1
|
Chuang HC, Chang JH, Fan YY, Hsieh CL, Lee YL. Interleukin-38-overexpressing adenovirus infection in dendritic cell-based treatment enhances immunotherapy for allergic asthma via inducing Foxp3 + regulatory T cells. Biomed Pharmacother 2024; 181:117738. [PMID: 39667222 DOI: 10.1016/j.biopha.2024.117738] [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: 08/31/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024] Open
Abstract
Allergic asthma is a chronic disease tied to unusual immune reactions involving type 2 T helper (Th2) cells specific to allergens. Dendritic cells (DCs) play a crucial role in guiding T-cell responses. Regulatory T (Treg) cells have the ability to suppress effector T-cell responses, and interleukin (IL)-38 is involved in Treg cell differentiation. In this study, we explored impacts of IL-38 on the activation and function of DCs, and we then developed an IL-38-overexpressing adenovirus (Ad-IL38) to evaluate its effectiveness in treating allergic asthma in mice through the adaptive transfer of Ad-IL38-infected DCs (IL38-DCs). Treating lipopolysaccharide (LPS)-activated bone marrow-derived DCs with recombinant IL-38 reduced cluster of differentiation 80 (CD80), CD86, and major histocompatibility complex (MHC) II expressions and decreased IL-1β, IL-6, and tumor necrosis factor (TNF)-α while increasing IL-10 secretion. The simultaneous culture of these semi-mature DCs with allogeneic CD4+ T cells facilitated the production of Forkhead box protein P3-positive (Foxp3+) Treg cells. A transcriptomic analysis revealed downregulation of the Chil3, Inhba, and Ctgf genes that are crucial for regulating inflammatory responses and cytokine-mediated signaling pathways in IL-38-treated DCs. In an animal model of asthma, IL38-DC treatment effectively decreased levels of an ovalbumin (OVA)-specific immunoglobulin E (IgE) antibody in serum, attenuated the severity of airway hyperresponsiveness, reduced the production of Th2-type cytokines (IL-4, IL-5, and IL-13) and proinflammatory cytokines (IL-6 and TNF-α) in bronchoalveolar lavage fluid, lowered expressions of the Th2-related cytokines IL-25 and thymic stromal-derived lymphopoietin (TSLP) by lung epithelial cells, and mitigated airway inflammation. Notably, enhanced expression of Foxp3+ Treg cells was linked to increased mRNA levels of transforming growth factor (TGF)-β production in vivo. In conclusion, we comprehensively clarified the immunomodulatory effects of IL-38 on DCs and provide a new treatment with IL-38 genetically modified DCs for alleviating Th2-mediated allergic diseases.
Collapse
Affiliation(s)
- Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yi Fan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Liao Y, Zhang X, Tao S, Wang S, Huang Q, Tang P, Tang A, Yang P, Yang G. Endoplasmic Reticulum Stress Promotes Telomerase Reverse Transcriptase Expression Contributes to Development of Allergic Rhinitis. Am J Rhinol Allergy 2024; 38:384-395. [PMID: 39093621 DOI: 10.1177/19458924241269686] [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] [Indexed: 08/04/2024]
Abstract
BACKGROUND The Th2 cell polarization is a crucial factor in the pathogenesis of allergic diseases. The underlying mechanism requires further investigation. Telomerase has an immune-regulating ability. The aim of this study is to elucidate the association between telomerase and Th2 cell polarization in patients with allergic rhinitis (AR). METHODS CD4+ T cells were isolated from blood samples collected from AR patients and healthy control subjects. RNA sequencing was employed to analyze RNA samples extracted from CD4+ T cells. An AR mouse model was established using the ovalbumin-alum protocol. RESULTS High telomerase gene activity and high endoplasmic reticulum (ER) stress status were observed in CD4+ T-cells in patients with AR. Positive correlation between the telomerase reverse transcriptase (TERT) gene expression in CD4+ T cells and AR response in patients with AR. TERT facilitated the degradation of Foxp3 proteins in CD4+ T cells, resulting in the polarization of Th2 cells. Sensitization with the ovalbumin-alum protocol enhanced the Tert expression in CD4+ T cells by exacerbating ER stress. Conditional inhibition of the Tert or eukaryotic translation initiation factor 2-α (Eif2a) expression in CD4+ T cells effectively attenuated experimental AR in mice. CONCLUSIONS Elevated amounts of telomerase in CD4+ T cells were found in CD4+ T cells of subjects with AR. Telomerase promoted Th2 cell polarization by inducing Foxp3 protein degradation and promotes GATA3 activation. Inhibition of TERT or eIF2a alleviated experimental AR.
Collapse
Affiliation(s)
- Yun Liao
- Department of Otolaryngology, Longgang Central Hospital affiliated to Shenzhen Clinical College, Guangzhou University of Chinese Traditional Medicine, Shenzhen, China
| | - Xiwen Zhang
- Department of Otolaryngology, Longgang Central Hospital affiliated to Shenzhen Clinical College, Guangzhou University of Chinese Traditional Medicine, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Shuang Tao
- Department of Otolaryngology, Longgang Central Hospital affiliated to Shenzhen Clinical College, Guangzhou University of Chinese Traditional Medicine, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Shiqi Wang
- Department of Otolaryngology, Longgang Central Hospital affiliated to Shenzhen Clinical College, Guangzhou University of Chinese Traditional Medicine, Shenzhen, China
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Qinmiao Huang
- Department of General Practice Medicine and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Ping Tang
- Department of General Practice Medicine and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Aifa Tang
- Department of General Practice Medicine and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Pingchang Yang
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy & Immunology of Shenzhen University, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital affiliated to Shenzhen Clinical College, Guangzhou University of Chinese Traditional Medicine, Shenzhen, China
| |
Collapse
|
3
|
Rong Y, Tang M, Liu L, Ma X, Liu M, Qu L, Liao X, Jiang Q, Zhang N, Xu X. Artemisia argyi essential oil alleviates asthma by regulating 5-LOX-CysLTs and IDO-1-KYN pathways: Insights from metabolomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118458. [PMID: 38871010 DOI: 10.1016/j.jep.2024.118458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia argyi essential oil (AAEO) is a traditional herbal remedy for asthma. However, the potential effect of AAEO on asthma has not been elucidated. AIM OF THE STUDY To investigate the protective properties of AAEO upon asthma and elucidate its mechanism. MATERIALS AND METHODS The effects of AAEO in asthma were assessed by histology and biochemical analysis. Then, we integrated real-time reverse transcription-quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, immunohistochemistry and metabolomics analysis to reveal its mechanism. RESULTS In vivo, AAEO reduced the counts of white blood cells (WBCs) and cytokines in bronchoalveolar lavage fluid (BALF), ameliorated pathologic alterations in lung tissues, and inhibited secretion of OVA-sIgE and muc5ac. Metabolomics results showed that AAEO can exert therapeutic effects on asthmatic mice by regulating disordered arachidonic acid metabolism and tryptophan metabolism. Further studies shown that AAEO inhibited the expression of 5-LOX and reduced the accumulation of CysLTs in mice. Meanwhile, AAEO promoted the activity of IDO-1, facilitated the conversion of tryptophan to kynurenine, and regulated the imbalance of Treg/Th17 immunity. Immunohistochemical results showed that AAEO promoted the expression of IDO-1. RT-qPCR results showed that AAEO promoted the expression of IL-10 and Foxp3 mRNA, and inhibited the expression of IL-17A and RORγt mRNA, thus regulated the imbalance of Treg/Th17 immunity and exerted its therapeutic effects. CONCLUSION AAEO treatment not only attenuates the clinical symptoms of asthma but is also involved in regulating lung tissue metabolism. The anti-asthmatic activity of AAEO may be achieved by reprogramming 5-LOX-CysLTs and IDO-1-KYN pathways.
Collapse
Affiliation(s)
- Ying Rong
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Mengqi Tang
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Luyao Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Xiaoge Ma
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Mengge Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Lingbo Qu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Xinglin Liao
- Nanyang LANHAISENYUAN Medical Technology Ltd.,CO, Nanyang, Henan, 473000, PR China
| | - Qiman Jiang
- Nanyang LANHAISENYUAN Medical Technology Ltd.,CO, Nanyang, Henan, 473000, PR China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China.
| | - Xia Xu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| |
Collapse
|
4
|
Wang Y, Zhang L, Shi B, Luo J. Trends and research foci in immunoregulatory mechanisms of allergic rhinitis: a bibliometric analysis (2014-2024). Front Immunol 2024; 15:1443954. [PMID: 39380999 PMCID: PMC11458462 DOI: 10.3389/fimmu.2024.1443954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Background This study aims to provide a comprehensive bibliometric analysis of research trends, hotspots, and future directions in the immunoregulatory mechanisms of allergic rhinitis (AR) from 2014 to 2024. Methods Data were sourced from the Web of Science Core Collection (WoSCC), covering articles and reviews published between April 1, 2014, and March 31, 2024. The search terms included "Allergic Rhinitis," "AR," and related terms along with specific keywords related to immune cells and inflammatory mediators. Bibliometric tools such as CiteSpace, VOSviewer, and SCImago Graphica were used to analyze institutional cooperation networks, keyword co-occurrence, citation bursts, and research topic evolution. Microsoft Excel 2019 was employed to display annual publication trends. Results A total of 2200 papers met the inclusion and exclusion criteria. The number of publications showed an upward trend over the past decade, with a significant peak in 2021. China (583 papers) and the United States (454 papers) were the major contributing countries. Imperial College London emerged as the leading institution. Key research frontiers identified include the roles of NF kappa B and air pollution in AR. Keyword burst analysis revealed emerging topics such as respiratory allergy and personalized treatment strategies. Notable limitations include the exclusive use of the WoSCC database and the restriction to English-language publications. Conclusion The field of immunoregulatory mechanisms in allergic rhinitis has seen significant growth, with China and the United States leading the research. Future research should focus on developing personalized treatment plans and understanding the comprehensive impact of environmental factors. Continued interdisciplinary collaboration and international cooperation will be essential for advancing therapeutic strategies in AR.
Collapse
Affiliation(s)
- Yandan Wang
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Liangran Zhang
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Baoyuan Shi
- Department of Otolaryngology, Huaihe Hospital, Henan University, Kaifeng, China
| | - Junpeng Luo
- Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| |
Collapse
|
5
|
Dong C, Lin JM, Wang Y, Zhu J, Lin L, Xu J, Du J. Exploring the Common Pathogenic Mechanisms of Psoriasis and Atopic Dermatitis: The Interaction between SGK1 and TIGIT Signaling Pathways. Inflammation 2024:10.1007/s10753-024-02115-1. [PMID: 39088121 DOI: 10.1007/s10753-024-02115-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
This study aims to explore the common pathogenic mechanisms of psoriasis and atopic dermatitis, two T-cell-mediated autoimmune diseases. Utilizing single-cell transcriptomic sequencing data, we revealed that Treg cells primarily express TIGIT in both psoriasis and atopic dermatitis, and identified a subset of macrophages that highly express SGK1. These cells can interact with T cells via the NECTIN2-TIGIT signaling pathway, inhibiting the differentiation of T cells into a pro-inflammatory phenotype, thereby uncovering a common immunoregulatory mechanism in both diseases. Furthermore, we discovered that inhibition of SGK1 exacerbates the inflammatory response in disease models of both conditions. These findings not only provide a new perspective for a common therapeutic strategy for psoriasis and atopic dermatitis but also highlight the importance of considering these molecular interactions in future treatments. Validation of these observations through further qPCR, immunofluorescence, and animal studies has identified potential new targets for the treatment of psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Canbin Dong
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jui-Ming Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Yilun Wang
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Junhao Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Lanmei Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jinhua Xu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Juan Du
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
6
|
Xue J, Liu Z, Liao Y, Zhang X, Liu Y, Mo L, Dong R, Li Q, Sun X, Xie J, Yang P. Undersized telomeres in regulatory T cells link to the pathogenesis of allergic rhinitis. iScience 2024; 27:108615. [PMID: 38205251 PMCID: PMC10777067 DOI: 10.1016/j.isci.2023.108615] [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: 09/05/2023] [Revised: 10/23/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024] Open
Abstract
Telomeres are an important biomarker in the cell destiny. The relationship between telomeres and regulatory T cells (Tregs) has not yet been investigated. The objective of this study is to evaluate the link between Tregs' telomere length and allergic rhinitis (AR)'s pathogenesis. Here, we report that low telomerase activity and high endoplasmic reticulum stress status were observed in Tregs from AR patients, as shown in the results. Immune regulatory molecules levels were correlated with the length of Tregs' telomeres. The immune-suppressive functions of Tregs were associated with the telomere length/Telomerase reverse transcriptase/Telomerase protein component 1 status in Tregs. The levels of telomere length/telomerase in airway Tregs were reduced by sensitization. Endoplasmic reticulum stress signaling pathway of proline-rich receptor-like protein kinase-eukaryotic translation initiation factor 2A (eIF2a) was associated with the regulation of telomerase. Inhibiting eIF2a had an effect on upregulating telomerase activity in Tregs and mitigating experimental AR.
Collapse
Affiliation(s)
- Jinmei Xue
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education of China, Shanxi Medical University, Taiyuan, China
| | - Yun Liao
- Shenzhen Clinical School of Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Xiwen Zhang
- Shenzhen Clinical School of Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Yu Liu
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Lihua Mo
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Rui Dong
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Qiang Li
- Department of Otolaryngology, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Xizhuo Sun
- Department of General Practice Medicine, Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education of China, Shanxi Medical University, Taiyuan, China
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
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: 65] [Impact Index Per Article: 32.5] [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
|
8
|
Moratin H, Thöle A, Lang J, Ehret Kasemo T, Stöth M, Hagen R, Scherzad A, Hackenberg S. Ag- but Not ZnO-Nanoparticles Disturb the Airway Epithelial Barrier at Subtoxic Concentrations. Pharmaceutics 2023; 15:2506. [PMID: 37896266 PMCID: PMC10610507 DOI: 10.3390/pharmaceutics15102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Inhalation is considered to be the most relevant source of human exposure to nanoparticles (NPs); however, only a few investigations have addressed the influence of exposing the respiratory mucosal barrier to subcytotoxic doses. In the nasal respiratory epithelium, cells of the mucosa represent one of the first contact points of the human organism with airborne NPs. Disruption of the epithelial barrier by harmful materials can lead to inflammation in addition to potential intrinsic toxicity of the particles. The aim of this study was to investigate whether subtoxic concentrations of zinc oxide (ZnO)- and silver (Ag)-NPs have an influence on upper airway barrier integrity. Nasal epithelial cells from 17 donors were cultured at the air-liquid interface and exposed to ZnO- and Ag-NPs. Barrier function, quantified by transepithelial electrical resistance (TEER), decreased after treatment with 10 µg/mL Ag-NPs, but FITC-dextran permeability remained stable and no change in mRNA levels of tight junction proteins and E-cadherin was detected by real-time quantitative PCR (RT-qPCR). The results indicate that subtoxic concentrations of Ag-NPs may already induce damage of the upper airway epithelial barrier in vitro. The lack of similar disruption by ZnO-NPs of similar size suggests a specific effect by Ag-NPs.
Collapse
Affiliation(s)
- Helena Moratin
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080 Wuerzburg, Germany (S.H.)
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Kolkhir P, Akdis CA, Akdis M, Bachert C, Bieber T, Canonica GW, Guttman-Yassky E, Metz M, Mullol J, Palomares O, Renz H, Ständer S, Zuberbier T, Maurer M. Type 2 chronic inflammatory diseases: targets, therapies and unmet needs. Nat Rev Drug Discov 2023; 22:743-767. [PMID: 37528191 DOI: 10.1038/s41573-023-00750-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 08/03/2023]
Abstract
Over the past two decades, significant progress in understanding of the pathogenesis of type 2 chronic inflammatory diseases has enabled the identification of compounds for more than 20 novel targets, which are approved or at various stages of development, finally facilitating a more targeted approach for the treatment of these disorders. Most of these newly identified pathogenic drivers of type 2 inflammation and their corresponding treatments are related to mast cells, eosinophils, T cells, B cells, epithelial cells and sensory nerves. Epithelial barrier defects and dysbiotic microbiomes represent exciting future drug targets for chronic type 2 inflammatory conditions. Here, we review common targets, current treatments and emerging therapies for the treatment of five major type 2 chronic inflammatory diseases - atopic dermatitis, chronic prurigo, chronic urticaria, asthma and chronic rhinosinusitis with nasal polyps - with a high need for targeted therapies. Unmet needs and future directions in the field are discussed.
Collapse
Affiliation(s)
- Pavel Kolkhir
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos, University of Zürich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos, University of Zürich, Davos, Switzerland
| | - Claus Bachert
- Department of Otorhinolaryngology - Head and Neck Surgery, University Hospital of Münster, Münster, Germany
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Division of ENT diseases, Karolinska Hospital, Stockholm, Sweden
| | - Thomas Bieber
- Department of Dermatology and Allergy, University Hospital, Bonn, Germany
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
- Davos Biosciences, Davos, Switzerland
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Asthma & Allergy Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Martin Metz
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clínic Barcelona, FRCB-IDIBAPS, Universitat de Barcelona, CIBERES, Barcelona, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Harald Renz
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
- Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Sonja Ständer
- Section Pruritus Medicine, Department of Dermatology and Center for Chronic Pruritus, University Hospital Münster, Münster, Germany
| | - Torsten Zuberbier
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| |
Collapse
|
10
|
Nahm DH. Regulatory T Cell-Targeted Immunomodulatory Therapy for Long-Term Clinical Improvement of Atopic Dermatitis: Hypotheses and Perspectives. Life (Basel) 2023; 13:1674. [PMID: 37629531 PMCID: PMC10455293 DOI: 10.3390/life13081674] [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: 06/29/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Atopic dermatitis (AD) is a chronically relapsing inflammatory skin disorder characterized by itching and eczematous lesions. It is often associated with a personal or familial history of allergic diseases. Allergic inflammation induced by immunoglobulin E and T-helper type 2 (Th2) cell responses to common environmental agents has been suggested to play an essential role in AD pathogenesis. The standard therapies for AD, including topical or systemic agents, focus on controlling skin inflammation. Recently developed monoclonal antibody to interleukin-4 receptor alpha or Janus kinase inhibitors can provide significant clinical improvements in patients with AD by inhibiting Th2 cell-mediated skin inflammation. However, the clinical efficacy of the Th2 cell-targeted therapy is transient and incomplete in patients with AD. Patients with AD are seeking a permanent cure. Therefore, the development of novel immunomodulatory strategies that can improve a long-term clinical outcome and provide a long-term treatment-free clinical remission of AD (disease-modifying therapy) is needed. Regulatory T (Treg) cells play a critical role in the maintenance of immune tolerance and suppress the development of autoimmune and allergic diseases. This review provides three working hypotheses and perspectives for the treatment of AD by Treg cell activation. (1) A decreased number or function of Treg cells is a critical event that causes the activation of Th2 cells, leading to the development and maintenance of AD. (2) Activation of Treg cells is an effective therapeutic approach for AD. (3) Many different immunomodulatory strategies activating Treg cells can provide a long-term clinical improvement of AD by induction of immune tolerance. The Treg cell-targeted immunomodulatory therapies for AD include allergen immunotherapy, microbiota, vitamin D, polyvalent human immunoglobulin G, monoclonal antibodies to the surface antigens of T cell or antigen-presenting cell, and adoptive transfer of autologous Treg cells or genetically engineered Treg cells expanded in vitro.
Collapse
Affiliation(s)
- Dong-Ho Nahm
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
11
|
Chang JH, Chuang HC, Fan CK, Hou TY, Chang YC, Lee YL. Norisoboldine exerts antiallergic effects on IgE/ovalbumin-induced allergic asthma and attenuates FcεRI-mediated mast cell activation. Int Immunopharmacol 2023; 121:110473. [PMID: 37331292 DOI: 10.1016/j.intimp.2023.110473] [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: 02/23/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/20/2023]
Abstract
Allergic asthma is an inflammatory lung disorder, and mast cells play crucial roles in the development of this allergic disease. Norisoboldine (NOR), the major isoquinoline alkaloid present in Radix Linderae, has received considerable attention because it has anti-inflammatory effects. Herein, the aim of this study was to explore the antiallergic effects of NOR on allergic asthma in mice and mast cell activation. In a murine model of ovalbumin (OVA)-induced allergic asthma, oral administration at 5 mg/kg body weight (BW) of NOR produced strong reductions in serum OVA-specific immunoglobulin E (IgE) levels, airway hyperresponsiveness, and bronchoalveolar lavage fluid (BALF) eosinophilia, while an increase in CD4+Foxp3+ T cells of the spleen was detected. Histological studies demonstrated that NOR treatment significantly ameliorated the progression of airway inflammation including the recruitment of inflammatory cells and mucus production by decreasing levels of histamine, prostaglandin D2 (PGD2), interleukin (IL)-4, IL-5, IL-6, and IL-13 in BALF. Furthermore, our results revealed that NOR (3 ∼ 30 μM) dose-dependently reduced expression of the high-affinity receptor for IgE (FcεRI) and the production of PGD2 and inflammatory cytokines (IL-4, IL-6, IL-13, and TNF-α), and also decreased degranulation of bone marrow-derived mast cells (BMMCs) activated by IgE/OVA. In addition, a similar suppressive effect on BMMC activation was observed by inhibition of the FcεRI-mediated c-Jun N-terminal kinase (JNK) signaling pathway using SP600125, a selective JNK inhibitor. Collectively, these results suggest that NOR may have therapeutic potential for allergic asthma at least in part through regulating the degranulation and the release of mediators by mast cells.
Collapse
Affiliation(s)
- Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chia-Kwung Fan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Yun Hou
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Cheng Chang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
12
|
Miwa T, Takemiya Y, Amesara K, Kawai H, Teranishi Y. Arginine-Rich Cell-Penetrating Peptide-Mediated Transduction of Mouse Nasal Cells with FOXP3 Protein Alleviates Allergic Rhinitis. Pharmaceutics 2023; 15:1770. [PMID: 37376217 PMCID: PMC10303077 DOI: 10.3390/pharmaceutics15061770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Intranasal corticosteroids are effective medications against allergic rhinitis (AR). However, mucociliary clearance promptly eliminates these drugs from the nasal cavity and delays their onset of action. Therefore, a faster, longer-lasting therapeutic effect on the nasal mucosa is required to enhance the efficacy of AR management. Our previous study showed that polyarginine, a cell-penetrating peptide, can deliver cargo to nasal cells; moreover, polyarginine-mediated cell-nonspecific protein transduction into the nasal epithelium exhibited high transfection efficiency with minimal cytotoxicity. In this study, poly-arginine-fused forkhead box P3 (FOXP3) protein, the "master transcriptional regulator" of regulatory T cells (Tregs), was administered into the bilateral nasal cavities of the ovalbumin (OVA)-immunoglobulin E mouse model of AR. The effects of these proteins on AR following OVA administration were investigated using histopathological, nasal symptom, flow cytometry, and cytokine dot blot analyses. Polyarginine-mediated FOXP3 protein transduction induced Treg-like cell generation in the nasal epithelium and allergen tolerance. Overall, this study proposes FOXP3 activation-mediated Treg induction as a novel and potential therapeutic strategy for AR, providing a potential alternative to conventional intranasal drug application for nasal drug delivery.
Collapse
Affiliation(s)
- Toru Miwa
- Department of Otolaryngology, Osaka Metropolitan University, Osaka 5458585, Japan; (Y.T.); (K.A.); (H.K.); (Y.T.)
- Department of Otolaryngology-Head and Neck Surgery, Kyoto University, Kyoto 6577575, Japan
| | - Yumi Takemiya
- Department of Otolaryngology, Osaka Metropolitan University, Osaka 5458585, Japan; (Y.T.); (K.A.); (H.K.); (Y.T.)
| | - Kazuki Amesara
- Department of Otolaryngology, Osaka Metropolitan University, Osaka 5458585, Japan; (Y.T.); (K.A.); (H.K.); (Y.T.)
| | - Hiroko Kawai
- Department of Otolaryngology, Osaka Metropolitan University, Osaka 5458585, Japan; (Y.T.); (K.A.); (H.K.); (Y.T.)
| | - Yuichi Teranishi
- Department of Otolaryngology, Osaka Metropolitan University, Osaka 5458585, Japan; (Y.T.); (K.A.); (H.K.); (Y.T.)
| |
Collapse
|
13
|
Wang T, Qiao W, Xie Y, Ma J, Hu W, Yang L, Li X, Duan C, Wu S, Wang Y, Cheng K, Zhang Y, Zhuang R. CD226 deficiency exacerbated intestinal immune dysregulation in mice with dinitrochlorobenzene-induced atopic dermatitis. Immunology 2023. [PMID: 36938934 DOI: 10.1111/imm.13640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/16/2023] [Indexed: 03/21/2023] Open
Abstract
Intestinal mucosal immunity plays a pivotal role in host defence. In this study, we found that cluster of differentiation 226 (CD226) gene knockout (KO) led to more severe atopic dermatitis (AD)-related skin pathologies and bowel abnormalities in a 2,4-dinitrochlorobenzene (DNCB)-induced AD-like mouse model. Following DNCB administration, the expression of CD226 was elevated in intestinal mucosal tissues, including group 3 innate lymphoid cells (ILC3s) and CD4+ T cells of Peyer's patches (PPs). CD226 deficiency led to an overactive intestinal immune response in the AD-like mice, as evidenced by increased inflammation and Th1/Th2-related cytokine levels as well as increased Paneth cell numbers and antimicrobial peptide (AMP) expression, which was likely due to the higher interleukin (IL)-22 production in the lamina propria. Additionally, CD226 deficiency increased the production of IL-4 and IL-17 in mesenteric lymph nodes as well as the number of PPs and expression of immunoglobulin (Ig) A in B cells. Moreover, insufficient expression of CD226 affected the characterization of intraepithelial and lamina propria lymphocytes in the intestinal mucosa. Finally, the number of PPs was increased in CD4+ T cell-specific CD226 KO and regulatory T (Treg) cell-specific CD226 KO mice; thus, loss of CD226 in Treg cells resulted in impaired Treg cell-suppressive function. Therefore, our findings indicate that CD226 deficiency alters intestinal immune functionality in inflammatory diseases.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Qiao
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yang Xie
- Department of Otolaryngological, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Hu
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lu Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuemei Li
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuwen Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuan Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
14
|
Zheng H, Dai H, Yan X, Xiang Q. Study on Intestinal Flora and Asthma: Knowledge Graph Analysis Based on CiteSpace (2001-2021). J Asthma Allergy 2023; 16:355-364. [PMID: 37041761 PMCID: PMC10083019 DOI: 10.2147/jaa.s402883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/12/2023] [Indexed: 04/13/2023] Open
Abstract
Asthma is a common chronic inflammatory disease of the airway. Intestinal flora, a significant risk factor for asthma, has become a widespread concern in the pathogenesis of asthma. To review the literature related to intestinal flora in asthma, summarize research direction, and report trends, this study used CiteSpace to perform bibliometric statistics and analysis on the research papers of intestinal flora and asthma collected in the Web of science core collection from 2001 to 2021. Eventually, a total of 613 articles were included. The results demonstrated that research on gut flora and asthma continued to heat up, with article numbers increasing, especially in the last decade. Moreover, analysis of the keywords showed that the research topics of intestinal flora and asthma range from confirming the link between intestinal flora and asthma to investigating mechanisms and then to asthma treatment. According to the summary of research hotspots, we expand on three emerging issues that require attention in the intestinal flora and asthma research, including (regulatory T)Treg cells, probiotics, and chain fatty acid. Evidence illustrated that Treg cells play a crucial role in the pathogenesis of asthma caused by dysbiosis of the gut flora. Furthermore, in contrast to probiotic supplements, which do not reduce the risk of developing asthma, short-chain fatty acids supplements do. Overall, the research direction in the field of intestinal flora and asthma has recently evolved from macro to micro with depth broadened. As a robust scientific evaluation, our study provided a comprehensive overview of the area, particularly for research focus, which could more precisely direct scholars on future research and clinical diagnosis, therapy, and individualized prevention.
Collapse
Affiliation(s)
- Hang Zheng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Xiumei Yan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Qiangwei Xiang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Correspondence: Qiangwei Xiang, Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China, Email
| |
Collapse
|
15
|
Agache I, Zemelka-Wiącek M, Shamji MH, Jutel M. Immunotherapy: State-of-the-art review of therapies and theratypes. J Allergy Clin Immunol 2022; 150:1279-1288. [PMID: 36328808 DOI: 10.1016/j.jaci.2022.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Through its disease-modifying potential, immunotherapy is the keystone to curing allergic diseases. Allergen immunotherapy, applied for more than a century, is currently supported by novel modalities such as mAb-based therapies or small molecules targeting the key nodes of the allergic inflammation network. In this review, a summary of the most significant advances in immunotherapy is presented, addressing not only novel approaches to stratifying patients but also major controlled clinical trials and real-world evidence that strengthen the role of immunotherapy in the treatment of allergies.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania.
| | | | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland; ALL-MED Medical Research Institute, Wroclaw, Poland
| |
Collapse
|
16
|
Jiao WE, Xu S, Qiao YL, Kong YG, Sun L, Deng YQ, Yang R, Tao ZZ, Hua QQ, Chen SM. Notch2-dependent GATA3+ Treg cells alleviate allergic rhinitis by suppressing the Th2 cell response. Int Immunopharmacol 2022; 112:109261. [PMID: 36155282 DOI: 10.1016/j.intimp.2022.109261] [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: 07/30/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
The aim of this study was to investigate the role and mechanism of Notch2-dependent GATA3+ Treg cells in allergic rhinitis (AR). Samples were collected from patients in the control and AR groups to detect differences in the numbers of GATA3+ Treg cells and their intracellular Notch2 levels. The effects of Notch2 on GATA3+ Treg cell differentiation and function in vitro were detected. AR mice were subjected to adoptive transfer of GATA3+ Treg cells to detect changes in the allergic inflammatory response and Th2 cells. Mice with Treg cell-specific knockout of Notch2 were constructed, and an AR model was established to detect the changes. The number of GATA3+ Treg cells and intracellular Notch2 expression in peripheral blood of the AR group were decreased compared with the controls (P < 0.05), and the number of GATA3+ Treg cells was significantly negatively correlated with the level of allergen-specific IgE (sIgE; P < 0.01). In vitro experiments showed that Notch2 promoted the differentiation and immunosuppressive function of GATA3+ Treg cells, and Notch2 directly promoted GATA3 transcription in Treg cells (P < 0.05). Animal experiments indicated that adoptive transfer of GATA3+ Treg cells reduced the allergic inflammatory response in AR mice (P < 0.05). The number of GATA3+ Treg cells was decreased in gene knockout mice (P < 0.05), and autoimmune inflammation was observed. After modeling, the allergic inflammatory response was further aggravated (P < 0.05). Overall, our findings indicate that Notch2 alleviates AR by specifically increasing GATA3+ Treg cell differentiation. Notch2 expressed in Treg cells is expected to be a new therapeutic target for AR.
Collapse
Affiliation(s)
- Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Liu Sun
- Department of Otolaryngology-Head and Neck Surgery, General Hospital of The Central Theater Command, Wuhan 430070, Hubei, PR China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Qing-Quan Hua
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|