of type 2 innate immune responses, especially by group 2 innate lymphoid cells (ILC2s). Neuroimmune interactions have emerged as critical modulators of lung inflammation. We found alcohol consumption induced the accumulation of ILC2 and reduced the production of the neuropeptide calcitonin gene-related peptide (CGRP), primarily released from sensory nerves and pulmonary neuroendocrine cells (PNECs). CGRP potently suppressed alcohol-driven type 2 cytokine signals in vivo. Vagal ganglia TRPV1+ afferents mediated immunosuppression occurs through the release of CGRP. Inactivation of the TRPV1 receptor enhanced bleomycin-induced fibrosis. In addition, mice lacking the CGRP receptor had the increased lung inflammation and fibrosis and type 2 cytokine production as well as exaggerated responses to alcohol feeding. Together, these data indicate that alcohol consumption regulates the interaction of CGRP and ILC2, which is a critical contributor of lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Liang Chen
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No. 1 People’s Hospital, Nanjing Medical University, Huai’an, Jiangsu, China
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Rui Sun
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Chao Lei
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Zhishan Xu
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Zhongbin Deng
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, United States
- Brown Cancer Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
13
|
Sedney CJ, Harvill ET. The Neonatal Immune System and Respiratory Pathogens. Microorganisms 2023; 11:1597. [PMID: 37375099 PMCID: PMC10301501 DOI: 10.3390/microorganisms11061597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Neonates are more susceptible to some pathogens, particularly those that cause infection in the respiratory tract. This is often attributed to an incompletely developed immune system, but recent work demonstrates effective neonatal immune responses to some infection. The emerging view is that neonates have a distinctly different immune response that is well-adapted to deal with unique immunological challenges of the transition from a relatively sterile uterus to a microbe-rich world, tending to suppress potentially dangerous inflammatory responses. Problematically, few animal models allow a mechanistic examination of the roles and effects of various immune functions in this critical transition period. This limits our understanding of neonatal immunity, and therefore our ability to rationally design and develop vaccines and therapeutics to best protect newborns. This review summarizes what is known of the neonatal immune system, focusing on protection against respiratory pathogens and describes challenges of various animal models. Highlighting recent advances in the mouse model, we identify knowledge gaps to be addressed.
Collapse
Affiliation(s)
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
14
|
Ryu S, Kim HY. Bone Marrow Progenitors and IL-2 Signaling Contribute to the Strain Differences of Kidney Innate Lymphoid Cells. Immune Netw 2023; 23:e15. [PMID: 37179753 PMCID: PMC10166654 DOI: 10.4110/in.2023.23.e15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/24/2022] [Accepted: 12/21/2022] [Indexed: 05/15/2023] Open
Abstract
Innate lymphoid cells (ILCs) are critical immune-response mediators. Although they largely reside in mucosal tissues, the kidney also bears substantial numbers. Nevertheless, kidney ILC biology is poorly understood. BALB/c and C57BL/6 mice are known to display type-2 and type-1 skewed immune responses, respectively, but it is unclear whether this extends to ILCs. We show here that indeed, BALB/c mice have higher total ILCs in the kidney than C57BL/6 mice. This difference was particularly pronounced for ILC2s. We then showed that three factors contributed to the higher ILC2s in the BALB/c kidney. First, BALB/c mice demonstrated higher numbers of ILC precursors in the bone marrow. Second, transcriptome analysis showed that compared to C57BL/6 kidneys, the BALB/c kidneys associated with significantly higher IL-2 responses. Quantitative RT-PCR also showed that compared to C57BL/6 kidneys, the BALB/c kidneys expressed higher levels of IL-2 and other cytokines known to promote ILC2 proliferation and/or survival (IL-7, IL-33, and thymic stromal lymphopoietin). Third, the BALB/c kidney ILC2s may be more sensitive to the environmental signals than C57BL/6 kidney ILC2s since they expressed their transcription factor GATA-3 and the IL-2, IL-7, and IL-25 receptors at higher levels. Indeed, they also demonstrated greater responsiveness to IL-2 than C57BL/6 kidney ILC2s, as shown by their greater STAT5 phosphorylation levels after culture with IL-2. Thus, this study demonstrates previously unknown properties of kidney ILC2s. It also shows the impact of mouse strain background on ILC2 behavior, which should be considered when conducting research on immune diseases with experimental mouse models.
Collapse
Affiliation(s)
- Seungwon Ryu
- Department of Microbiology, Gachon University College of Medicine, Incheon 21999, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
15
|
Airway epithelial ITGB4 deficiency induces airway remodeling in a mouse model. J Allergy Clin Immunol 2023; 151:431-446.e16. [PMID: 36243221 DOI: 10.1016/j.jaci.2022.09.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Airway epithelial cells (AECs) with impaired barrier function contribute to airway remodeling through the activation of epithelial-mesenchymal trophic units (EMTUs). Although the decreased expression of ITGB4 in AECs is implicated in the pathogenesis of asthma, how ITGB4 deficiency impacts airway remodeling remains obscure. OBJECTIVE This study aims to determine the effect of epithelial ITGB4 deficiency on the barrier function of AECs, asthma susceptibility, airway remodeling, and EMTU activation. METHODS AEC-specific ITGB4 conditional knockout mice (ITGB4-/-) were generated and an asthma model was employed by the sensitization and challenge of house dust mite (HDM). EMTU activation-related growth factors were examined in ITGB4-silenced primary human bronchial epithelial cells of healthy subjects after HDM stimulation. Dexamethasone, the inhibitors of JNK phosphorylation or FGF2 were administered for the identification of the molecular mechanisms of airway remodeling in HDM-exposed ITGB4-/- mice. RESULTS ITGB4 deficiency in AECs enhanced asthma susceptibility and airway remodeling by disrupting airway epithelial barrier function. Aggravated airway remodeling in HDM-exposed ITGB4-/- mice was induced through the enhanced activation of EMTU mediated by Src homology domain 2-containing protein tyrosine phosphatase 2/c-Jun N-terminal kinase/Jun N-terminal kinase-dependent transcription factor/FGF2 (SHP2/JNK/c-Jun/FGF2) signaling pathway, which was partially independent of airway inflammation. Both JNK and FGF2 inhibitors significantly inhibited the aggravated airway remodeling and EMTU activation in HDM-exposed ITGB4-/- mice. CONCLUSIONS Airway epithelial ITGB4 deficiency induces airway remodeling in a mouse model of asthma through enhanced EMTU activation that is regulated by the SHP2/JNK/c-Jun/FGF2 pathway.
Collapse
|
16
|
Deng Y, Shi S, Luo J, Zhang Y, Dong H, Wang X, Zhou J, Wei Z, Li J, Xu C, Xu S, Sun Y, Ni B, Wu Y, Yang D, Han C, Tian Y. Regulation of mRNA stability contributes to the function of innate lymphoid cells in various diseases. Front Immunol 2023; 14:1118483. [PMID: 36776864 PMCID: PMC9909350 DOI: 10.3389/fimmu.2023.1118483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Innate lymphoid cells (ILCs) are important subsets of innate immune cells that regulate mucosal immunity. ILCs include natural killer cells, innate lymphoid cells-1 (ILC1s), ILC2s, and ILC3s, which have extremely important roles in the immune system. In this review, we summarize the regulation of mRNA stability mediated through various factors in ILCs (e.g., cytokines, RNA-binding proteins, non-coding RNAs) and their roles in mediating functions in different ILC subsets. In addition, we discuss potential therapeutic targets for diseases such as chronic obstructive pulmonary disease, cancer, and pulmonary fibrosis by regulation of mRNA stability in ILCs, which may provide novel directions for future clinical research.
Collapse
Affiliation(s)
- Yuanyu Deng
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Saiyu Shi
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiwei Zhang
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Dong
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xian Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Jian Zhou
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyuan Wei
- The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiahui Li
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chen Xu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuai Xu
- Department of Stomatology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Sun
- The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Di Yang
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| | - Chao Han
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| | - Yi Tian
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| |
Collapse
|
17
|
Bickel J, Bermudez LE. Induction of killing of Mycobacterium avium subsp. hominissuis in macrophages by cytokine stimulated innate-like lymphoid cells is negatively affected by the pathogen. INTERNATIONAL MICROBIOLOGY : THE OFFICIAL JOURNAL OF THE SPANISH SOCIETY FOR MICROBIOLOGY 2023:10.1007/s10123-023-00326-4. [PMID: 36662342 PMCID: PMC10397143 DOI: 10.1007/s10123-023-00326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
Mycobacterium avium subsp. hominissuis (MAH) is a common environmental bacterium that causes infection in immunocompromised patients such as those with HIV/AIDS, or patients with chronic lung disease such as cystic fibrosis. There are many strains of MAH with varying levels of virulence. Infection with MAH strains 100 and 104 has been associated with different immune responses in mice and outcome of the disease. While MAH 100 infection tends to be cleared from mice, MAH 104 is virulent and grows in host tissue. What is currently unknown are the mechanisms related to this difference in host defense and virulence. Our hypothesis is that differences in circulating innate lymphocytes response are associated with increased protection from infection. Innate lymphoid cells (ILC) are lymphoid cells with an important role in regulation of innate immune systems. ILCs can be categorized into three subpopulations ILC1, ILC2, and ILC3 based on their cytokine production and regulatory transcription factors. Investigation was carried out on how macrophage anti-MAH response change depending on activation by primary mouse lymphocytes activated with IL-12, IL-33, and IL-23, triggering differentiation into ILC-like subpopulations. Our results do not affirm the role of any one ILC subpopulation in macrophage anti-M. avium ability. Our findings instead support the conclusion that MAH infection of macrophages suppresses the stimulatory function of ILCs.
Collapse
Affiliation(s)
- Jay Bickel
- Department of Biomedical Science, College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Luiz E Bermudez
- Department of Biomedical Science, College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA.
- Department of Microbiology, College of Science, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
18
|
How an Immune-Factor-Based Formulation of Micro-Immunotherapy Could Interfere with the Physiological Processes Involved in the Atopic March. Int J Mol Sci 2023; 24:ijms24021483. [PMID: 36675006 PMCID: PMC9864899 DOI: 10.3390/ijms24021483] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Allergic diseases consist of improper inflammatory reactions to antigens and are currently an important healthcare concern, especially considering their increasing worldwide development in recent decades. The "atopic march" defines the paradigm of allergic diseases occurring in chronological order and displaying specific spatial manifestations, as they usually start as atopic dermatitis (AD) and food allergies during infancy and progressively evolve into allergic asthma (AA) and allergic rhinitis (AR) or rhino-conjunctivitis in childhood. Many immune cell subtypes and inflammatory factors are involved in these hypersensitivity reactions. In particular, the T helpers 2 (Th2) subset, through its cytokine signatures made of interleukins (ILs), such as IL-4, IL-5, IL-10, and IL-13, as well as mast cells and their related histamine pathways, contribute greatly to the perpetuation and evolution of the atopic march. By providing low doses (LD) and ultra-low doses (ULD) of ILs and immune factors to the body, micro-immunotherapy (MI) constitutes an interesting therapeutic strategy for the management of the atopic march and its symptoms. One of the aims of this review is to shed light on the current concept of the atopic march and the underlying immune reactions occurring during the IgE-mediated responses. Moreover, the different classes of traditional and innovative treatments employed in allergic diseases will also be discussed, with a special emphasis on the potential benefits of the MI medicine 2LALERG® formulation in this context.
Collapse
|
19
|
Biggs CM, Cordeiro-Santanach A, Prykhozhij SV, Deveau AP, Lin Y, Del Bel KL, Orben F, Ragotte RJ, Saferali A, Mostafavi S, Dinh L, Dai D, Weinacht KG, Dobbs K, Ott de Bruin L, Sharma M, Tsai K, Priatel JJ, Schreiber RA, Rozmus J, Hosking MC, Shopsowitz KE, McKinnon ML, Vercauteren S, Seear M, Notarangelo LD, Lynn FC, Berman JN, Turvey SE. Human JAK1 gain of function causes dysregulated myelopoeisis and severe allergic inflammation. JCI Insight 2022; 7:e150849. [PMID: 36546480 PMCID: PMC9869972 DOI: 10.1172/jci.insight.150849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
Primary atopic disorders are a group of inborn errors of immunity that skew the immune system toward severe allergic disease. Defining the biology underlying these extreme monogenic phenotypes reveals shared mechanisms underlying common polygenic allergic disease and identifies potential drug targets. Germline gain-of-function (GOF) variants in JAK1 are a cause of severe atopy and eosinophilia. Modeling the JAK1GOF (p.A634D) variant in both zebrafish and human induced pluripotent stem cells (iPSCs) revealed enhanced myelopoiesis. RNA-Seq of JAK1GOF human whole blood, iPSCs, and transgenic zebrafish revealed a shared core set of dysregulated genes involved in IL-4, IL-13, and IFN signaling. Immunophenotypic and transcriptomic analysis of patients carrying a JAK1GOF variant revealed marked Th cell skewing. Moreover, long-term ruxolitinib treatment of 2 children carrying the JAK1GOF (p.A634D) variant remarkably improved their growth, eosinophilia, and clinical features of allergic inflammation. This work highlights the role of JAK1 signaling in atopic immune dysregulation and the clinical impact of JAK1/2 inhibition in treating eosinophilic and allergic disease.
Collapse
Affiliation(s)
- Catherine M. Biggs
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | | | | | - Adam P. Deveau
- Department of Internal Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yi Lin
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Kate L. Del Bel
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Felix Orben
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Robert J. Ragotte
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Aabida Saferali
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Mostafavi
- Department of Medical Genetics and
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louie Dinh
- Department of Medical Genetics and
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Darlene Dai
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Katja G. Weinacht
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, California, USA
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Lisa Ott de Bruin
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mehul Sharma
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Kevin Tsai
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine and
| | - John J. Priatel
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine and
| | - Richard A. Schreiber
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Jacob Rozmus
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Martin C.K. Hosking
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Kevin E. Shopsowitz
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine and
| | | | | | - Michael Seear
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Francis C. Lynn
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason N. Berman
- CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Departments of Pediatrics and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Shimizu Y, Horigane-Konakai Y, Ishii Y, Sugimoto C, Wakao H. Mucosal-associated invariant T cells repress group 2 innate lymphoid cells in Alternaria alternata-induced model of allergic airway inflammation. Front Immunol 2022; 13:1005226. [PMID: 36458017 PMCID: PMC9706205 DOI: 10.3389/fimmu.2022.1005226] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/27/2022] [Indexed: 08/13/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells, a blossoming member of the innate-like T cells, play a pivotal role in host defense through engaging the mucosal immunity. Although it has been suggested that MAIT cells are somehow implicated in the allergic airway inflammation mediated by group 2 innate lymphoid cells (ILC2s) such as asthma, the precise role(s) of MAIT cells in such inflammation has remained elusive. To explore the possible roles of MAIT cells in the inflammation, we examined whether MAIT cells suppressed the production of T helper (Th) 2 and inflammatory cytokines from ILC2s, and constrained the proliferation of ILC2s, both of which are prerequisite for airway inflammation. Given that laboratory mice are poor at MAIT cells, a novel mouse line rich in MAIT cells was used. We found that mice rich in MAIT cells showed alleviated airway inflammation as evidenced by reduced infiltration of the immune cells and hyperplasia in goblet cells in the lung concomitant with compromised production of Th2 and inflammatory cytokines, while wild type mice exhibited severe inflammation upon challenge with the fungal extracts. In vitro coculture experiments using purified ILC2s and MAIT cells unrevealed that cytokine-stimulated MAIT cells suppressed ILC2s to produce the cytokines as well as to proliferate most likely via production of IFN-γ. Furthermore, reconstitution of the allergic airway inflammation in the highly immunocompromised mice showed that ILC2-mediated inflammation was alleviated in mice that received MAIT cells along with ILC2s. We concluded that MAIT cells played a crucial role in suppressing the cytokine-producing capacity of ILC2s and ILC2 proliferation, that ultimately led to decrease in the allergic airway inflammation. The results open up a novel therapeutic horizon in ILC2-mediated inflammatory diseases by modulating MAIT cell activity.
Collapse
Affiliation(s)
- Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
- Respiratory Endoscopy Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Yukiko Horigane-Konakai
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Yoshii Ishii
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Chie Sugimoto
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Hiroshi Wakao
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi, Japan
| |
Collapse
|
21
|
Chen Y, Huang A, Tan G, Liu H. Antagonism of m3 Alleviates Type 2 Inflammation in Allergic Rhinitis Mice. Am J Rhinol Allergy 2022; 37:264-272. [PMID: 36343939 DOI: 10.1177/19458924221137977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background Type 2 immune cells play a pivotal role in allergic rhinitis (AR). Increasing evidence shows that inhibition of cholinergic nerve activity decreases the severity of airway diseases including asthma and AR. However, the role of the cholinergic receptor muscarinic 3 (m3) in type 2 inflammation in AR is unknown. Objective We aimed to investigate the effect of m3 on the type 2 immune response, including both T helper 2 (Th2)-mediated and type 2 innate lymphocyte (ILC2)-mediated inflammation, in AR. Methods Peripheral blood mononuclear cells (PBMCs) from human were cultured in vitro. Treatment with the m3 antagonist 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide (4-DAMP) was used. The percentages of Th2 and ILC2 cells in PBMCs were evaluated by flow cytometry. AR mouse models were established by house dust mite (HDM) sensitization, and treated with tiotropium intranasally. The expression of Th2 cytokines, ILC2 cytokines and related factors in the nasal mucosa was assessed by immunohistochemistry and quantitative real-time polymerase chain reaction. Serum HDM-specific immunoglobulin E (sIgE) level was detected by enzyme-linked immunosorbent assay. Results Both Th2 and ILC2 percentages in PBMCs were decreased after 4-DAMP treatment. Similarly, the levels of Th2 cytokines (interleukin 4 [IL-4] and IL-13) and ILC2 cytokines and related factors (IL-25, IL-33, GATA3 and RORα) were significantly decreased in the nasal mucosa of AR mice after tiotropium treatment. Furthermore, tiotropium treatment decreased the nasal symptom score, the serum sIgE level and eosinophil infiltration in AR mice. In addition, tiotropium decreased phospholipase Cγ1 (PLCγ1), PLCγ2, nuclear factor of activated T cell 1 (NFATc1), and NFATc2 mRNA levels in AR mice. Conclusion Antagonism of m3 alleviated type 2 inflammation in the nasal mucosa of AR mice.
Collapse
Affiliation(s)
- Yu Chen
- Department of Otorhinolaryngology—Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Aijie Huang
- Department of Otorhinolaryngology—Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guolin Tan
- Department of Otorhinolaryngology—Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Honghui Liu
- Department of Otorhinolaryngology—Head Neck Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
22
|
Nedunchezhiyan U, Varughese I, Sun AR, Wu X, Crawford R, Prasadam I. Obesity, Inflammation, and Immune System in Osteoarthritis. Front Immunol 2022; 13:907750. [PMID: 35860250 PMCID: PMC9289681 DOI: 10.3389/fimmu.2022.907750] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity remains the most important risk factor for the incidence and progression of osteoarthritis (OA). The leading cause of OA was believed to be overloading the joints due to excess weight which in turn leads to the destruction of articular cartilage. However, recent studies have proved otherwise, various other factors like adipose deposition, insulin resistance, and especially the improper coordination of innate and adaptive immune responses may lead to the initiation and progression of obesity-associated OA. It is becoming increasingly evident that multiple inflammatory cells are recruited into the synovial joint that serves an important role in pathological changes in the synovial joint. Polarization of macrophages and macrophage-produced mediators are extensively studied and linked to the inflammatory and destructive responses in the OA synovium and cartilage. However, the role of other major innate immune cells such as neutrophils, eosinophils, and dendritic cells in the pathogenesis of OA has not been fully evaluated. Although cells of the adaptive immune system contribute to the pathogenesis of obesity-induced OA is still under exploration, a quantity of literature indicates OA synovium has an enriched population of T cells and B cells compared with healthy control. The interplay between a variety of immune cells and other cells that reside in the articular joints may constitute a vicious cycle, leading to pathological changes of the articular joint in obese individuals. This review addresses obesity and the role of all the immune cells that are involved in OA and summarised animal studies and human trials and knowledge gaps between the studies have been highlighted. The review also touches base on the interventions currently in clinical trials, different stages of the testing, and their shortcomings are also discussed to understand the future direction which could help in understanding the multifactorial aspects of OA where inflammation has a significant function.
Collapse
Affiliation(s)
- Udhaya Nedunchezhiyan
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ibin Varughese
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia RuJia Sun
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Xiaoxin Wu
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- Department of Orthopedic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ross Crawford
- Orthopedic Department, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
- *Correspondence: Indira Prasadam,
| |
Collapse
|
23
|
Ikutani M, Nakae S. Heterogeneity of Group 2 Innate Lymphoid Cells Defines Their Pleiotropic Roles in Cancer, Obesity, and Cardiovascular Diseases. Front Immunol 2022; 13:939378. [PMID: 35844571 PMCID: PMC9278653 DOI: 10.3389/fimmu.2022.939378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/02/2022] [Indexed: 11/24/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are typically known for their ability to respond rapidly to parasitic infections and play a pivotal role in the development of certain allergic disorders. ILC2s produce cytokines such as Interleukin (IL)-5 and IL-13 similar to the type 2 T helper (Th2) cells. Recent findings have highlighted that ILC2s, together with IL-33 and eosinophils, participate in a considerably broad range of physiological roles such as anti-tumor immunity, metabolic regulation, and vascular disorders. Therefore, the focus of the ILC2 study has been extended from conventional Th2 responses to these unexplored areas of research. However, disease outcomes accompanied by ILC2 activities are paradoxical mostly in tumor immunity requiring further investigations. Although various environmental factors that direct the development, activation, and localization of ILC2s have been studied, IL-33/ILC2/eosinophil axis is presumably central in a multitude of inflammatory conditions and has guided the research in ILC2 biology. With a particular focus on this axis, we discuss ILC2s across different diseases.
Collapse
Affiliation(s)
- Masashi Ikutani
- Laboratory of Immunology, Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- *Correspondence: Masashi Ikutani, ; Susumu Nakae,
| | - Susumu Nakae
- Laboratory of Immunology, Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama, Japan
- *Correspondence: Masashi Ikutani, ; Susumu Nakae,
| |
Collapse
|
24
|
Beeraka NM, Zhou R, Wang X, Vikram P R H, Kumar TP, Liu J, Greeshma MV, Mandal SP, Gurupadayya BM, Fan R. Immune Repertoire and Advancements in Nanotherapeutics for the Impediment of Severe Steroid Resistant Asthma (SSR). Int J Nanomedicine 2022; 17:2121-2138. [PMID: 35592101 PMCID: PMC9112344 DOI: 10.2147/ijn.s364693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/17/2022] [Indexed: 11/28/2022] Open
Abstract
Severe steroid-resistant asthma (SSR) patients do not respond to the corticosteroid therapies due to the heterogeneity, and genome-wide variations. However, there are very limited reports pertinent to the molecular signaling underlying SSR and making pharmacologists, and formulation scientists to identify the effective therapeutic targets in order to produce novel therapies using novel drug delivery systems (NDDS). We have substantially searched literature for the peer-reviewed and published reports delineating the role of glucocorticoid-altered gene expression, and the mechanisms responsible for SSR asthma, and NDDS for treating SSR asthma using public databases PubMed, National Library of Medicine (NLM), google scholar, and medline. Subsequently, we described reports underlying the SSR pathophysiology through several immunological and inflammatory phenotypes. Furthermore, various therapeutic strategies and the role of signaling pathways such as mORC1-STAT3-FGFBP1, NLRP3 inflammasomes, miR-21/PI3K/HDAC2 axis, PI3K were delineated and these can be considered as the therapeutic targets for mitigating the pathophysiology of SSR asthma. Finally, the possibility of nanomedicine-based formulation and their applications in order to enhance the long term retention of several antioxidant and anti-asthmatic drug molecules as a significant therapeutic modality against SSR asthma was described vividly.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Department of Human Anatomy, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical college, Mysuru, Karnataka, India
| | - Runze Zhou
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Xiaoyan Wang
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Hemanth Vikram P R
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, 570015, Karnataka, India
| | - Tegginamath Pramod Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysore, Karnataka, 570015, India
| | - Junqi Liu
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical college, Mysuru, Karnataka, India
| | - Subhankar P Mandal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, 570015, Karnataka, India
| | - B M Gurupadayya
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Ruitai Fan
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Correspondence: Ruitai Fan, Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, People’s Republic of China, Email
| |
Collapse
|
25
|
Sugimura R, Wang CY. The Role of Innate Lymphoid Cells in Cancer Development and Immunotherapy. Front Cell Dev Biol 2022; 10:803563. [PMID: 35557940 PMCID: PMC9086356 DOI: 10.3389/fcell.2022.803563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/11/2022] [Indexed: 11/29/2022] Open
Abstract
Innate Lymphoid Cells (ILCs) are an elusive type of innate immune cell that was only discovered recently. Their tissue residency and dependency makes them a niche group of cells that bridge the adaptive and innate immune system. The nomenclature and classification of ILCs have been challenging due to their heterogeneity. The currently agreed ILC classification splits the cells into two categories including cytotoxic and helper ILCs. The tumour microenvironment is often hostile for immune cells. Remodeling the microenvironment and regulating other immune cells—achieved by ILCs-can enhance anti-tumor effects. How ILCs regulate other immune cells in the tumor microenvironment remains to be understood. Here we review current understanding of the role of ILCs in the tumor microenvironment. ILCs recruit CD8 positive T and memory T cells in PDAC, ILCs are also able to help CD108 positive B cells migrate toward tumour locations. In NSCLC, ILC3s are seen helping resident macrophages enhancing the mucus immunity to cancer cells. We then highlight the roles of cytokines and immune checkpoint pathways in ILCs and its implication in immunotherapy.
Collapse
|
26
|
Lao JC, Bui CB, Pang MA, Cho SX, Rudloff I, Elgass K, Schröder J, Maksimenko A, Mangan NE, Starkey MR, Skuza EM, Sun YBY, Beker F, Collins CL, Kamlin OF, König K, Malhotra A, Tan K, Theda C, Young MJ, McLean CA, Wilson NJ, Sehgal A, Hansbro PM, Pearson JT, Polo JM, Veldman A, Berger PJ, Nold-Petry CA, Nold MF. Type 2 immune polarization is associated with cardiopulmonary disease in preterm infants. Sci Transl Med 2022; 14:eaaz8454. [PMID: 35385341 DOI: 10.1126/scitranslmed.aaz8454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Postnatal maturation of the immune system is poorly understood, as is its impact on illnesses afflicting term or preterm infants, such as bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension. These are both cardiopulmonary inflammatory diseases that cause substantial mortality and morbidity with high treatment costs. Here, we characterized blood samples collected from 51 preterm infants longitudinally at five time points, 20 healthy term infants at birth and age 3 to 16 weeks, and 5 healthy adults. We observed strong associations between type 2 immune polarization in circulating CD3+CD4+ T cells and cardiopulmonary illness, with odds ratios up to 24. Maternal magnesium sulfate therapy, delayed hepatitis B vaccination, and increasing fetal, but not maternal, chorioamnionitis severity were associated with attenuated type 2 polarization. Blocking type 2 mediators such as interleukin-4 (IL-4), IL-5, IL-13, or signal transducer and activator of transcription 6 (STAT6) in murine neonatal cardiopulmonary disease in vivo prevented changes in cell type composition, increases in IL-1β and IL-13, and losses of pulmonary capillaries, but not gains in larger vessels. Thereby, type 2 blockade ameliorated lung inflammation, protected alveolar and vascular integrity, and confirmed the pathological impact of type 2 cytokines and STAT6. In-depth flow cytometry and single-cell transcriptomics of mouse lungs further revealed complex associations between immune polarization and cardiopulmonary disease. Thus, this work advances knowledge on developmental immunology and its impact on early life disease and identifies multiple therapeutic approaches that may relieve inflammation-driven suffering in the youngest patients.
Collapse
Affiliation(s)
- Jason C Lao
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Christine B Bui
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Merrin A Pang
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Steven X Cho
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Ina Rudloff
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Kirstin Elgass
- Monash Micro Imaging, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Jan Schröder
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Anton Maksimenko
- Imaging and Medical Beamline, Australian Synchrotron, Melbourne, Victoria 3168, Australia
| | - Niamh E Mangan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, Victoria 3168, Australia
| | - Malcolm R Starkey
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Elisabeth M Skuza
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Yu B Y Sun
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Friederike Beker
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4101, Australia.,Neonatal Services, Mercy Hospital for Women, Melbourne, Victoria 3084, Australia
| | - Clare L Collins
- Neonatal Services, Mercy Hospital for Women, Melbourne, Victoria 3084, Australia
| | - Omar F Kamlin
- Department of Newborn Research, Royal Women's Hospital, Melbourne, Victoria 3052, Australia.,University of Melbourne, Melbourne, Victoria 3010, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria 3052, Australia
| | - Kai König
- Department of Paediatrics, Medicum Wesemlin, Lucerne 6006, Switzerland
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Kenneth Tan
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Christiane Theda
- Department of Newborn Research, Royal Women's Hospital, Melbourne, Victoria 3052, Australia.,University of Melbourne, Melbourne, Victoria 3010, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria 3052, Australia
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Health, Melbourne, Victoria 3004, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Victoria 3800, Australia
| | - Nicholas J Wilson
- CSL Limited, Bio21 Institute, Parkville, Melbourne, Victoria 3052, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Philip M Hansbro
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Ultimo, Sydney, New South Wales 2007, Australia
| | - James T Pearson
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia.,Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.,Victorian Heart Institute, Melbourne, Victoria 3168, Australia
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Adelaide Centre for Epigenetics, University of Adelaide, Adelaide, South Australia 5005, Australia.,South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Alex Veldman
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Department of Pediatrics, Helios HSK, Wiesbaden 65199, Germany.,Department of Pediatric Cardiology, J. Liebig University, Gießen 35392, Germany
| | - Philip J Berger
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Claudia A Nold-Petry
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| |
Collapse
|
27
|
Orimo K, Tamari M, Takeda T, Kubo T, Rückert B, Motomura K, Sugiyama H, Yamada A, Saito K, Arae K, Kuriyama M, Hara M, Soyka MB, Ikutani M, Yamaguchi S, Morimoto N, Nakabayashi K, Hata K, Matsuda A, Akdis CA, Sudo K, Saito H, Nakae S, Tamaoki J, Tagaya E, Matsumoto K, Morita H. Direct platelet adhesion potentiates group 2 innate lymphoid cell functions. Allergy 2022; 77:843-855. [PMID: 34402091 DOI: 10.1111/all.15057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Platelets are thought to be involved in the pathophysiology of asthma, presumably through direct adhesion to inflammatory cells, including group 2 innate lymphoid cells (ILC2s). Here, we tried to elucidate the effects of platelet adhesion to ILC2s in vitro and in vivo, as well as the mechanisms involved. METHODS Alternaria-induced ILC2-dependent airway inflammation models using wild-type and c-mpl-/- mice were evaluated. Both purified CD41+ and CD41- ILC2s were cultured with IL-2 and IL-33 to determine in vitro Type 2 (T2) cytokine production and cell proliferation. RNA-seq data of flow-cytometry-sorted CD41+ and CD41- ILC2s were used to isolate ILC2-specific genes. Flow cytometry was performed to determine the expression of CD41 and adhesion-related molecules on ILC2s in both mouse and human tissues. RESULTS T2 inflammation and T2 cytokine production from ILC2s were significantly reduced in the c-mpl-/- mice compared to wild-type mice. Platelet-adherent ILC2s underwent significant proliferation and showed enhanced T2 cytokine production when exposed to IL-2 and IL-33. The functions of ILC2-specific genes were related to cell development and function. Upstream regulator analysis identified 15 molecules, that are thought to be involved in ILC2 activation. CD41 expression levels were higher in ILC2s from human PBMCs and mouse lung than in those from secondary lymphoid tissues, but they did not correlate with the P-selectin glycoprotein ligand-1 or CD24 expression level. CONCLUSION Platelets spontaneously adhere to ILC2s, probably in the peripheral blood and airways, thereby potentiating ILC2s to enhance their responses to IL-33.
Collapse
Affiliation(s)
- Keisuke Orimo
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Masato Tamari
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Pediatrics Jikei University School of Medicine Tokyo Japan
| | - Tomohiro Takeda
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Health Science Kansai University of Health Sciences Osaka Japan
| | - Terufumi Kubo
- Department of Pathology Sapporo Medical University School of Medicine Sapporo Japan
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Hiroki Sugiyama
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Ayako Yamada
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Kyoko Saito
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Otorhinolaryngology Head and Neck Surgery University of Fukui Fukui Japan
| | - Ken Arae
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Immunology Faculty of Health Sciences Kyorin University Tokyo Japan
| | - Motohiro Kuriyama
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Mariko Hara
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Michael B. Soyka
- Department of Otorhinolaryngology, Head and Neck Surgery University Hospital Zurich and University of Zurich Zurich Switzerland
| | - Masashi Ikutani
- Graduate School of Integrated Sciences for Life Hiroshima University Higashi‐Hiroshima City Japan
- Department of Immune Regulation Research Institute, National Center for Global Health and Medicine Ichikawa Japan
| | - Sota Yamaguchi
- Division of Otolaryngology Department of Surgical Specialties National Center for Child Health and Development Tokyo Japan
| | - Noriko Morimoto
- Division of Otolaryngology Department of Surgical Specialties National Center for Child Health and Development Tokyo Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal–Fetal Biology National Research Institute for Child Health and Development Tokyo Japan
| | - Kenichiro Hata
- Department of Maternal–Fetal Biology National Research Institute for Child Health and Development Tokyo Japan
| | - Akio Matsuda
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Katsuko Sudo
- Animal Research Center Tokyo Medical University Tokyo Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Susumu Nakae
- Department of Immune Regulation Research Institute, National Center for Global Health and Medicine Ichikawa Japan
- Laboratory of Systems Biology Center for Experimental Medicine and Systems Biology The Institute of Medical Science, The University of Tokyo Tokyo Japan
- Precursory Research for Embryonic Science and Technology (PRESTO Japan Science and Technology Agency Saitama Japan
| | - Jun Tamaoki
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Etsuko Tagaya
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Allergy Center National Center for Child Health and Development Tokyo Japan
| |
Collapse
|
28
|
Pivniouk V, Gimenes-Junior JA, Ezeh P, Michael A, Pivniouk O, Hahn S, VanLinden SR, Malone SP, Abidov A, Anderson D, Gozdz J, DeVries A, Martinez FD, Pasquali C, Vercelli D. Airway administration of OM-85, a bacterial lysate, blocks experimental asthma by targeting dendritic cells and the epithelium/IL-33/ILC2 axis. J Allergy Clin Immunol 2022; 149:943-956. [PMID: 34560105 PMCID: PMC8901455 DOI: 10.1016/j.jaci.2021.09.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Microbial interventions against allergic asthma have robust epidemiologic underpinnings and the potential to recalibrate disease-inducing immune responses. Oral administration of OM-85, a standardized lysate of human airways bacteria, is widely used empirically to prevent respiratory infections and a clinical trial is testing its ability to prevent asthma in high-risk children. We previously showed that intranasal administration of microbial products from farm environments abrogates experimental allergic asthma. OBJECTIVES We sought to investigate whether direct administration of OM-85 to the airway compartment protects against experimental allergic asthma; and to identify protective cellular and molecular mechanisms activated through this natural route. METHODS Different strains of mice sensitized and challenged with ovalbumin or Alternaria received OM-85 intranasally, and cardinal cellular and molecular asthma phenotypes were measured. Airway transfer experiments assessed whether OM-85-treated dendritic cells protect allergen-sensitized, OM-85-naive mice against asthma. RESULTS Airway OM-85 administration suppressed allergic asthma in all models acting on multiple innate and adaptive immune targets: the airway epithelium/IL-33/ILC2 axis, lung allergen-induced type 2 responses, and dendritic cells whose Myd88/Trif-dependent tolerogenic reprogramming was sufficient to transfer OM-85-induced asthma protection. CONCLUSIONS We provide the first demonstration that administering a standardized bacterial lysate to the airway compartment protects from experimental allergic asthma by engaging multiple immune pathways. Because protection required a cumulative dose 27- to 46-fold lower than the one reportedly active through the oral route, the efficacy of intranasal OM-85 administration may reflect its direct access to the airway mucosal networks controlling the initiation and development of allergic asthma.
Collapse
Affiliation(s)
- Vadim Pivniouk
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Ariz; Asthma and Airway Disease Research Center, The University of Arizona, Tucson, Ariz; BIO5 Institute, The University of Arizona, Tucson, Ariz.
| | - Joao A. Gimenes-Junior
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Peace Ezeh
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Ashley Michael
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Oksana Pivniouk
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Seongmin Hahn
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Sydney R. VanLinden
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Sean P. Malone
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Amir Abidov
- Medical Student Research Program, College of Medicine, The
University of Arizona, Tucson, AZ, 85721, USA
| | - Dayna Anderson
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Justyna Gozdz
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA
| | - Avery DeVries
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA
| | - Fernando D. Martinez
- Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA
| | | | - Donata Vercelli
- Department of Cellular and Molecular Medicine, The
University of Arizona, Tucson, AZ, 85721, USA,Asthma and Airway Disease Research Center, The University
of Arizona, Tucson, AZ, 85721, USA,The BIO5 Institute, The University of Arizona, Tucson, AZ,
85721, USA,Arizona Center for the Biology of Complex Diseases, The
University of Arizona
| |
Collapse
|
29
|
Xiao Q, Han X, Liu G, Zhou D, Zhang L, He J, Xu H, Zhou P, Yang Q, Chen J, Zhou J, Jiang G, Yao Z. Adenosine restrains ILC2-driven allergic airway inflammation via A2A receptor. Mucosal Immunol 2022; 15:338-350. [PMID: 34921233 DOI: 10.1038/s41385-021-00475-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/05/2021] [Accepted: 12/05/2021] [Indexed: 02/04/2023]
Abstract
Although group 2 Innate Lymphoid Cells (ILC2s) play important roles in driving the pathogenesis of allergic airway inflammation, the molecular mechanisms regulating ILC2 responses remain to be fully elucidated. Adenosine signaling is emerging as an important factor to limit excessive inflammation and tissue damage, its role in ILC2-driven airway inflammation remains to be understood. Here we identify adenosine as a negative regulator of ILC2s and allergic airway inflammation. Elevation of adenosine was observed in lungs after protease papain challenge. Adenosine receptor A2A was abundantly expressed in lung ILC2s. The adenosine analog NECA significantly suppress ILC2s responses and relieved airway inflammation induced by IL-33 or papain. Conversely, blockage of adenosine synthesis by CD73 inhibitor APCP or deficiency of A2A aggravated murine airway inflammation. Adoptive transfer of ILC2s into immunodeficiency NCG mice demonstrated that the regulation of ILC2 by adenosine was cell intrinsic. Mechanistic studies showed that the effects of adenosine on ILC2s were associated with changes in transcriptional profiling, and the elevation of intracellular cAMP and resulted NF-κB downregulation. These observations indicate that adenosine-A2A signaling is a negative regulator of ILC2s, which confers protection against airway inflammation and represents a novel therapeutic target for controlling asthma.
Collapse
Affiliation(s)
- Qiang Xiao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Xu Han
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Gaoyu Liu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Dongmei Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijuan Zhang
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Juan He
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Haixu Xu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Pan Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiangfan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Guanmin Jiang
- Department of Clinical laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China.
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
30
|
Mu M, Li B, Zou Y, Wang W, Cao H, Zhang Y, Sun Q, Chen H, Ge D, Tao H, Hu D, Yuan L, Tao X, Wang J. Coal dust exposure triggers heterogeneity of transcriptional profiles in mouse pneumoconiosis and Vitamin D remedies. Part Fibre Toxicol 2022; 19:7. [PMID: 35057792 PMCID: PMC8772169 DOI: 10.1186/s12989-022-00449-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/22/2022] Open
Abstract
Background Coal dust particles (CDP), an inevitable by-product of coal mining for the environment, mainly causes coal workers’ pneumoconiosis (CWP). Long-term exposure to coal dust leads to a complex alternation of biological processes during regeneration and repair in the healing lung. However, the cellular and complete molecular changes associated with pulmonary homeostasis caused by respiratory coal dust particles remain unclear. Methods This study mainly investigated the pulmonary toxicity of respirable-sized CDP in mice using unbiased single-cell RNA sequencing. CDP (< 5 μm) collected from the coal mine was analyzed by Scanning Electron Microscope (SEM) and Mass Spectrometer. In addition, western blotting, Elisa, QPCR was used to detect gene expression at mRNA or protein levels. Pathological analysis including HE staining, Masson staining, immunohistochemistry, and immunofluorescence staining were performed to characterize the structure and functional alternation in the pneumoconiosis mouse and verify the reliability of single-cell sequencing results. Results SEM image and Mass Spectrometer analysis showed that coal dust particles generated during coal mine production have been crushed and screened with a diameter of less than 5 µm and contained less than 10% silica. Alveolar structure and pulmonary microenvironment were destroyed, inflammatory and death (apoptosis, autophagy, and necrosis) pathways were activated, leading to pneumoconiosis in post 9 months coal dust stimulation. A distinct abnormally increased alveolar type 2 epithelial cell (AT2) were classified with a highly active state but reduced the antimicrobial-related protein expression of LYZ and Chia1 after CDP exposure. Beclin1, LC3B, LAMP2, TGF-ß, and MLPH were up-regulated induced by CDP, promoting autophagy and pulmonary fibrosis. A new subset of macrophages with M2-type polarization double expressed MLPH + /CD206 + was found in mice having pneumoconiosis but markedly decreased after the Vitamin D treatment. Activated MLPH + /CD206 + M2 macrophages secreted TGF-β1 and are sensitive to Vitamin D treatment. Conclusions This is the first study to reconstruct the pathologic progression and transcriptome pattern of coal pneumoconiosis in mice. Coal dust had obvious toxic effects on lung epithelial cells and macrophages and eventually induced pulmonary fibrosis. CDP-induced M2-type macrophages could be inhibited by VD, which may be related to the alleviation of the pulmonary fibrosis process. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00449-y.
Collapse
|
31
|
Baker JR, Rasky AJ, Landers JJ, Janczak KW, Totten TD, Lukacs NW, O’Konek JJ. Intranasal delivery of allergen in a nanoemulsion adjuvant inhibits allergen-specific reactions in mouse models of allergic airway disease. Clin Exp Allergy 2021; 51:1361-1373. [PMID: 33999457 PMCID: PMC11155263 DOI: 10.1111/cea.13903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 05/07/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Atopic diseases are an increasing problem that involve both immediate hypersensitivity reactions mediated by IgE and unique cellular inflammation. Many forms of specific immunotherapy involve the administration of allergen to suppress allergic immune responses but are focused on IgE-mediated reactions. In contrast, the effect of allergen-specific immunotherapy on allergic inflammation is complex, not entirely consistent and not well understood. We have previously demonstrated the ability of allergen administered in a nanoemulsion (NE) mucosal adjuvant to suppress IgE-mediated allergic responses and protect from allergen challenge in murine food allergy models. This activity was associated with decreases in allergen-specific IL-10 and reductions in allergic cytokines and increases in regulatory T cells. OBJECTIVE Here, we extend these studies to using 2 distinct models, the ovalbumin (OVA) and cockroach (CRA) models of allergic airway disease, which are based predominantly on allergic inflammation. METHODS Acute or chronic allergic airway disease was induced in mice using ovalbumin and cockroach allergen models. Mice received three therapeutic immunizations with allergen in NE, and reactivity to airway challenge was determined. RESULTS Therapeutic immunization with cockroach or OVA allergen in NE markedly reduced pathology after airway challenge. The 2 models demonstrated protection from allergen challenge-induced pathology that was associated with suppression of Th2-polarized immune responses in the lung. In addition, the reduction in ILC2 numbers in the lungs of allergic mice along with reduction in epithelial cell alarmins, IL-25 and IL-33, suggests an overall change in the lung immune environment induced by the NE immunization protocol. CONCLUSIONS AND CLINICAL RELEVANCE These results demonstrate that suppression of allergic airway inflammation and bronchial hyper-reactivity can be achieved using allergen-specific immunotherapy without significant reductions in allergen-specific IgE and suggest that ILC2 cells may be critical targets for this activity.
Collapse
Affiliation(s)
- James R. Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J. Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey J. Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Tiffanie D. Totten
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jessica J. O’Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Kuchibhotla VNS, Starkey MR, Reid AT, Heijink IH, Nawijn MC, Hansbro PM, Knight DA. Inhibition of β-Catenin/CREB Binding Protein Signaling Attenuates House Dust Mite-Induced Goblet Cell Metaplasia in Mice. Front Physiol 2021; 12:690531. [PMID: 34385933 PMCID: PMC8353457 DOI: 10.3389/fphys.2021.690531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
Abstract
Excessive mucus production is a major feature of allergic asthma. Disruption of epithelial junctions by allergens such as house dust mite (HDM) results in the activation of β-catenin signaling, which has been reported to stimulate goblet cell differentiation. β-catenin interacts with various co-activators including CREB binding protein (CBP) and p300, thereby regulating the expression of genes involved in cell proliferation and differentiation, respectively. We specifically investigated the role of the β-catenin/CBP signaling pathway in goblet cell metaplasia in a HDM-induced allergic airway disease model in mice using ICG-001, a small molecule inhibitor that blocks the binding of CBP to β-catenin. Female 6- 8-week-old BALB/c mice were sensitized to HDM/saline on days 0, 1, and 2, followed by intranasal challenge with HDM/saline with or without subcutaneous ICG-001/vehicle treatment from days 14 to 17, and samples harvested 24 h after the last challenge/treatment. Differential inflammatory cells in bronchoalveolar lavage (BAL) fluid were enumerated. Alcian blue (AB)/Periodic acid–Schiff (PAS) staining was used to identify goblet cells/mucus production, and airway hyperresponsiveness (AHR) was assessed using invasive plethysmography. Exposure to HDM induced airway inflammation, goblet cell metaplasia and increased AHR, with increased airway resistance in response to the non-specific spasmogen methacholine. Inhibition of the β-catenin/CBP pathway using treatment with ICG-001 significantly attenuated the HDM-induced goblet cell metaplasia and infiltration of macrophages, but had no effect on eosinophils, neutrophils, lymphocytes or AHR. Increased β-catenin/CBP signaling may promote HDM-induced goblet cell metaplasia in mice.
Collapse
Affiliation(s)
- Virinchi N S Kuchibhotla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre GrowUpWell and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Andrew T Reid
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Irene H Heijink
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands.,Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martijn C Nawijn
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Providence Health Care Research Institute, Vancouver, BC, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Blomme EE, Provoost S, De Smet EG, De Grove KC, Van Eeckhoutte HP, De Volder J, Hansbro PM, Bonato M, Saetta M, Wijnant SR, Verhamme F, Joos GF, Bracke KR, Brusselle GG, Maes T. Quantification and role of innate lymphoid cell subsets in Chronic Obstructive Pulmonary Disease. Clin Transl Immunology 2021; 10:e1287. [PMID: 34136217 PMCID: PMC8178740 DOI: 10.1002/cti2.1287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 01/02/2023] Open
Abstract
Objectives Innate lymphoid cells (ILCs) secrete cytokines, such as IFN‐γ, IL‐13 and IL‐17, which are linked to chronic obstructive pulmonary disease (COPD). Here, we investigated the role of pulmonary ILCs in COPD pathogenesis. Methods Lung ILC subsets in COPD and control subjects were quantified using flow cytometry and associated with clinical parameters. Tissue localisation of ILC and T‐cell subsets was determined by immunohistochemistry. Mice were exposed to air or cigarette smoke (CS) for 1, 4 or 24 weeks to investigate whether pulmonary ILC numbers and activation are altered and whether they contribute to CS‐induced innate inflammatory responses. Results Quantification of lung ILC subsets demonstrated that ILC1 frequency in the total ILC population was elevated in COPD and was associated with smoking and severity of respiratory symptoms (COPD Assessment Test [CAT] score). All three ILC subsets localised near lymphoid aggregates in COPD. In the COPD mouse model, CS exposure in C57BL/6J mice increased ILC numbers at all time points, with relative increases in ILC1 in bronchoalveolar lavage (BAL) fluid. Importantly, CS exposure induced increases in neutrophils, monocytes and dendritic cells that remained elevated in Rag2/Il2rg‐deficient mice that lack adaptive immune cells and ILCs. However, CS‐induced CXCL1, IL‐6, TNF‐α and IFN‐γ levels were reduced by ILC deficiency. Conclusion The ILC1 subset is increased in COPD patients and correlates with smoking and severity of respiratory symptoms. ILCs also increase upon CS exposure in C57BL/6J mice. In the absence of adaptive immunity, ILCs contribute to CS‐induced pro‐inflammatory mediator release, but are redundant in CS‐induced innate inflammation.
Collapse
Affiliation(s)
- Evy E Blomme
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Sharen Provoost
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Elise G De Smet
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Katrien C De Grove
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Hannelore P Van Eeckhoutte
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Joyceline De Volder
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Philip M Hansbro
- Centre for Inflammation Centenary Institute Sydney NSW Australia.,Faculty of Science University of Technology Sydney Ultimo NSW Australia
| | - Matteo Bonato
- Faculty of Science University of Technology Sydney Ultimo NSW Australia
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health University of Padova Padova Italy
| | - Sara Ra Wijnant
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium.,Department of Epidemiology Erasmus Medical Center Rotterdam The Netherlands.,Department of Bioanalysis Faculty of Pharmaceutical Sciences Ghent University Ghent Belgium
| | - Fien Verhamme
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Guy F Joos
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Ken R Bracke
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| | - Tania Maes
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent Belgium
| |
Collapse
|
35
|
Marega M, Chen C, Bellusci S. Cross-Talk Between Inflammation and Fibroblast Growth Factor 10 During Organogenesis and Pathogenesis: Lessons Learnt From the Lung and Other Organs. Front Cell Dev Biol 2021; 9:656883. [PMID: 34136479 PMCID: PMC8201783 DOI: 10.3389/fcell.2021.656883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/19/2021] [Indexed: 11/21/2022] Open
Abstract
The adult human lung is constantly exposed to irritants like particulate matter, toxic chemical compounds, and biological agents (bacteria and viruses) present in the external environment. During breathing, these irritants travel through the bronchi and bronchioles to reach the deeper lung containing the alveoli, which constitute the minimal functional respiratory units. The local biological responses in the alveoli that follow introduction of irritants need to be tightly controlled in order to prevent a massive inflammatory response leading to loss of respiratory function. Cells, cytokines, chemokines and growth factors intervene collectively to re-establish tissue homeostasis, fight the aggression and replace the apoptotic/necrotic cells with healthy cells through proliferation and/or differentiation. Among the important growth factors at play during inflammation, members of the fibroblast growth factor (Fgf) family regulate the repair process. Fgf10 is known to be a key factor for organ morphogenesis and disease. Inflammation is influenced by Fgf10 but can also impact Fgf10 expression per se. Unfortunately, the connection between Fgf10 and inflammation in organogenesis and disease remains unclear. The aim of this review is to highlight the reported players between Fgf10 and inflammation with a focus on the lung and to propose new avenues of research.
Collapse
Affiliation(s)
- Manuela Marega
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Member of the German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, Giessen, Germany
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Saverio Bellusci
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Member of the German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
36
|
Loering S, Cameron GJM, Bhatt NP, Belz GT, Foster PS, Hansbro PM, Starkey MR. Differences in pulmonary group 2 innate lymphoid cells are dependent on mouse age, sex and strain. Immunol Cell Biol 2021; 99:542-551. [PMID: 33295058 DOI: 10.1111/imcb.12430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 01/06/2023]
Abstract
Innate lymphoid cells (ILCs) are resident in the lung and are involved in both the maintenance of homeostasis and the pathogenesis of respiratory diseases. In this study, murine lung ILCs were characterized using flow cytometry and the impact of mouse age, sex and strain were assessed. Lung ILCs were found as early as postnatal day 4 and numbers peaked at 2 weeks, and then decreased as the lung matured. During postnatal lung development, ILC expressed differential amounts of group 2 ILC (ILC2)-associated cell surface antigens including ST2, CD90.2 and ICOS. Using Il5venus Il13td-tomato dual reporter mice, neonates were found to have increased constitutive interleukin (IL)-13 expression compared with adult mice. Neonates and adults had similar ratios of IL-5+ CD45+ leukocytes; however, these cells were mostly composed of ILCs in neonates and T cells in adults. Sex-specific differences in ILC numbers were also observed, with females having greater numbers of lung ILCs than males in both neonatal and adult mice. Female lung ILCs also expressed higher levels of ICOS and decreased KLRG1. Mouse strain also impacted on lung ILCs with BALB/c mice having more ILCs in the lung and increased expression of ST2 and ICOS compared with C57BL/6J mice. Collectively, these data show that lung ILC numbers, cell surface antigen expression, IL-5 and IL-13 levels differed between neonatal and adult lung ILCs. In addition, cell surface antigens commonly used for ILC2 quantification, such as ST2, CD90.2 and ICOS, differ depending on age, sex and strain and these are important considerations for consistent universal identification of lung ILC2s.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Guy J M Cameron
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nirmal P Bhatt
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Gabrielle T Belz
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland Translational Research Institute, Woolloongabba, QLD, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Paul S Foster
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology, Sydney, NSW, Australia
| | - Malcolm R Starkey
- Priority Research Centre's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Fonseca W, Lukacs NW, Elesela S, Malinczak CA. Role of ILC2 in Viral-Induced Lung Pathogenesis. Front Immunol 2021; 12:675169. [PMID: 33953732 PMCID: PMC8092393 DOI: 10.3389/fimmu.2021.675169] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Innate lymphoid type-2 cells (ILC2) are a population of innate cells of lymphoid origin that are known to drive strong Type 2 immunity. ILC2 play a key role in lung homeostasis, repair/remodeling of lung structures following injury, and initiation of inflammation as well as more complex roles during the immune response, including the transition from innate to adaptive immunity. Remarkably, dysregulation of this single population has been linked with chronic lung pathologies, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrotic diseases (IPF). Furthermore, ILC2 have been shown to increase following early-life respiratory viral infections, such as respiratory syncytial virus (RSV) and rhinovirus (RV), that may lead to long-term alterations of the lung environment. The detrimental roles of increased ILC2 following these infections may include pathogenic chronic inflammation and/or alterations of the structural, repair, and even developmental processes of the lung. Respiratory viral infections in older adults and patients with established chronic pulmonary diseases often lead to exacerbated responses, likely due to previous exposures that leave the lung in a dysregulated functional and structural state. This review will focus on the role of ILC2 during respiratory viral exposures and their effects on the induction and regulation of lung pathogenesis. We aim to provide insight into ILC2-driven mechanisms that may enhance lung-associated diseases throughout life. Understanding these mechanisms will help identify better treatment options to limit not only viral infection severity but also protect against the development and/or exacerbation of other lung pathologies linked to severe respiratory viral infections.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Srikanth Elesela
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
38
|
Maruyama K. Senso-immunology: crosstalk between nociceptive and immune systems. FEBS J 2021; 289:4132-4145. [PMID: 33780155 DOI: 10.1111/febs.15846] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/03/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022]
Abstract
Understanding the molecular mechanisms of nociception has recently grown impressively. Nociception is mediated by mechanical, chemical, or microbial stimuli that evoke unpleasant feelings, alerting the host of the risk of tissue damage. Such diverse arrays of noxious stimuli trigger various escape reactions, usually altering immune homeostasis. Notably, nociceptors can recognize cytokines or pathogens via sensory molecules or innate immune receptors, participating in immune responses. Accumulating evidence suggests that activated nociceptors produce various humoral factors that affect the immune system and act like endocrine/paracrine signals. Thus, understanding the interplay between the nociceptive and immune systems may open new avenues for the development of an interdisciplinary research field, hereinafter referred to as 'senso-immunology'. This review will discuss the physiological relevance of the senso-immune system in the host defense context, focusing on how senso-immune research might yield novel treatments to cure pain and inflammation.
Collapse
Affiliation(s)
- Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| |
Collapse
|
39
|
Nakatsuka Y, Yaku A, Handa T, Vandenbon A, Hikichi Y, Motomura Y, Sato A, Yoshinaga M, Tanizawa K, Watanabe K, Hirai T, Chin K, Suzuki Y, Uehata T, Mino T, Tsujimura T, Moro K, Takeuchi O. Profibrotic function of pulmonary group 2 innate lymphoid cells is controlled by regnase-1. Eur Respir J 2021; 57:13993003.00018-2020. [PMID: 32978308 DOI: 10.1183/13993003.00018-2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 09/11/2020] [Indexed: 12/23/2022]
Abstract
Regnase-1 is an RNase critical for post-transcriptional control of pulmonary immune homeostasis in mice by degrading immune-related mRNAs. However, little is known about the cell types Regnase-1 controls in the lung, and its relevance to human pulmonary diseases.Regnase-1-dependent changes in lung immune cell types were examined by a competitive bone marrow transfer mouse model, and group 2 innate lymphoid cells (ILC2s) were identified. Then the associations between Regnase-1 in ILC2s and human diseases were investigated by transcriptome analysis and a bleomycin-induced pulmonary fibrosis mouse model. The clinical significance of Regnase-1 in ILC2s was further assessed using patient-derived cells.Regnase-1-deficiency resulted in the spontaneous proliferation and activation of ILC2s in the lung. Intriguingly, genes associated with pulmonary fibrosis were highly upregulated in Regnase-1-deficient ILC2s compared with wild-type, and supplementation of Regnase-1-deficient ILC2s augmented bleomycin-induced pulmonary fibrosis in mice. Regnase-1 suppresses mRNAs encoding transcription factors Gata3 and Egr1, which are potent to regulate fibrosis-associated genes. Clinically, Regnase-1 protein levels in ILC2 negatively correlated with the ILC2 population in bronchoalveolar lavage fluid. Furthermore, idiopathic pulmonary fibrosis (IPF) patients with ILC2s >1500 cells·mL-1 peripheral blood exhibited poorer prognosis than patients with lower numbers, implying the contribution of Regnase-1 in ILC2s for the progression of IPF.Collectively, Regnase-1 was identified as a critical post-transcriptional regulator of the profibrotic function of ILC2s both in mouse and human, suggesting that Regnase-1 may be a novel therapeutic target for IPF.
Collapse
Affiliation(s)
- Yoshinari Nakatsuka
- Dept of Respiratory Care and Sleep Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ai Yaku
- Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Dept of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Handa
- Dept of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Alexis Vandenbon
- Laboratory of Systems Virology, Dept of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yuki Hikichi
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Yasutaka Motomura
- Dept of Microbiology and Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ayuko Sato
- Dept of Pathology, Hyogo College of Medicine, Hyogo, Japan
| | - Masanori Yoshinaga
- Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiminobu Tanizawa
- Dept of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kizuku Watanabe
- Dept of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Dept of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuo Chin
- Dept of Respiratory Care and Sleep Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yutaka Suzuki
- Laboratory of Functional Genomics, Dept of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Takuya Uehata
- Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Mino
- Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.,Dept of Microbiology and Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Osamu Takeuchi
- Dept of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
40
|
Farazuddin M, Landers JJ, Janczak KW, Lindsey HK, Finkelman FD, Baker JR, O'Konek JJ. Mucosal Nanoemulsion Allergy Vaccine Suppresses Alarmin Expression and Induces Bystander Suppression of Reactivity to Multiple Food Allergens. Front Immunol 2021; 12:599296. [PMID: 33717078 PMCID: PMC7946984 DOI: 10.3389/fimmu.2021.599296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
We have demonstrated that intranasal immunotherapy with allergens formulated in a nanoemulsion (NE) mucosal adjuvant suppresses Th2/IgE-mediated allergic responses and protects from allergen challenge in murine food allergy models. Protection conferred by this therapy is associated with strong suppression of allergen specific Th2 cellular immunity and increased Th1 cytokines. Here we extend these studies to examine the effect of NE-allergen immunization in mice sensitized to multiple foods. Mice were sensitized to both egg and peanut and then received NE vaccine formulated with either one or both of these allergens. The animals were then subjected to oral challenges with either egg or peanut to assess reactivity. Immunization with NE formulations containing both egg and peanut markedly reduced reactivity after oral allergen challenge with either allergen. Interestingly, mice that received the vaccine containing only peanut also had reduced reactivity to challenge with egg. Protection from oral allergen challenge was achieved despite the persistence of allergen-specific IgE and was associated with strong suppression of both Th2-polarized immune responses, alarmins and type 2 innate lymphoid cells (ILC2). NE-induced bystander suppression of reactivity required IFN-γ and the presence of an allergen in the NE vaccine. These results demonstrate that anaphylactic reactions to food allergens can be suppressed using allergen-specific immunotherapy without having to eliminate allergen-specific IgE and suggests that modulation of Th2 immunity towards one allergen may induce bystander effects that suppress reactivity to other allergens through the induction of IFN-γ and suppression of alarmins in the intestine. In addition, these data suggest that a NE vaccine for a single food allergen may lead to a global suppression of allergic responses to multiple foods.
Collapse
Affiliation(s)
- Mohammad Farazuddin
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Katarzyna W Janczak
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Hayley K Lindsey
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Fred D Finkelman
- Division of Allergy, Immunology and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| | - Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
41
|
Zhang H, He F, Li P, Hardwidge PR, Li N, Peng Y. The Role of Innate Immunity in Pulmonary Infections. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6646071. [PMID: 33553427 PMCID: PMC7847335 DOI: 10.1155/2021/6646071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Innate immunity forms a protective line of defense in the early stages of pulmonary infection. The primary cellular players of the innate immunity against respiratory infections are alveolar macrophages (AMs), dendritic cells (DCs), neutrophils, natural killer (NK) cells, and innate lymphoid cells (ILCs). They recognize conserved structures of microorganisms through membrane-bound and intracellular receptors to initiate appropriate responses. In this review, we focus on the prominent roles of innate immune cells and summarize transmembrane and cytosolic pattern recognition receptor (PRR) signaling recognition mechanisms during pulmonary microbial infections. Understanding the mechanisms of PRR signal recognition during pulmonary pathogen infections will help us to understand pulmonary immunopathology and lay a foundation for the development of effective therapies to treat and/or prevent pulmonary infections.
Collapse
Affiliation(s)
- Huihui Zhang
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Fang He
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Pan Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | | | - Nengzhang Li
- College of Animal Medicine, Southwest University, Chongqing, China
| | - Yuanyi Peng
- College of Animal Medicine, Southwest University, Chongqing, China
| |
Collapse
|
42
|
Shastri MD, Chong WC, Dua K, Peterson GM, Patel RP, Mahmood MQ, Tambuwala M, Chellappan DK, Hansbro NG, Shukla SD, Hansbro PM. Emerging concepts and directed therapeutics for the management of asthma: regulating the regulators. Inflammopharmacology 2020; 29:15-33. [PMID: 33152094 DOI: 10.1007/s10787-020-00770-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Asthma is a common, heterogeneous and serious disease, its prevalence has steadily risen in most parts of the world, and the condition is often inadequately controlled in many patients. Hence, there is a major need for new therapeutic approaches. Mild-to-moderate asthma is considered a T-helper cell type-2-mediated inflammatory disorder that develops due to abnormal immune responses to otherwise innocuous allergens. Prolonged exposure to allergens and persistent inflammation results in myofibroblast infiltration and airway remodelling with mucus hypersecretion, airway smooth muscle hypertrophy, and excess collagen deposition. The airways become hyper-responsive to provocation resulting in the characteristic wheezing and obstructed airflow experienced by patients. Extensive research has progressed the understanding of the underlying mechanisms and the development of new treatments for the management of asthma. Here, we review the basis of the disease, covering new areas such as the role of vascularisation and microRNAs, as well as associated potential therapeutic interventions utilising reports from animal and human studies. We also cover novel drug delivery strategies that are being developed to enhance therapeutic efficacy and patient compliance. Potential avenues to explore to improve the future of asthma management are highlighted.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Malik Q Mahmood
- Faculty of Health, School of Medicine, Deakin University, Melbourne, Australia
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Belfast, Northern Ireland, UK
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia. .,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia. .,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
43
|
Akama Y, Satoh-Takayama N, Kawamoto E, Ito A, Gaowa A, Park EJ, Imai H, Shimaoka M. The Role of Innate Lymphoid Cells in the Regulation of Immune Homeostasis in Sepsis-Mediated Lung Inflammation. Diagnostics (Basel) 2020; 10:diagnostics10100808. [PMID: 33053762 PMCID: PMC7600279 DOI: 10.3390/diagnostics10100808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Septic shock/severe sepsis is a deregulated host immune system response to infection that leads to life-threatening organ dysfunction. Lung inflammation as a form of acute lung injury (ALI) is often induced in septic shock. Whereas macrophages and neutrophils have been implicated as the principal immune cells regulating lung inflammation, group two innate lymphoid cells (ILC2s) have recently been identified as a new player regulating immune homeostasis. ILC2 is one of the three major ILC subsets (ILC1s, ILC2s, and ILC3s) comprised of newly identified innate immune cells. These cells are characterized by their ability to rapidly produce type 2 cytokines. ILC2s are predominant resident ILCs and, thereby, have the ability to respond to signals from damaged tissues. ILC2s regulate the immune response, and ILC2-derived type 2 cytokines may exert protective roles against sepsis-induced lung injury. This focused review not only provides readers with new insights into the signaling mechanisms by which ILC2s modulate sepsis-induced lung inflammation, but also proposes ILC2 as a novel therapeutic target for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yuichi Akama
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
- Correspondence: (Y.A.); (M.S.)
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan;
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
| | - Atsushi Ito
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Thoracic and Cardiovascular Surgery, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
| | - Hiroshi Imai
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Correspondence: (Y.A.); (M.S.)
| |
Collapse
|
44
|
Virus-Induced Asthma Exacerbations: SIRT1 Targeted Approach. J Clin Med 2020; 9:jcm9082623. [PMID: 32823491 PMCID: PMC7464235 DOI: 10.3390/jcm9082623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of asthma has increased worldwide. Asthma exacerbations triggered by upper respiratory tract viral infections remain a major clinical problem and account for hospital admissions and time lost from work. Virus-induced asthma exacerbations cause airway inflammation, resulting in worsening asthma and deterioration in the patients’ quality of life, which may require systemic corticosteroid therapy. Despite recent advances in understanding the cellular and molecular mechanisms underlying asthma exacerbations, current therapeutic modalities are inadequate for complete prevention and treatment of these episodes. The pathological role of cellular senescence, especially that involving the silent information regulator 2 homolog sirtuin (SIRT) protein family, has recently been demonstrated in stable and exacerbated chronic respiratory disease states. This review discusses the role of SIRT1 in the pathogenesis of bronchial asthma. It also discusses the role of SIRT1 in inflammatory cells that play an important role in virus-induced asthma exacerbations. Recent studies have hypothesized that SIRT1 is one of major contributors to cellular senescence. SIRT1 levels decrease in Th2 and non-Th2-related airway inflammation, indicating the role of SIRT1 in several endotypes and phenotypes of asthma. Moreover, several models have demonstrated relationships between viral infection and SIRT1. Therefore, targeting SIRT1 is a novel strategy that may be effective for treating virus-induced asthma exacerbations in the future.
Collapse
|
45
|
Schulz-Kuhnt A, Wirtz S, Neurath MF, Atreya I. Regulation of Human Innate Lymphoid Cells in the Context of Mucosal Inflammation. Front Immunol 2020; 11:1062. [PMID: 32655549 PMCID: PMC7324478 DOI: 10.3389/fimmu.2020.01062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since their identification as a unique cell population, innate lymphoid cells (ILCs) have revolutionized our understanding of immune responses, leaving their impact on multiple inflammatory and fibrotic pathologies without doubt. Thus, a tightly controlled regulation of local ILC numbers and their activity is of crucial importance. Even though this has been extensively studied in murine ILCs in the last few years, our knowledge of human ILCs is still lagging behind. Our review article will therefore summarize recent insights into the function of human ILCs and will particularly focus on their regulation under inflammatory conditions. The quality and intensity of ILC involvement into local immune responses at mucosal sites of the human body can potentially be modulated via three different axes: (1) activation of tissue-resident mature ILCs, (2) plasticity and local transdifferentiation of specific ILC subsets, and (3) tissue migration and accumulation of peripheral ILCs. Despite a still ongoing scientific effort in this field, already existing data on the fate of human ILCs under different pathologic conditions clearly indicate that all three of these mechanisms are of relevance for the clinical course of chronic inflammatory and autoimmune diseases and might likewise provide new target structures for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
46
|
Boberg E, Johansson K, Malmhäll C, Weidner J, Rådinger M. House Dust Mite Induces Bone Marrow IL-33-Responsive ILC2s and T H Cells. Int J Mol Sci 2020; 21:E3751. [PMID: 32466530 PMCID: PMC7312993 DOI: 10.3390/ijms21113751] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/05/2023] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) and their adaptive counterpart type 2 T helper (TH2) cells respond to interleukin-33 (IL-33) by producing IL-5, which is a crucial cytokine for eosinophil development in the bone marrow. The aim of this study was to determine if bone marrow ILC2s, TH cells, and eosinophils are locally regulated by IL-33 in terms of number and activation upon exposure to the common aeroallergen house dust mite (HDM). Mice that were sensitized and challenged with HDM by intranasal exposures induced eosinophil development in the bone marrow with an initial increase of IL5Rα+ eosinophil progenitors, following elevated numbers of mature eosinophils and the induction of airway eosinophilia. Bone marrow ILC2s, TH2, and eosinophils all responded to HDM challenge by increased IL-33 receptor (ST2) expression. However, only ILC2s, but not TH cells, revealed increased ST2 expression at the onset of eosinophil development, which significantly correlated with the number of eosinophil progenitors. In summary, our findings suggest that airway allergen challenges with HDM activates IL-33-responsive ILC2s, TH cells, and eosinophils locally in the bone marrow. Targeting the IL-33/ST2 axis in allergic diseases including asthma may be beneficial by decreasing eosinophil production in the bone marrow.
Collapse
Affiliation(s)
| | | | | | | | - Madeleine Rådinger
- Krefting Research Centre, Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (E.B.); (K.J.); (C.M.); (J.W.)
| |
Collapse
|
47
|
Schulz-Kuhnt A, Greif V, Hildner K, Knipfer L, Döbrönti M, Zirlik S, Fuchs F, Atreya R, Zundler S, López-Posadas R, Neufert C, Ramming A, Kiefer A, Grüneboom A, Strasser E, Wirtz S, Neurath MF, Atreya I. ILC2 Lung-Homing in Cystic Fibrosis Patients: Functional Involvement of CCR6 and Impact on Respiratory Failure. Front Immunol 2020; 11:691. [PMID: 32457736 PMCID: PMC7221160 DOI: 10.3389/fimmu.2020.00691] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/26/2020] [Indexed: 01/10/2023] Open
Abstract
Cystic fibrosis patients suffer from a progressive, often fatal lung disease, which is based on a complex interplay between chronic infections, locally accumulating immune cells and pulmonary tissue remodeling. Although group-2 innate lymphoid cells (ILC2s) act as crucial initiators of lung inflammation, our understanding of their involvement in the pathogenesis of cystic fibrosis remains incomplete. Here we report a marked decrease of circulating CCR6+ ILC2s in the blood of cystic fibrosis patients, which significantly correlated with high disease severity and advanced pulmonary failure, strongly implicating increased ILC2 homing from the peripheral blood to the chronically inflamed lung tissue in cystic fibrosis patients. On a functional level, the CCR6 ligand CCL20 was identified as potent promoter of lung-directed ILC2 migration upon inflammatory conditions in vitro and in vivo using a new humanized mouse model with light-sheet fluorescence microscopic visualization of lung-accumulated human ILC2s. In the lung, blood-derived human ILC2s were able to augment local eosinophil and neutrophil accumulation and induced a marked upregulation of pulmonary type-VI collagen expression. Studies in primary human lung fibroblasts additionally revealed ILC2-derived IL-4 and IL-13 as important mediators of this type-VI collagen-inducing effect. Taken together, the here acquired results suggest that pathologically increased CCL20 levels in cystic fibrosis airways induce CCR6-mediated lung homing of circulating human ILC2s. Subsequent ILC2 activation then triggers local production of type-VI collagen and might thereby drive extracellular matrix remodeling potentially influencing pulmonary tissue destruction in cystic fibrosis patients. Thus, modulating the lung homing capacity of circulating ILC2s and their local effector functions opens new therapeutic avenues for cystic fibrosis treatment.
Collapse
Affiliation(s)
- Anja Schulz-Kuhnt
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Vicky Greif
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Lisa Knipfer
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Michael Döbrönti
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Florian Fuchs
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Medicine 3, University Hospital of Erlangen, Erlangen, Germany
| | - Alexander Kiefer
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Anika Grüneboom
- Department of Medicine 3, University Hospital of Erlangen, Erlangen, Germany
| | - Erwin Strasser
- Department of Transfusion Medicine and Haemostaseology, University Hospital of Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
48
|
A pulmonary ILC3 niche promotes neonatal mucosal immunity to respiratory bacterial infection and is associated with postnatal lung development. Mucosal Immunol 2020; 13:385-387. [PMID: 32203064 DOI: 10.1038/s41385-020-0283-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/04/2023]
|
49
|
Regulation of Innate Lymphoid Cells in Acute Kidney Injury: Crosstalk between Cannabidiol and GILZ. J Immunol Res 2020; 2020:6056373. [PMID: 32185239 PMCID: PMC7060850 DOI: 10.1155/2020/6056373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022] Open
Abstract
Innate lymphoid cells (ILCs) have emerged as largely tissue-resident archetypal cells of the immune system. We tested the hypotheses that renal ischemia-reperfusion injury (IRI) is a contributing factor to polarization of ILCs and that glucocorticoid-induced leucine zipper (GILZ) and cannabidiol regulate them in this condition. Mice subjected to unilateral renal IRI were treated with the following agents before restoration of renal blood flow: cannabidiol, DMSO, transactivator of transcription- (TAT-) GILZ, or the TAT peptide. Thereafter, kidney cells were prepared for flow cytometry analyses. Sham kidneys treated with either cannabidiol or TAT-GILZ displayed similar frequencies of each subset of ILCs compared to DMSO or TAT, respectively. Renal IRI increased ILC1s and ILC3s but reduced ILC2s compared to the sham group. Cannabidiol or TAT-GILZ treatment of IRI kidneys reversed this pattern as evidenced by reduced ILC1s and ILC3s but increased ILC2s compared to their DMSO- or TAT-treated counterparts. While TAT-GILZ treatment did not significantly affect cells positive for cannabinoid receptors subtype 2 (CB2+), cannabidiol treatment increased frequency of both CB2+ and GILZ-positive (GILZ+) cells of IRI kidneys. Subsequent studies showed that IRI reduced GILZ+ subsets of ILCs, an effect less marked for ILC2s. Treatment with cannabidiol increased frequencies of each subset of GILZ+ ILCs, but the effect was more marked for ILC2s. Indeed, cannabidiol treatment increased CB2+ GILZ+ ILC2s. Collectively, the results indicate that both cannabidiol and GILZ regulate ILC frequency and phenotype, in acute kidney injury, and that the effects of cannabidiol likely relate to modulation of endogenous GILZ.
Collapse
|
50
|
Entwistle LJ, Gregory LG, Oliver RA, Branchett WJ, Puttur F, Lloyd CM. Pulmonary Group 2 Innate Lymphoid Cell Phenotype Is Context Specific: Determining the Effect of Strain, Location, and Stimuli. Front Immunol 2020; 10:3114. [PMID: 32038635 PMCID: PMC6987460 DOI: 10.3389/fimmu.2019.03114] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are enriched at mucosal sites, including the lung, and play a central role in type 2 immunity and maintaining tissue homeostasis. As a result, since their discovery in 2010, research into ILC2s has increased markedly. Numerous strategies have been used to define ILC2s by flow cytometry, often utilizing different combinations of surface markers despite their expression being variable and context-dependent. In this study, we sought to generate a comprehensive characterization of pulmonary ILC2s, identifying stable and context specific markers from different pulmonary compartments following different airway exposures in different strains of mice. Our analysis revealed that pulmonary ILC2 surface marker phenotype is heterogeneous and is influenced by mouse strain, pulmonary location, in vivo treatment/exposure and ex vivo stimulation. Therefore, we propose that a lineage negative cell expressing CD45 and Gata3 defines an ILC2, and subsequent surface marker expression should be used to describe their phenotype in context-specific scenarios.
Collapse
Affiliation(s)
- Lewis J Entwistle
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Lisa G Gregory
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Robert A Oliver
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - William J Branchett
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Franz Puttur
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Clare M Lloyd
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|