Review

Scoping review on severe asthma: Cytokines, cells, triggers and multi-omics approaches

  • Received: 09 December 2023 Revised: 16 March 2024 Accepted: 22 March 2024 Published: 10 April 2024
  • Severe asthma (SA) is a refractory condition that does not respond well to conventional treatments. Patients with SA have heterogenetic endotypes. Endotypes for SA can be classified as type 2 cytokine-high and type 2 cytokine-low. The condition can also be classified as eosinophilic, neutrophilic, mixed, and paucigranulocytic SA. Abnormalities in TH1 and TH17 cytokines can be present in some SA patients. Innate lymphoid cells, airway smooth muscle cells, and lung epithelial cells have important roles in the disease. Viral infections, bacterial infections, fungal infections, smoking, allergens, and pollutants are major triggers that determine the severity of the disease. Biologics have been proven to be effective for some type 2 high patients. Multi-omics approaches, including genomics, transcriptomics, proteomics, metabolomics, and metagenomics, have identified many novel genes or molecules that could serve as biomarkers or potential therapeutic targets for SA. Investigations of the mechanisms of novel genes and molecules will help us understand the condition and find new treatment means for SA in the future.

    Citation: Hongchun Du, Min Zhang, Jinming Gao, Youming Zhang. Scoping review on severe asthma: Cytokines, cells, triggers and multi-omics approaches[J]. AIMS Allergy and Immunology, 2024, 8(1): 56-79. doi: 10.3934/Allergy.2024005

    Related Papers:

  • Severe asthma (SA) is a refractory condition that does not respond well to conventional treatments. Patients with SA have heterogenetic endotypes. Endotypes for SA can be classified as type 2 cytokine-high and type 2 cytokine-low. The condition can also be classified as eosinophilic, neutrophilic, mixed, and paucigranulocytic SA. Abnormalities in TH1 and TH17 cytokines can be present in some SA patients. Innate lymphoid cells, airway smooth muscle cells, and lung epithelial cells have important roles in the disease. Viral infections, bacterial infections, fungal infections, smoking, allergens, and pollutants are major triggers that determine the severity of the disease. Biologics have been proven to be effective for some type 2 high patients. Multi-omics approaches, including genomics, transcriptomics, proteomics, metabolomics, and metagenomics, have identified many novel genes or molecules that could serve as biomarkers or potential therapeutic targets for SA. Investigations of the mechanisms of novel genes and molecules will help us understand the condition and find new treatment means for SA in the future.



    加载中

    Acknowledgments



    Hongchun Du was awarded a scholarship from China Scholarship Council (201906385024). Jinming Gao was supported by a grant from the National Natural Sciences Foundation of China (No. 81970025).

    Conflict of interest



    All authors declare no conflicts of interest in this paper.

    [1] Merhej T, Zein JG (2023) Epidemiology of asthma: Prevalence and burden of disease. Adv Exp Med Biol 1426: 3-23. https://doi.org/10.1007/978-3-031-32259-4_1
    [2] Heffler E, Blasi F, Latorre M, et al. (2019) The severe asthma network in Italy: Findings and perspectives. J Allergy Clin Immunol Pract 7: 1462-1468. https://doi.org/10.1016/j.jaip.2018.10.016
    [3] Chung KF, Wenzel SE, Brozek JL, et al. (2014) International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J 43: 343-373. https://doi.org/10.1183/09031936.00202013
    [4] Delgado Romero J, Miralles Lopez JC, Alvarez Puebla M, et al. (2021) Severe asthma units accredited by the Spanish Society of Allergology and Clinical Immunology (SEAIC): Experience and future. J Investig Allergol Clin Immunol 31: 182-184. https://doi.org/10.18176/jiaci.0618
    [5] Agache I (2019) Severe asthma phenotypes and endotypes. Semin Immunol 46: 101301. https://doi.org/10.1016/j.smim.2019.101301
    [6] Lotvall J, Akdis CA, Bacharier LB, et al. (2011) Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol 127: 355-360. https://doi.org/10.1016/j.jaci.2010.11.037
    [7] Fahy JV (2015) Type 2 inflammation in asthma--present in most, absent in many. Nat Rev Immunol 15: 57-65. https://doi.org/10.1038/nri3786
    [8] Zhang YM, Fear DJ, Willis-Owen SAG, et al. (2016) Global gene regulation during activation of immunoglobulin class switching in human B cells. Sci Rep-Uk 6. https://doi.org/10.1038/srep37988
    [9] Pelaia C, Paoletti G, Puggioni F, et al. (2019) Interleukin-5 in the pathophysiology of severe asthma. Front Physiol 10: 1514. https://doi.org/10.3389/fphys.2019.01514
    [10] Marone G, Granata F, Pucino V, et al. (2019) The intriguing role of interleukin 13 in the pathophysiology of asthma. Front Pharmacol 10: 1387. https://doi.org/10.3389/fphar.2019.01387
    [11] Woodruff PG, Modrek B, Choy DF, et al. (2009) T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med 180: 388-395. https://doi.org/10.1164/rccm.200903-0392OC
    [12] Chan BCL, Lam CWK, Tam LS, et al. (2019) IL33: Roles in allergic inflammation and therapeutic perspectives. Front Immunol 10: 364. https://doi.org/10.3389/fimmu.2019.00364
    [13] Saglani S, Lui S, Ullmann N, et al. (2013) IL-33 promotes airway remodeling in pediatric patients with severe steroid-resistant asthma. J Allergy Clin Immunol 132: 676-685.E13. https://doi.org/10.1016/j.jaci.2013.04.012
    [14] Valizadeh A, Khosravi A, Zadeh LJ, et al. (2015) Role of IL-25 in immunity. J Clin Diagn Res 9: OE01-04. https://doi.org/10.7860/JCDR/2015/12235.5814
    [15] Cheng D, Xue Z, Yi L, et al. (2014) Epithelial interleukin-25 is a key mediator in Th2-high, corticosteroid-responsive asthma. Am J Respir Crit Care Med 190: 639-648. https://doi.org/10.1164/rccm.201403-0505OC
    [16] Paplinska-Goryca M, Grabczak EM, Dabrowska M, et al. (2018) Sputum interleukin-25 correlates with asthma severity: a preliminary study. Postepy Dermatol Alergol 35: 462-469. https://doi.org/10.5114/ada.2017.71428
    [17] Ying S, O'Connor B, Ratoff J, et al. (2005) Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J immunol 174: 8183-8190. https://doi.org/10.4049/jimmunol.174.12.8183
    [18] Ying S, O'Connor B, Ratoff J, et al. (2008) Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J immunol 181: 2790-2798. https://doi.org/10.4049/jimmunol.181.4.2790
    [19] Ferreira MA, Matheson MC, Tang CS, et al. (2014) Genome-wide association analysis identifies 11 risk variants associated with the asthma with hay fever phenotype. J Allergy Clin Immunol 133: 1564-1571. https://doi.org/10.1016/j.jaci.2013.10.030
    [20] Lai JF, Thompson LJ, Ziegler SF (2020) TSLP drives acute T(H)2-cell differentiation in lungs. J Allergy Clin Immunol 146: 1406-1418.E7. https://doi.org/10.1016/j.jaci.2020.03.032
    [21] He JQ, Hallstrand TS, Knight D, et al. (2009) A thymic stromal lymphopoietin gene variant is associated with asthma and airway hyperresponsiveness. J Allergy Clin Immunol 124: 222-229. https://doi.org/10.1016/j.jaci.2009.04.018
    [22] Fornasa G, Tsilingiri K, Caprioli F, et al. (2015) Dichotomy of short and long thymic stromal lymphopoietin isoforms in inflammatory disorders of the bowel and skin. J Allergy Clin Immunol 136: 413-422. https://doi.org/10.1016/j.jaci.2015.04.011
    [23] Mitchell PD, O'Byrne PM (2017) Biologics and the lung: TSLP and other epithelial cell-derived cytokines in asthma. Pharmacol Ther 169: 104-112. https://doi.org/10.1016/j.pharmthera.2016.06.009
    [24] Verstraete K, Peelman F, Braun H, et al. (2017) Structure and antagonism of the receptor complex mediated by human TSLP in allergy and asthma. NatCommun 8: 14937. https://doi.org/10.1038/ncomms14937
    [25] Matera MG, Rogliani P, Calzetta L, et al. (2020) TSLP inhibitors for asthma: Current status and future prospects. Drugs 80: 449-458. https://doi.org/10.1007/s40265-020-01273-4
    [26] Hinks TSC, Levine SJ, Brusselle GG (2021) Treatment options in type-2 low asthma. Eur Respir J 57. https://doi.org/10.1183/13993003.00528-2020
    [27] Nelson BH (2004) IL-2, regulatory T cells, and tolerance. J Immunol 172: 3983-3988. https://doi.org /10.4049/jimmunol.172.7.3983/
    [28] Ishihara C, Ochiai K, Kagami M, et al. (1997) Human peripheral eosinophils express functional interferon-gamma receptors (IFN-gammaR). Clin Exp Immunol 110: 524-529. https://doi.org/10.1046/j.1365-2249.1997.4511469.x
    [29] Yamaguchi Y, Suda T, Shiozaki H, et al. (1990) Role of IL-5 in IL-2-induced eosinophilia. In vivo and in vitro expression of IL-5 mRNA by IL-2. J Immunol 145: 873-877. https://doi.org/10.4049/jimmunol.145.3.873
    [30] Camiolo MJ, Kale SL, Oriss TB, et al. (2021) Immune responses and exacerbations in severe asthma. Curr Opin Immunol 72: 34-42. https://doi.org/10.1016/j.coi.2021.03.004
    [31] Gauthier M, Chakraborty K, Oriss TB, et al. (2017) Severe asthma in humans and mouse model suggests a CXCL10 signature underlies corticosteroid-resistant Th1 bias. JCI Insight 2. https://doi.org/10.1172/jci.insight.94580
    [32] Muehling LM, Heymann PW, Wright PW, et al. (2020) Human T(H)1 and T(H)2 cells targeting rhinovirus and allergen coordinately promote allergic asthma. J Allergy Clin Immunol 146: 555-570. https://doi.org/10.1016/j.jaci.2020.03.037
    [33] Meyts I, Hellings PW, Hens G, et al. (2006) IL-12 contributes to allergen-induced airway inflammation in experimental asthma. J Immunol 177: 6460-6470. https://doi.org/10.4049/jimmunol.177.9.6460
    [34] Korn T, Bettelli E, Oukka M, et al. (2009) IL-17 and Th17 Cells. Annu Rev Immunol 27: 485-517. https://doi.org/10.1146/annurev.immunol.021908.132710
    [35] Newcomb DC, Peebles RS (2013) Th17-mediated inflammation in asthma. Curr Opin Immunol 25: 755-760. https://doi.org/10.1016/j.coi.2013.08.002
    [36] Ito T, Hirose K, Nakajima H (2019) Bidirectional roles of IL-22 in the pathogenesis of allergic airway inflammation. Allergol Int 68: 4-8. https://doi.org/10.1016/j.alit.2018.10.002
    [37] Nakajima H, Hirose K (2010) Role of IL-23 and Th17 cells in airway inflammation in asthma. Immune Netw 10: 1-4. https://doi.org/10.4110/in.2010.10.1.1
    [38] Svenningsen S, Nair P (2017) Asthma endotypes and an overview of targeted therapy for asthma. Front Med (Lausanne) 4: 158. https://doi.org/10.3389/fmed.2017.00158
    [39] Nakagome K, Nagata M (2018) Involvement and possible role of eosinophils in asthma exacerbation. Front Immunol 9: 2220. https://doi.org/10.3389/fimmu.2018.02220
    [40] Ray A, Kolls JK (2017) Neutrophilic inflammation in asthma and association with disease severity. Trends Immunol 38: 942-954. https://doi.org/10.1016/j.it.2017.07.003
    [41] Tecchio C, Micheletti A, Cassatella MA (2014) Neutrophil-derived cytokines: facts beyond expression. Front Immunol 5: 508. https://doi.org/10.3389/fimmu.2014.00508
    [42] Jatakanon A, Uasuf C, Maziak W, et al. (1999) Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med 160: 1532-1539. https://doi.org/10.1164/ajrccm.160.5.9806170
    [43] Norzila MZ, Fakes K, Henry RL, et al. (2000) Interleukin-8 secretion and neutrophil recruitment accompanies induced sputum eosinophil activation in children with acute asthma. Am J Respir Crit Care Med 161: 769-774. https://doi.org/10.1164/ajrccm.161.3.9809071
    [44] Klose CS, Kiss EA, Schwierzeck V, et al. (2013) A T-bet gradient controls the fate and function of CCR6-RORgammat+ innate lymphoid cells. Nature 494: 261-265. https://doi.org/10.1038/nature11813
    [45] Malik B, Bartlett NW, Upham JW, et al. (2023) Severe asthma ILC2s demonstrate enhanced proliferation that is modified by biologics. Respirology 28: 758-766. https://doi.org/10.1111/resp.14506
    [46] Kim CH, Hashimoto-Hill S, Kim M (2016) Migration and tissue tropism of innate lymphoid cells. Trends Immunol 37: 68-79. https://doi.org/10.1016/j.it.2015.11.003
    [47] Kim HY, Lee HJ, Chang YJ, et al. (2014) Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med 20: 54-61. https://doi.org/10.1038/nm.3423
    [48] Jonckheere AC, Seys SF, Steelant B, et al. (2022) Innate lymphoid cells are required to induce airway hyperreactivity in a murine neutrophilic asthma model. Front Immunol 13: 849155. https://doi.org/10.3389/fimmu.2022.849155
    [49] He LX, Yang L, Liu T, et al. (2023) Group 3 innate lymphoid cells secret neutrophil chemoattractants and are insensitive to glucocorticoid via aberrant GR phosphorylation. Respir Res 24: 90. https://doi.org/10.1186/s12931-023-02395-5
    [50] Dilasser F, Rose L, Hassoun D, et al. (2021) Essential role of smooth muscle Rac1 in severe asthma-associated airway remodelling. Thorax 7: 326-334. https://doi.org/10.1136/thoraxjnl-2020-216271
    [51] Janssen LJ, Killian K (2006) Airway smooth muscle as a target of asthma therapy: history and new directions. Respir Res 7: 123. https://doi.org/10.1186/1465-9921-7-123
    [52] Rutting S, Xenaki D, Reddy KD, et al. (2021) Airway smooth muscle cells from severe asthma patients with fixed airflow obstruction are responsive to steroid and bronchodilator treatment in vitro. ERJ Open Res 7. https://doi.org/10.1183/23120541.00117-2021
    [53] Wills-Karp M (2010) Allergen-specific pattern recognition receptor pathways. Curr Opin Immunol 22: 777-782. https://doi.org/10.1016/j.coi.2010.10.01
    [54] Heijink IH, Kuchibhotla VNS, Roffel MP, et al. (2020) Epithelial cell dysfunction, a major driver of asthma development. Allergy 75: 1902-1917. https://doi.org/10.1111/all.14421
    [55] Zhang Y, Moffatt MF, Cookson WO (2012) Genetic and genomic approaches to asthma: new insights for the origins. Curr Opin Pulm Med 18: 6-13. https://doi.org/10.1097/MCP.0b013e32834dc532
    [56] Zhang Y, Poobalasingam T, Yates LL, et al. (2018) Manipulation of dipeptidylpeptidase 10 in mouse and human in vivo and in vitro models indicates a protective role in asthma. Dis Model Mech 11. https://doi.org/10.1242/dmm.031369
    [57] Zhang YM (2021) Orosomucoid-like protein 3, rhinovirus and asthma. World J Crit Care Med 10: 170-182. https://doi.org/10.5492/wjccm.v10.i5.170
    [58] Johnston SL (2007) Innate immunity in the pathogenesis of virus-induced asthma exacerbations. Proc Am Thorac Soc 4: 267-270. https://doi.org/10.1513/pats.200701-030AW
    [59] Bizzintino J, Lee WM, Laing IA, et al. (2011) Association between human rhinovirus C and severity of acute asthma in children. Eur Respir J 37: 1037-1042. https://doi.org/10.1183/09031936.00092410
    [60] Dougherty RH, Fahy JV (2009) Acute exacerbations of asthma: epidemiology, biology and the exacerbation-prone phenotype. Clin Exp Allergy 39: 193-202. https://doi.org/10.1111/j.1365-2222.2008.03157.x
    [61] Zhang Y, Willis-Owen SAG, Spiegel S, et al. (2019) The ORMDL3 asthma gene regulates ICAM1 and has multiple effects on cellular inflammation. Am J Respir Crit Care Med 199: 478-488. https://doi.org/10.1164/rccm.201803-0438OC
    [62] Cantero-Recasens G, Fandos C, Rubio-Moscardo F, et al. (2010) The asthma-associated ORMDL3 gene product regulates endoplasmic reticulum-mediated calcium signaling and cellular stress. Hum Mol Genet 19: 111-121. https://doi.org/10.1093/hmg/ddp471
    [63] Ha SG, Ge XN, Bahaie NS, et al. (2013) ORMDL3 promotes eosinophil trafficking and activation via regulation of integrins and CD48. Natcommun 4: 2479. https://doi.org/10.1038/ncomms3479
    [64] Corne JM, Marshall C, Smith S, et al. (2002) Frequency, severity, and duration of rhinovirus infections in asthmatic and non-asthmatic individuals: a longitudinal cohort study. Lancet 359: 831-834. https://doi.org/10.1016/S0140-6736(02)07953-9
    [65] Wennergren G, Kristjansson S (2001) Relationship between respiratory syncytial virus bronchiolitis and future obstructive airway diseases. Eur Respir J 18: 1044-1058. https://doi.org/10.1183/09031936.01.00254101
    [66] Zhou Y, Bacharier LB, Isaacson-Schmid M, et al. (2016) Azithromycin therapy during respiratory syncytial virus bronchiolitis: Upper airway microbiome alterations and subsequent recurrent wheeze. J Allergy Clin Immunol 138: 1215-1219.E5. https://doi.org/10.1016/j.jaci.2016.03.054
    [67] Polosa R, Russo C, Caponnetto P, et al. (2011) Greater severity of new onset asthma in allergic subjects who smoke: a 10-year longitudinal study. Respir Res 12: 16. https://doi.org/10.1186/1465-9921-12-16
    [68] Arbeitskreis Blut U (2009) Influenza Virus. Transfus Med Hemother 36: 32-39. https://doi.org/10.1159/000197314
    [69] Veerapandian R, Snyder JD, Samarasinghe AE (2018) Influenza in asthmatics: For better or for worse?. Front Immunol 9: 1843. https://doi.org/10.3389/fimmu.2018.01843
    [70] Machhi J, Herskovitz J, Senan AM, et al. (2020) The natural history, pathobiology, and clinical manifestations of SARS-CoV-2 infections. J Neuroimmune Pharmacol 15: 359-386. https://doi.org/10.1007/s11481-020-09944-5
    [71] Kwok WC, Tam TCC, Lam DCL, et al. (2023) Worsening of asthma control after recovery from mild to moderate COVID-19 in patients from Hong Kong. Respir Res 24: 53. https://doi.org/10.1186/s12931-023-02363-z
    [72] Agondi RC, Menechino N, Marinho A, et al. (2022) Worsening of asthma control after COVID-19. Front Med (Lausanne) 9: 882665. https://doi.org/10.3389/fmed.2022.882665
    [73] Kraft M (2000) The role of bacterial infections in asthma. Clin Chest Med 21: 301-313. https://doi.org/10.1016/s0272-5231(05)70268-9
    [74] Just J, Fayon M, Charavel A, et al. (1997) Role of bacterial infections in children with asthma. Pediatr Pulmonol Suppl 16: 76. https://doi.org/10.1002/ppul.1950230844
    [75] Weiser JN (2010) The pneumococcus: why a commensal misbehaves. J Mol Med (Berl) 88: 97-102. https://doi.org/10.1007/s00109-009-0557-x
    [76] Leiberman A, Dagan R, Leibovitz E, et al. (1999) The bacteriology of the nasopharynx in childhood. Int J Pediatr Otorhinolaryngol 49: S151-153. https://doi.org/10.1016/s0165-5876(99)00151-2
    [77] Faden H, Duffy L, Wasielewski R, et al. (1997) Relationship between nasopharyngeal colonization and the development of otitis media in children. Tonawanda/Williamsville Pediatrics. J Infect Dis 175: 1440-1445. https://doi.org/10.1086/516477
    [78] Tan TQ (2020) Asthma and invasive pneumococcal disease in the age of pneumococcal conjugate vaccines. Pediatrics 145. https://doi.org/10.1542/peds.2019-3360
    [79] Talbot TR, Hartert TV, Mitchel E, et al. (2005) Asthma as a risk factor for invasive pneumococcal disease. N Engl J Med 352: 2082-2090. https://doi.org/10.1056/NEJMoa044113
    [80] Kuschner WG, Kuschner RA (2005) Asthma and invasive pneumococcal disease. N Engl J Med 353: 738-739. https://doi.org/10.1056/NEJM200508183530720
    [81] Castro-Rodriguez JA, Abarca K, Forno E (2020) Asthma and the risk of invasive pneumococcal disease: A meta-analysis. Pediatrics 145. https://doi.org/10.1542/peds.2019-1200
    [82] Morey P, Cano V, Marti-Lliteras P, et al. (2011) Evidence for a non-replicative intracellular stage of nontypable Haemophilus influenzae in epithelial cells. Microbiology (Reading) 157: 234-250. https://doi.org/10.1099/mic.0.040451-0
    [83] Ackland J, Barber C, Heinson A, et al. (2022) Nontypeable Haemophilus influenzae infection of pulmonary macrophages drives neutrophilic inflammation in severe asthma. Allergy 77: 2961-2973. https://doi.org/10.1111/all.15375
    [84] Versi A, Ivan FX, Abdel-Aziz MI, et al. (2023) Haemophilus influenzae and Moraxella catarrhalis in sputum of severe asthma with inflammasome and neutrophil activation. Allergy 78: 2906-2920. https://doi.org/10.1111/all.15776
    [85] Garcia-Clemente M, de la Rosa D, Maiz L, et al. (2020) Impact of pseudomonas aeruginosa infection on patients with chronic inflammatory airway diseases. J Clin Med 9. https://doi.org/10.3390/jcm9123800
    [86] Denning DW, O'Driscoll BR, Hogaboam CM, et al. (2006) The link between fungi and severe asthma: a summary of the evidence. Eur Respir J 27: 615-626. https://doi.org/10.1183/09031936.06.00074705
    [87] Denning DW, O'Driscoll BR, Powell G, et al. (2009) Randomized controlled trial of oral antifungal treatment for severe asthma with fungal sensitization: The Fungal Asthma Sensitization Trial (FAST) study. Am J Respir Crit Care Med 179: 11-18. https://doi.org/10.1164/rccm.200805-737OC
    [88] Overton NL, Simpson A, Bowyer P, et al. (2017) Genetic susceptibility to severe asthma with fungal sensitization. Int J Immunogenet 44: 93-106. https://doi.org/10.1111/iji.12312
    [89] Kennedy JL, Heymann PW, Platts-Mills TA (2012) The role of allergy in severe asthma. Clin Exp Allergy 42: 659-669. https://doi.org/10.1111/j.1365-2222.2011.03944.x
    [90] Mthembu N, Ikwegbue P, Brombacher F, et al. (2021) Respiratory viral and bacterial factors that influence early childhood asthma. Front Allergy 2: 692841. https://doi.org/10.3389/falgy.2021.692841
    [91] Boulet LP, Lemiere C, Archambault F, et al. (2006) Smoking and asthma: clinical and radiologic features, lung function, and airway inflammation. Chest 129: 661-668. https://doi.org/10.1378/chest.129.3.661
    [92] Lazarus SC, Chinchilli VM, Rollings NJ, et al. (2007) Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma. Am J Respir Crit Care Med 175: 783-790. https://doi.org/10.1164/rccm.200511-1746OC
    [93] Diaz-Sanchez D, Rumold R, Gong H (2006) Challenge with environmental tobacco smoke exacerbates allergic airway disease in human beings. J Allergy Clin Immunol 118: 441-446. https://doi.org/10.1016/j.jaci.2006.04.047
    [94] Thomson NC, Chaudhuri R, Livingston E (2004) Asthma and cigarette smoking. Eur Respir J 24: 822-833. https://doi.org/10.1183/09031936.04.00039004
    [95] Li X, Zhang Y, Zhang R, et al. (2022) Association between e-cigarettes and asthma in adolescents: A systematic review and meta-analysis. Am J Prev Med 62: 953-960. https://doi.org/10.1016/j.amepre.2022.01.015
    [96] Rage E, Siroux V, Kunzli N, et al. (2009) Epidemiological study on the G, environment of A: Air pollution and asthma severity in adults. Occup Environ Med 66: 182-188. https://doi.org/10.1136/oem.2007.038349
    [97] Neuhaus-Steinmetz U, Uffhausen F, Herz U, et al. (2000) Priming of allergic immune responses by repeated ozone exposure in mice. Am J Respir Cell Mol Biol 23: 228-233. https://doi.org/10.1165/ajrcmb.23.2.3898
    [98] Peden DB, Boehlecke B, Horstman D, et al. (1997) Prolonged acute exposure to 0.16 ppm ozone induces eosinophilic airway inflammation in asthmatic subjects with allergies. J Allergy Clin Immunol 100: 802-808. https://doi.org/10.1016/s0091-6749(97)70277-x
    [99] Nurmatov UB, Tagiyeva N, Semple S, et al. (2015) Volatile organic compounds and risk of asthma and allergy: a systematic review. Eur Respir Rev 24: 92-101. https://doi.org/10.1183/09059180.00000714
    [100] Pelaia C, Crimi C, Vatrella A, et al. (2020) Molecular targets for biological therapies of severe asthma. Front Immunol 11: 603312. https://doi.org/10.3389/fimmu.2020.603312
    [101] Patel VH, Thannir S, Dhanani M, et al. (2023) Current limitations and recent advances in the management of asthma. Dis Mon 69: 101483. https://doi.org/10.1016/j.disamonth.2022.101483
    [102] Ivanisevic T, Sewduth RN (2023) Multi-Omics integration for the design of novel therapies and the identification of novel biomarkers. Proteomes 11. https://doi.org/10.3390/proteomes11040034
    [103] Chang D, Hunkapiller J, Bhangale T, et al. (2022) A whole genome sequencing study of moderate to severe asthma identifies a lung function locus associated with asthma risk. Sci Rep 12: 5574. https://doi.org/10.1038/s41598-022-09447-8
    [104] Shrine N, Portelli MA, John C, et al. (2019) Moderate-to-severe asthma in individuals of European ancestry: a genome-wide association study. Lancet Respir Med 7: 20-34. https://doi.org/10.1016/S2213-2600(18)30389-8
    [105] Yan Q, Forno E, Herrera-Luis E, et al. (2021) A genome-wide association study of severe asthma exacerbations in Latino children and adolescents. Eur Respir J 57. https://doi.org/10.1183/13993003.02693-2020
    [106] Wan YI, Shrine NR, Soler Artigas M, et al. (2012) Genome-wide association study to identify genetic determinants of severe asthma. Thorax 67: 762-768. https://doi.org/10.1136/thoraxjnl-2011-201262
    [107] Tsutsui K, Manabe R, Yamada T, et al. (2010) ADAMTSL-6 is a novel extracellular matrix protein that binds to fibrillin-1 and promotes fibrillin-1 fibril formation. J Biol Chem 285: 4870-4882. https://doi.org/10.1074/jbc.M109.076919
    [108] Bonser LR, Erle DJ (2017) Airway mucus and asthma: The role of MUC5AC and MUC5B. J Clin Med 6. https://doi.org/10.3390/jcm6120112
    [109] Moffatt MF, Kabesch M, Liang L, et al. (2007) Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 448: 470-473. https://doi.org/10.1038/nature06014
    [110] Moffatt MF, Gut IG, Demenais F, et al. (2010) A large-scale, consortium-based genomewide association study of asthma. N Engl J Med 363: 1211-1221. https://doi.org/10.1056/NEJMoa0906312
    [111] Chen Q, Shi P, Wang Y, et al. (2019) GSDMB promotes non-canonical pyroptosis by enhancing caspase-4 activity. J Mol Cell Biol 11: 496-508. https://doi.org/10.1093/jmcb/mjy056
    [112] Persson H, Kwon AT, Ramilowski JA, et al. (2015) Transcriptome analysis of controlled and therapy-resistant childhood asthma reveals distinct gene expression profiles. J Allergy Clin Immunol 136: 638-648. https://doi.org/10.1016/j.jaci.2015.02.026
    [113] Tsitsiou E, Williams AE, Moschos SA, et al. (2012) Transcriptome analysis shows activation of circulating CD8+ T cells in patients with severe asthma. J Allergy Clin Immunol 129: 95-103. https://doi.org/10.1016/j.jaci.2011.08.011
    [114] Zeng X, Qing J, Li CM, et al. (2023) Blood transcriptomic signature in type-2 biomarker-low severe asthma and asthma control. J Allergy Clin Immunol 152: 876-886. https://doi.org/10.1016/j.jaci.2023.05.023
    [115] Baines KJ, Simpson JL, Wood LG, et al. (2011) Transcriptional phenotypes of asthma defined by gene expression profiling of induced sputum samples. J Allergy Clin Immunol 127: 153-160.E9. https://doi.org/10.1016/j.jaci.2010.10.024
    [116] Berthon BS, Gibson PG, Wood LG, et al. (2017) A sputum gene expression signature predicts oral corticosteroid response in asthma. Eur Respir J 49. https://doi.org/10.1183/13993003.00180-2017
    [117] Hodge S (2020) Sputum transcriptomics: A tool for identifying gene expression underlying mechanistic pathways in severe asthma. Respirology 25: 668-669. https://doi.org/10.1111/resp.13769
    [118] Woodruff PG, Boushey HA, Dolganov GM, et al. (2007) Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci USA 104: 15858-15863. https://doi.org/10.1073/pnas.0707413104
    [119] Li B, Sun WX, Zhang WY, et al. (2021) The transcriptome characteristics of severe asthma from the prospect of co-expressed gene modules. Front Genet 12: 765400. https://doi.org/10.3389/fgene.2021.765400
    [120] Zhang L, Chun Y, Irizar H, et al. (2023) Integrated study of systemic and local airway transcriptomes in asthma reveals causal mediation of systemic effects by airway key drivers. Genome Med 15: 71. https://doi.org/10.1186/s13073-023-01222-2
    [121] Suzuki M, Cole JJ, Konno S, et al. (2021) Large-scale plasma proteomics can reveal distinct endotypes in chronic obstructive pulmonary disease and severe asthma. Clin Transl Allergy 11. https://doi.org/10.1002/clt2.12091
    [122] Takahashi K, Pavlidis S, Ng Kee Kwong F, et al. (2018) Sputum proteomics and airway cell transcripts of current and ex-smokers with severe asthma in U-BIOPRED: an exploratory analysis. Eur Respir J 51. https://doi.org/10.1183/13993003.02173-2017
    [123] Cao C, Li W, Hua W, et al. (2017) Proteomic analysis of sputum reveals novel biomarkers for various presentations of asthma. J Transl Med 15: 171. https://doi.org/10.1186/s12967-017-1264-y
    [124] Dunn WB, Broadhurst DI, Atherton HJ, et al. (2011) Systems level studies of mammalian metabolomes: the roles of mass spectrometry and nuclear magnetic resonance spectroscopy. Chem Soc Rev 40: 387-426. https://doi.org/10.1039/b906712b
    [125] Reinke SN, Gallart-Ayala H, Gomez C, et al. (2017) Metabolomics analysis identifies different metabotypes of asthma severity. Eur Respir J 49. https://doi.org/10.1183/13993003.01740-2016
    [126] Gupta R, Radicioni G, Abdelwahab S, et al. (2019) Intercellular communication between airway epithelial cells is mediated by exosome-like vesicles. Am J Respir Cell Mol Biol 60: 209-220. https://doi.org/10.1165/rcmb.2018-0156OC
    [127] Pite H, Aguiar L, Morello J, et al. (2020) Metabolic dysfunction and asthma: Current perspectives. J Asthma Allergy 13: 237-247. https://doi.org/10.2147/JAA.S208823
    [128] Xu W, Comhair SAA, Janocha AJ, et al. (2017) Arginine metabolic endotypes related to asthma severity. PLoS One 12. https://doi.org/10.1371/journal.pone.0183066
    [129] Peters MC, McGrath KW, Hawkins GA, et al. (2016) Plasma interleukin-6 concentrations, metabolic dysfunction, and asthma severity: a cross-sectional analysis of two cohorts. Lancet Respir Med 4: 574-584. https://doi.org/10.1016/S2213-2600(16)30048-0
    [130] Weinstock GM (2012) Genomic approaches to studying the human microbiota. Nature 489: 250-256. https://doi.org/10.1038/nature11553
    [131] Depner M, Ege MJ, Cox MJ, et al. (2017) Bacterial microbiota of the upper respiratory tract and childhood asthma. J Allergy Clin Immunol 139: 826-834.E13. https://doi.org/10.1016/j.jaci.2016.05.050
    [132] Good JT, Kolakowski CA, Groshong SD, et al. (2012) Refractory asthma: importance of bronchoscopy to identify phenotypes and direct therapy. Chest 141: 599-606. https://doi.org/10.1378/chest.11-0741
    [133] Perez-Garcia J, Hernandez-Perez JM, Gonzalez-Perez R, et al. (2020) The genomics and metagenomics of asthma severity (GEMAS) study: Rationale and design. J Pers Med 10. https://doi.org/10.3390/jpm10030123
    [134] Huang YJ, Nariya S, Harris JM, et al. (2015) The airway microbiome in patients with severe asthma: Associations with disease features and severity. J Allergy Clin Immunol 136: 874-884. https://doi.org/10.1016/j.jaci.2015.05.044
  • Reader Comments
  • © 2024 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(596) PDF downloads(63) Cited by(0)

Article outline

Figures and Tables

Tables(4)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog