Journal of Shandong University (Health Sciences) ›› 2024, Vol. 62 ›› Issue (5): 1-6.doi: 10.6040/j.issn.1671-7554.0.2024.0121

• Precision medicine in chronic airway diseases—Expert Overview •    

Airway epithelium and epithelial-derived cytokines in asthma: reflection and outlook

ZHANG Jintao, DONG Liang   

  1. Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Disease, Jinan 250014, Shandong, China
  • Published:2024-05-29

Abstract: The airway epithelium plays a central role in the pathogenesis of asthma by tightly interacting with immune cells and finely regulating the formation of the airway microenvironment. Epithelial-derived cytokines have been recognized as key players in triggering and sustaining airway inflammation in asthma, making them attractive targets for the development of novel asthma drugs. This article provides an overview of the role of the airway epithelium and its derived cytokines in asthma, as well as the progress in research on targeted drugs, offering new perspectives and insights for related studies.

Key words: Airway epithelium, Cytokines, Asthma, Targeted medicine, Outlook

CLC Number: 

  • R562
[1] Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases[J]. Eur J Immunol, 2024, 54(1): e2249982. doi: 10.1002/eji.202249982.
[2] Noureddine N, Chalubinski M, Wawrzyniak P. The role of defective epithelial barriers in allergic lung disease and asthma development[J]. J Asthma Allergy, 2022, 15: 487-504. doi: 10.2147/JAA.S324080.
[3] Chen CY, Wu KH, Guo BC, et al. Personalized medicine in severe asthma: from biomarkers to biologics[J]. Int J Mol Sci, 2023, 25(1): 182. doi: 10.3390/ijms25010182.
[4] Hellings PW, Steelant B. Epithelial barriers in allergy and asthma[J]. J Allergy Clin Immunol, 2020, 145(6): 1499-1509.
[5] Vieira Braga FA, Kar G, Berg M, et al. A cellular census of human lungs identifies novel cell states in health and in asthma[J]. Nat Med, 2019, 25(7): 1153-1163.
[6] Zhang N, Xu J, Jiang C, et al.Neuro-immune regulation in inflammation and airway remodeling of allergic asthma[J]. Front Immunol, 2022, 13: 894047. doi: 10.3389/fimmu.2022.894047.
[7] Kohanski MA, Workman AD, Patel NN, et al. Solitary chemosensory cells are a primary epithelial source of IL-25 in patients with chronic rhinosinusitis with nasal polyps[J]. J Allergy Clin Immunol, 2018, 142(2): 460-469.e7.
[8] Waghray A, Monga I, Lin B, et al. A deep lung cell atlas reveals cytokine-mediated lineage switching of a rare cell progenitor of the human airway epithelium[J]. bioRxiv, 2023. doi: 10.1101/2023.11.28.569028.
[9] Gauvreau GM, Obyrne PM, Boulet LP, et al. Effects of an anti-TSLP antibody on allergen-induced asthmatic responses[J]. N Engl J Med, 2014, 370(22): 2102-2110.
[10] Barlow JL, Peel S, Fox J, et al. IL-33 is more potent than IL-25 in provoking IL-13-producing nuocytes(type 2 innate lymphoid cells)and airway contraction[J]. J Allergy Clin Immunol, 2013, 132(4): 933-941.
[11] Duchesne M, Okoye I, Lacy P. Epithelial cell alarmin cytokines: frontline mediators of the asthma inflammatory response[J]. Front Immunol, 2022, 13: 975914. doi: 10.3389/fimmu.2022.975914.
[12] Liu T, Liu Y, Miller M, et al. Autophagy plays a role in FSTL1-induced epithelial mesenchymal transition and airway remodeling in asthma[J]. Am J Physiol Lung Cell Mol Physiol, 2017, 313(1): L27-L40.
[13] Zhang J, Zhang D, Pan Y, et al. The TL1A-DR3 axis in asthma: membrane-bound and secreted TL1A co-determined the development of airway remodeling[J]. Allergy Asthma Immunol Res, 2022, 14(2): 233-253.
[14] Liu F, Zhang J, Zhang D, et al. Follistatin-related protein 1 in asthma: miR-200b-3p interactions affect airway remodeling and inflammation phenotype[J]. Int Immunopharmacol, 2022, 109: 108793. doi: 10.1016/j.intimp.2022.108793.
[15] Guo Z, Wu J, Zhao J, et al. IL-33 promotes airway remodeling and is a marker of asthma disease severity[J]. J Asthma, 2014, 51(8): 863-869.
[16] Cao L, Liu F, Liu Y, et al. TSLP promotes asthmatic airway remodeling via p38-STAT3 signaling pathway in human lung fibroblast[J]. Exp Lung Res, 2018, 44(6): 288-301.
[17] Zhang J, Dong L. Status and prospects: personalized treatment and biomarker for airway remodeling in asthma[J]. J Thorac Dis, 2020, 12(10): 6090-6101.
[18] Andreasson LM, Dyhre-Petersen N, Hvidtfeldt M, et al. Airway hyperresponsiveness correlates with airway TSLP in asthma independent of eosinophilic inflammation[J]. J Allergy Clin Immunol, 2023: S0091-6749(23)02409-0. doi: 10.1016/j.jaci.2023.11.915.
[19] Chatziparasidis G, Bush A, Chatziparasidi MR, et al. Airway epithelial development and function: a key player in asthma pathogenesis?[J]. Paediatr Respir Rev, 2023, 47: 51-61. doi: 10.1016/j.prrv.2023.04.005.
[20] Frey A, Lunding LP, Ehlers JC, et al. More than just a barrier: the immune functions of the airway epithelium in asthma pathogenesis[J]. Front Immunol, 2020, 11: 761. doi: 10.3389/fimmu.2020.00761.
[21] Basil MC, Katzen J, Engler AE, et al. The cellular and physiological basis for lung repair and regeneration: past, present, and future[J]. Cell Stem Cell, 2020, 26(4): 482-502.
[22] Basil MC, Cardenas-Diaz FL, Kathiriya JJ, et al. Human distal airways contain a multipotent secretory cell that can regenerate alveoli[J]. Nature, 2022, 604(7904): 120-126.
[23] Smolinska S, Antolín-Amérigo D, Popescu FD, et al. Thymic stromal lymphopoietin(TSLP), its isoforms and the interplay with the epithelium in allergy and asthma[J]. Int J Mol Sci, 2023, 24(16): 12725. doi: 10.3390/ijms241612725.
[24] Chen W, Chen S, Yan C, et al. Allergen protease-activated stress granule assembly and gasdermin D fragmentation control interleukin-33 secretion[J]. Nat Immunol, 2022, 23(7): 1021-1030.
[25] Zhang D, Zhang J, Xu C, et al. A humanized mouse model to study asthmatic airway remodeling and Muc-5ac secretion via the human IL-33[J]. Allergy, 2024. 79(5): 1364-1367.
[26] Qi Q, Xu J, Wang Y, et al. Decreased sphingosine due to down-regulation of acid ceramidase expression in airway of bronchiectasis patients: a potential contributor to pseudomonas aeruginosa infection[J]. Infect Drug Resist, 2023, 16: 2573-2588. doi: 10.2147/IDR.S407335.
[27] Saikumar Jayalatha AK, Jonker MR, Carpaij OA, et al. Lack of a transcriptional response of primary bronchial epithelial cells from patients with asthma and controls to IL-33[J]. Am J Physiol Lung Cell Mol Physiol, 2024, 326(1): L65-L70.
[28] Ruhl A, Antão AV, Dietschmann A, et al. STAT6-induced production of mucus and resistin-like molecules in lung Club cells does not protect against helminth or influenza A virus infection[J]. Eur J Immunol, 2024, 54(1): e2350558. doi: 10.1002/eji.202350558.
[29] Li Y, Zhang Q, Li L, et al. LKB1 deficiency upregulates RELM-α to drive airway goblet cell metaplasia[J]. Cell Mol Life Sci, 2021, 79(1): 42. doi: 10.1007/s00018-021-04044-w.
[30] Kortekaas RK, Geillinger-Kästle KE, Borghuis T, et al. Interleukin-11 disrupts alveolar epithelial progenitor function[J]. ERJ Open Res, 2023, 9(3): 00679-2022. doi: 10.1183/23120541.00679-2022.
[31] Thomas D, Mcdonald VM, Pavord ID, et al. Asthma remission: what is it and how can it be achieved?[J]. Eur Respir J, 2022, 60(5): 2102583. doi: 10.1183/13993003.02583-2021.
[32] Mümmler C, Milger K. Biologics for severe asthma and beyond[J]. Pharmacol Ther, 2023, 252: 108551. doi: 10.1016/j.pharmthera.2023.108551.
[33] Chan R, Stewart K, Misirovs R, et al. Targeting downstream type 2 cytokines or upstream epithelial alarmins for severe asthma[J]. J Allergy Clin Immunol Pract, 2022, 10(6): 1497-1505.
[34] Shinkai M, Yabuta T. Tezepelumab: an anti-thymic stromal lymphopoietin monoclonal antibody for the treatment of asthma[J]. Immunotherapy, 2023, 15(17): 1435-1447.
[35] Wechsler ME, Ruddy MK, Pavord ID, et al. Efficacy and safety of itepekimab in patients with moderate-to-severe asthma[J]. N Engl J Med, 2021, 385(18): 1656-1668.
[36] Maspero J, Agache IO, Kamei T, et al. Long-term safety and exploratory efficacy of fevipiprant in patients with inadequately controlled asthma: the SPIRIT randomised clinical trial[J]. Respir Res, 2021, 22(1): 311. doi: 10.1186/s12931-021-01904-8.
[37] Eger K, Kroes JA, Ten Brinke A, et al. Long-term therapy response to anti-IL-5 biologics in severe asthma-a real-life evaluation[J]. J Allergy Clin Immunol Pract, 2021, 9(3): 1194-1200.
[38] Demarche SF, Schleich FN, Paulus VA, et al. Is it possible to claim or refute sputum eosinophils ≥ 3% in asthmatics with sufficient accuracy using biomarkers?[J]. Respir Res, 2017, 18(1): 133. doi: 10.1186/s12931-017-0615-9.
[39] Gautam S, Chu JH, Cohen AJ, et al. Sputum alarmins delineate distinct T2 cytokine pathways and unique subtypes of patients with asthma[J]. Allergy, 2023, 78(12): 3274-3277.
[40] Banno A, Reddy AT, Lakshmi SP, et al. Bidirectional interaction of airway epithelial remodeling and inflammation in asthma[J]. Clin Sci(Lond), 2020, 134(9): 1063-1079.
[41] Vannella KM, Ramalingam TR, Borthwick LA, et al. Combinatorial targeting of TSLP, IL-25, and IL-33 in type 2 cytokine-driven inflammation and fibrosis[J]. Sci Transl Med, 2016, 8(337): 337ra65. doi: 10.1126/scitranslmed.aaf1938.
[1] BU Meiling, WANG Jinrong, FENG Mei, SUN Lifeng. Mechanism of FOXM1 in acute exacerbation of asthma induced by respiratory virus infection in mice [J]. Journal of Shandong University (Health Sciences), 2023, 61(6): 1-9.
[2] LI Huayu, SHI Xiaohan, ZHANG Xinrui, LI Feng. Association between sleep disturbance and inflammatory cytokines in 203 patients with glioma [J]. Journal of Shandong University (Health Sciences), 2022, 60(12): 26-30.
[3] ZHANG Weiwei, HUA Fang, LIANG Chaoshuai, CHU Miaomiao, SUN Jiayi, FRANK Zaucke, XIN Wei. Thyroid stimulating hormone promotes chondrocyte differentiation via anti-inflammatory protein CTRP3 [J]. Journal of Shandong University (Health Sciences), 2022, 60(10): 1-8.
[4] GE Shaohua, DING Tian, LIU Hongrui. Role and regulatory mechanism of type 2 immunity in tissue repair [J]. Journal of Shandong University (Health Sciences), 2021, 59(9): 51-56.
[5] ZHANG Qian, QIN Mingming, HE Xuejia, CAI Qiujing, ZHANG Yamin, LI Qingsu, ZHU Weiwei. Effects of calcitriol on EMT induced by TGF-β1 in asthma [J]. Journal of Shandong University (Health Sciences), 2021, 59(7): 10-18.
[6] LI Xiangqing, YIN Xin, ZHAO Xuelian, ZHAO Peiqing. Expression and clinical significance of circulating CD56bright subset of NK cells in patients with Parkinsons disease [J]. Journal of Shandong University (Health Sciences), 2021, 59(2): 34-40.
[7] LIU Xiaofei, LIANG Ying, ZHANG Congxi, WANG Juan, PAN Yun, XU Jiawei, CHANG Chun, DONG Liang. Association of serum leptin with induced sputum eosinophils in 92 asthmatic patients [J]. Journal of Shandong University (Health Sciences), 2020, 1(9): 27-33.
[8] CAI Qiujing, ZHANG Qian, HE Xuejia, SUN Wenli, GUO Aili, ZHANG Nan, ZHU Weiwei. Airway smooth muscle cells regulate IL-33 expression through TGF-β1/Smad3 signaling pathway to participate in asthma [J]. Journal of Shandong University (Health Sciences), 2020, 58(4): 78-83.
[9] YANG Liping, MU Tingting, YANG Yujuan, ZHANG Yu, SONG Xicheng. Effects of inhaled allergens on pulmonary function in children with adenoid hypertrophy complicated with bronchial asthma [J]. Journal of Shandong University (Health Sciences), 2020, 58(3): 107-112.
[10] LI Yan, NIU Rui, WANG Chaochao. Analysis of the bronchodilator test of 122 patients with asthma [J]. Journal of Shandong University (Health Sciences), 2020, 58(11): 81-84.
[11] WANG Haixia, LI Yizhang, BAI Chenxiao, JIANG Di, WANG Liwen, CHEN Ou. A Meta-analysis of the efficacy and safety of clarithromycin in adjuvant treatment of asthma [J]. Journal of Shandong University (Health Sciences), 2019, 57(11): 27-33.
[12] CHEN Ou, LI Guoyong, LIU Aihong, ZHU Xiaobo, CHEN Shaojie, WANG Yibiao. Anti-inflammatory mechanism of ephedra treatment of asthma based on network pharmacology [J]. Journal of Shandong University (Health Sciences), 2019, 57(1): 55-61.
[13] GONG Xiaodan, ZHAO Fangzheng, CAO Ke, DENG Penghui, ZHANG Caiqing. Effect of myosin light chain kinase on airway inflammation and lung function in asthma model mice [J]. Journal of Shandong University (Health Sciences), 2017, 55(12): 18-23.
[14] LIU Xiaojuan, DING Lijie, KANG Fengling, ZHOU Miao, XUE Fuzhong. A prediction model for bronchial asthma risk based on a health management population [J]. Journal of Shandong University (Health Sciences), 2017, 55(12): 56-61.
[15] LIU Qingfa, WANG Chao, SUN Qijing, GONG Xiaodan, ZHANG Caiqing. Effect of interleukin-25 on inducing the airway remodeling of asthma mice by nuocyte cells [J]. JOURNAL OF SHANDONG UNIVERSITY (HEALTH SCIENCES), 2016, 54(8): 28-33.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!