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:

    [1] Haiyan Guo, Mingsheng Lei, Jinan Ma, Hongchun Du, Youming Zhang . Allergy and ferroptosis. AIMS Allergy and Immunology, 2025, 9(1): 8-26. doi: 10.3934/Allergy.2025002
    [2] Hiroto Matsuse, Tohru Yamagishi, Norio Kodaka, Chihiro Nakano, Chizu Fukushima, Yasushi Obase, Hiroshi Mukae . Therapeutic modality of plasmacytoid dendritic cells in a murine model of Aspergillus fumigatus sensitized and infected asthma. AIMS Allergy and Immunology, 2017, 1(4): 232-241. doi: 10.3934/Allergy.2017.4.232
    [3] Sabrine Louhaichi, Mariem Salhi, Anissa Berraïes, Besma Hamdi, Jamel Ammar, Kamel Hamzaoui, Agnès Hamzaoui . Co-inhibitory receptors in female asthmatic patients: Correlation with IL-17 and IL-26. AIMS Allergy and Immunology, 2018, 2(1): 10-23. doi: 10.3934/Allergy.2018.1.10
    [4] Norio Kodaka, Chihiro Nakano, Takeshi Oshio, Hiroto Matsuse . The treatment of severe uncontrolled asthma using biologics. AIMS Allergy and Immunology, 2020, 4(1): 1-13. doi: 10.3934/Allergy.2020001
    [5] Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda . IL-17 signaling is regulated through intrinsic stability control of mRNA during inflammation. AIMS Allergy and Immunology, 2022, 6(3): 188-199. doi: 10.3934/Allergy.2022014
    [6] Omer Qibi, Severine Audusseau, Andrea Mogas, Zoulfia Allakhverdi, Abdelilah Soussi Gounni, Saba Al Heialy, Qutayba Hamid . No evidence for IgE receptor FcεRI expression on bronchial epithelial cells of asthmatic patients. AIMS Allergy and Immunology, 2018, 2(4): 165-179. doi: 10.3934/Allergy.2018.4.165
    [7] Izabela Krzemień, Angelika Szatan, Paulina Skalska, Martyna Cieślik, Angelika Domagała, Magdalena Gębicka, Krzysztof Bryniarski, Katarzyna Nazimek . Serum cytokine concentrations correlate with the severity of imiquimod and IL-23-induced psoriatic inflammation in mice. AIMS Allergy and Immunology, 2024, 8(4): 265-282. doi: 10.3934/Allergy.2024016
    [8] Federica Serra, Lisa Aielli, Erica Costantini . The role of miRNAs in the inflammatory phase of skin wound healing. AIMS Allergy and Immunology, 2021, 5(4): 264-278. doi: 10.3934/Allergy.2021020
    [9] Ken S. Rosenthal . Immune monitoring of the body’s borders. AIMS Allergy and Immunology, 2018, 2(3): 148-164. doi: 10.3934/Allergy.2018.3.148
    [10] Boel De Paepe . A recipe for myositis: nuclear factor κB and nuclear factor of activated T-cells transcription factor pathways spiced up by cytokines. AIMS Allergy and Immunology, 2017, 1(1): 31-42. doi: 10.3934/Allergy.2017.1.31
  • 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.



    Asthma is the most common chronic respiratory disease and is a highly heterogeneous condition characterised by reversible bronchial obstruction [1]. In most cases, asthma can be managed by using standard inhaled corticosteroids alone or in combination with other treatments; however, 5–10% of patients are inadequately controlled [2]. Clinically, it is not easy to define severe asthma (SA); the term includes all cases of therapy-resistant disease in all age groups. The European Respiratory Society and American Thoracic Society define SA as asthma requiring treatment with high-dose inhaled corticosteroids (ICS) plus a controller with or without systemic corticosteroids to maintain control of disease, or, despite this therapy, having sub-optimally controlled asthma [3]. SA is currently estimated to affect 3% to 10% of asthmatic adults and up to 2.5% of asthmatic children [4]. The difficulty in managing SA is due to the heterogenic phenotypes and endotypes from individual patients. Endotypes are referred to as mechanisms of disease [5] and are now widely accepted in the guidance of appropriate treatments in clinic. There are many endotypes proposed for asthma [6]. For SA, endotypes can be divided as type 2 cytokine high or type 2 cytokine low due to expression levels of cytokine clusters. Type 2 inflammation is induced by cytokines secreted by T helper 2 cells and other inflammatory cells. High type 2 cytokine expression is present in many, but not all, SA patients. Other cytokine pathways such as TH1 and TH17 also have important roles in SA. Endotypes of SA can also be defined as eosinophilic, neutrophilic, mixed, and paucigranulocytic due to inflammatory cells in the lungs. Innate lymphoid cells, epithelial cells, and airway smooth muscle also have roles for SA in some patients. SA can be triggered by viral infection, bacterial infection, fungi infection, allergens, smoking, and pollutants. Biologics to treat type 2 high SA have been shown to control the symptoms well. They have a great impact on the outcomes such as exacerbations, oral corticosteroids (OCS) use, and lung function improvement. For type 2 low SA, there is still a lack of effective drugs. The unmet need for managing SA promotes extensive investigations for this condition. There are many review studies available on asthma, from mechanisms to clinical managements. In this review, we searched for progress on investigations of SA, over the past ten years, and we briefly discuss the roles of major cytokines, immune-related cells, and triggers for SA. We focus on recent progress from multi-omics approaches for SA including genomics, transcriptomics, metabolics, and metagenomics and update the novel findings that could serve as biomarkers or potential therapeutic targets for SA in the future.

    Human T lymphocytes are derived from the thymus and consist of T helper cells, T cytotoxic cells, T regulatory (Treg) cells, and T natural killer (NK) cells. T helper cells can be divided as TH1 helper cells and TH2 helper cells. TH1 cells mainly release interleukin-2 (IL-2), interferon-γ (IFNγ), and lymphotoxin-α that can activate macrophage and cytotoxic T-cells. TH2 helper cells can release cytokines IL-4, IL-5, and IL-13 [7], which activate B lymphocytes. B lymphocytes produce immunoglobulins and can undergo class switching to generate IgE due to allergen or antigen stimulation [8].

    TH2 cytokines are termed as type 2 cytokines and are released from TH2 cells but can also be released from eosinophils, basophils, mast cells, group2 innate lymphoid cells (ILC2s), and B lymphocytes [7]. IL-4 regulates antibody production, haematopoiesis, and inflammation. IL-5 has important effects on eosinophils, promoting the cells' maturation, activation, and survival. IL-5 helps eosinophils' migrate from blood and recruit to the airways [9]. IL-13 exerts eosinophilic inflammation, mucus secretion, and airway hyperresponsiveness [10]. Clinically, TH2 response increases fractional exhaled nitric oxide (FeNO), eosinophilia in blood, and type-2 inflammatory epithelial gene signatures [11]. Other cytokines and regulators in TH2 response are IL-25, IL-33, and thymic stromal lymphopoietin (TSLP). They can be activated in airway epithelial cells by exposure to oxidants, viral and bacterial infections, and other pollutants, initialling TH2 response.

    IL-33: IL-33 is a member of the IL-1 cytokine family which is generated from lining and structural cells including endothelial cells, fibroblasts, and epithelial cells of the lungs when exposed to the environment. Nuclear cytokine IL-33 can be released during cell necrosis, suggesting that IL-33 may function as an alarmin that alerts the human immune system during physical stress, infection, and trauma. IL-33 activates basophils, eosinophils, macrophages, mast cells, and ILC2s through IL-33 receptor ST2 [12]. In paediatric patients who had severe steroid-resistant asthma, IL-33 promotes airway remodelling [13].

    IL-25: IL-25 previously was called IL-17E. The cytokine IL-17 family has 6 members, from IL-17A to IL-17F. IL-25 has a unique structure and function. The main TH2 cells highly express receptor of IL-25 [14]. The expression of IL-25 in airway epithelial cells is a key determinant of type 2 response activation in asthma. Plasma IL-25 level can reflect airway type 2 response and can be used as a factor to predict response to therapy [15]. Sputum IL-25 levels correlate with asthma severity [16].

    TSLP: Levels of TSLP protein and mRNA were found to increase in airways of asthma patients, and the level of expression correlated with disease severity [17],[18]. TSLP encodes a hemopoietic cytokine and signals through a heterodimeric receptor complex. The complex consists of the thymic stromal lymphopoietin receptor (TSLPR) and IL-7R alpha chain. TSLP induces the release of chemokines from monocytes and promotes the maturation of CD11c (+) dendritic cells (DCs) [19]. TSLP promotes TH2 cell reactions in asthma patients [20],[21]. The shorter isoform of TSLP has antifungal and antibacterial activities [22]. TSLP is mainly secreted by airway smooth muscle cells (ASMCs), epithelial cells, fibroblasts, keratinocytes, mast cells, stromal cells, monocytes, macrophages, granulocytes, and DCs [23],[24]. TSLP underlies asthma pathophysiology through multiple pathways to influence airway inflammation: for example, as the signal transducer and activator of transcription 3 (STAT3), transcription 5 (STAT5), nuclear factor kappa light chain enhancer of activated B cells (NF-kB), and mitogen-activated protein kinases (MAPKs) [25].

    TH2 low asthma patients have low TH2 cytokines in blood and are often not associated with systemic and airway eosinophils. These patients lack response to glucocorticoids and inhibitors of type 2 inflammation. One-third of SA patients are type 2 low, and this status is clinically challenging [26]. Type 1 T cells release cytokines including IL-2, IFNγ, and IL-12.

    IL-2: IL-2 affects Treg cells growth, survival, and activity [27], and it is a chemoattractant for eosinophils [28]. An indirect role of IL-2 is to regulate IL-5 production [29] that is involved in SA.

    IFN-γ: IFN-γ is an epithelial mediator with SA [30]. Type 1 T cells can release IFN-γ to influence cytokines CXCL9, CXCL10, and the transcription factor interferon regulatory factor 5 (IRF5) of macrophages/dendritic cells in SA [31],[32].

    IL-12: IL-12 normally opposes TH2 response, but it can contribute to allergic airway disease upon allergen exposure in the post-sensitization phase, by recruiting eosinophils and CD4+ T cells with up-regulation of TH2 chemokines, cytokines, and vascular cell adhesion molecule 1 (VCAM-1). IFN-γ-producing cells play an important role in this unexpected proinflammatory effect of IL-12 in allergic airway disease [33].

    Naïve T cells differentiate to TH17 cells with the presence of TGF-β, IL-6, IL-1β, and IL-23, leading to activation of STAT-3 and orphan nuclear receptor RORC2 [34]. In some SA patients, it has been found that increased neutrophils are associated with increased TH17 cytokines including IL-17A, IL-17F, and IL-22 in the bronchoalveolar lavage fluid (BALF). TH17 cytokines recruit neutrophils to the airway, induce mucous cell metaplasia, and have multiple effects on airway smooth muscle to result in narrowing of the airways [35].

    IL-22: IL-22 is released by TH17 cells and type 3 ILCs and exclusively functions on non-hematopoietic cells including epithelial cells. It displays a broad range of action in host protection and regeneration. IL-22 is involved in SA through airway allergic inflammation [36].

    IL-23: IL-23 is an IL-12-related cytokine. It is essential for the survival and functional maturation of TH17 cells involved in airway inflammation induced by antigens [37] in SA.

    The major cytokines and their roles in SA are listed in Table 1.

    Table 1.  Major cytokines and mediators in severe asthma.
    Endotypes involved Roles in severe asthma Biologic treatments
    TH2 cytokines and mediators
    IL-4 Type 2 high Regulating B cells to produce IgE that mediates allergic severe asthma Dupilumab
    IL-5 Type2 high Promoting eosinophil maturation, activation, survival, migration, and recruitment to airway Mepolizumab, Reslizumab, Benralizumab
    IL-13 Type 2 high Eosinophilic inflammation, mucus secretion, and airway hyperresponsiveness
    IgE Type2 high Mediating allergic severe asthma Omalizumab
    IL-33 Type2 high Acting as alarmin to activate allergic inflammation REGN3500
    IL-25 Type2 high Regulating internal safety of adaptive immune responses
    TSLP Type2 high Activating MAPKs, NF-kB, STAT3, and STAT5 to induce inflammatory response Tezepelumab
    Th1 cytokines and mediators
    IL-2 Type 2 low Influencing Treg cell growth, survival, and activity
    IFN-γ Type 2low Influencing expression of CXCL9 and CXCL10 as well as the macrophage/dendritic cell transcription factor IRF5
    IL-12 Type 2 high Counteracting Th2 sensitization and contributing to full-blown allergic airway disease upon airway allergen exposure in the post-sensitization phase, with enhanced recruitment of CD4+ T cells and eosinophils and with up-regulation of Th2 cytokines
    Th17 cytokines
    IL-17 Type 2 low Recruiting neutrophils to the airway by increasing secretion of epithelial-derived neutrophilic chemokines
    IL-22 Type 2 low Acting on non-hematopoietic cells including epithelial cells of mucosal surface and exhibits a broad range of action in regeneration and host protection
    IL-23 Type 2 high Essential for survival and functional maturation of Th17 cells and involved in antigen-induced airway inflammation

     | Show Table
    DownLoad: CSV

    For SA, human T and B lymphocytes are important immune cells in influencing the process. T helper cells release type 2 cytokines, and B cells produce IgE antibodies when stimulated by antigens or allergens through class switching [8]. Other cells, such as eosinophils and neutrophils, are all shown to have associations with SA. SA can then also be classified as the endotypes eosinophilic, neutrophilic, mixed, and paucigranulocytic. Mixed cell endotypes present with the characteristics of both eosinophilic and neutrophilic SA. Paucigranulocytic SA does not show any abnormalities in eosinophils or neutrophils in the patients' lungs or blood. Innate lymphoid (ILC) cells including ILC2, ILC3, airway smooth muscle cells and airway epithelial cells can also have roles in SA. Anti-inflammatory therapy may not work with patients who have paucigranulocytic disease [38]. The symptoms of these patients could be driven by airway hyper-responsiveness. These patients may benefit from the use of airway smooth muscle-directed therapies.

    Eosinophils are one of the major cell types in the development of asthma exacerbations. The TH2 cytokines network in the airways can be maintained with eosinophilic inflammation. It comprises a cascade consisting of vascular cell adhesion molecule 1 (VCAM-1), granulocyte-macrophage colony-stimulating factor (GM-CSF), CC chemokines, and eosinophil growth factors. Periostin is an extracellular matrix protein and is considered a biomarker of the TH2 immune response. It directly activates eosinophils in vitro. Adhesion of eosinophils to intercellular adhesion molecule (ICAM-1) on airway epithelial cells can upregulate the functions of eosinophils. ICAM-1 is a receptor of rhinovirus (RVs). In virus-induced asthma, virus infection can upregulate the expressions of cysteinyl leukotrienes (cysLTs) and CXCL10. CysLTs can directly induce eosinophilic infiltration in vivo, and CXCL10 can activate eosinophils. Eosinophil activation has several mechanisms to contribute to the development of SA including (1) through TH2 cytokines (e.g., IL-5 or GM-CSF), (2) through proteins induced by viral infection (e.g., CXCL10), and (3) through interaction with other cells (e.g., neutrophils) [39]. Eosinophils of greater than 3% in sputum can indicate a response to corticosteroids treatments or biologics therapies against type 2 cytokines [38].

    Neutrophils are one other major cell type in the development of SA. In some patients, asthma severity is associated with airway neutrophilia. Neutrophilia correlates with asthma refractory to corticosteroid treatment [40]. Neutrophils can release many cytokines and chemokines after stimulation [41]. An increase in neutrophil counts has been reported in the sputum of adults with persistent asthma and of children with acute asthma exacerbations [42],[43]. IL-8 is a key chemoattractant for neutrophils, and has been found to be upregulated in some cases of SA. Lipopolysaccharide (LPS) can induce release of IL-8 from airway epithelial cells, and IL-8 levels were also found to be raised in the lower airways of asthma patients who were corticosteroid-resistant. Even without chemo-attractants for eosinophils, IL-8 or LPS can stimulate neutrophils to increase the trans-basement membrane migration of eosinophils. Neutrophil SA may represent a non-T2-driven condition, and it can be a predictor of response to antibiotics and as well as possibly a predictor of treatment targeted at pathways that lead to neutrophil recruitment [38].

    ILCs are derived from common lymphoid progenitors (CLPs). The ILCs can be classified into three groups based on the differences in their transcription factor and cytokine profiles [44]. ILC2 cells are a novel family of haematopoietic effector cells. ILC2 cells specifically produce type 2 cytokines and depend on GATA3 and retinoic acid-related orphan receptor alpha (RORα) for their functions [7]. ILC2 cells in SA demonstrate an increase in proliferative capacity, expression of TSLP receptor (TSLPR), GATA3, and NFATc1 protein, enhancing IL-5 and IL-13 release. ILC2 cells can release IL-6 after stimulation. ILC2 cells have active phenotypes in severe allergic and eosinophilic asthma, which include increased proliferation; TSLPR, GATA3 and NFATc1 expression; and increased IL-5, IL-13 and IL-6 levels. Mepolizumab for anti-IL-5 can decrease markers of ILC2 cell activation [45].

    ILC3 cells are constitutively presented at mucosal barrier sites in many organs such as the lungs, gut, spleen, liver, and skin [46]. In an asthma model from obese mice, ILC3 cells have been found to be involved in the development of airway hyperreactivity (AHR) induced by IL-1β. Blockade of IL-1β reduces ILC3 cell numbers and abolishes the AHR [47]. In lungs from non-allergic neutrophilic asthma mice, ILC3 cell numbers are found to be increased [48]. IL-17+ ILC cells are also found in bronchoalveolar lavage fluid (BALF) from human SA patients [47]. ILC3 cells are elevated in patients with non-eosinophilic asthma and associated with neutrophilic inflammation from the release of neutrophil chemo-attractants. They are also resistant to steroid treatment [49].

    ASMCs are effector cells for bronchoconstriction in asthma and are responsible in secreting chemokines and cytokines in inflammatory responses. Small G proteins of the Rho family, including RhoA, Rac1, and Cdc42, are major functional regulators of ASMCs. In patients with SA, Rac1 is overactive and is essential for ASMC proliferation [50]. Drugs directed at ion channels on the smooth muscle membrane have been found to be ineffective in SA [51]. ASMCs are major targets for both β2-agonist and ICS treatments, but ASMCs derived from SA patients have similar levels to mild asthma patients in fixed airflow obstruction (FAO) responding to β2-agonists and corticosteroids [52]. This suggests ASMCs may have other mechanisms of ASMCs except airflow obstruction in SA patients.

    Table 2.  Major cell endotypes in severe asthma.
    Cells Endotypes involved Roles in severe asthma
    Eosinophils Eosinophilic TH2 network, which comprises a cascade of vascular cell adhesion molecule-1/CC chemokines/eosinophil growth factors, including granulocyte-macrophage colony-stimulating factor (GM-CSF), involving virus induced severe asthma
    Neutrophils Neutrophilic Can release many cytokines and chemokines after stimulation, helping recruit cytokines TNF, IL-1, IL-6, IL-8, IL-23, and IL-17
    ILC2 Eosinophilic Producing type 2 cytokines and depends on GATA-binding protein 3 and retinoic acid-receptor-related orphan receptor-a for their development and proliferative capacity, TSLP receptor (TSLPR), and NFATc1 protein expressions and increasing IL-5 and IL-13 release
    ILC3 Neutrophilic Influencing non-allergic neutrophilic asthma and associating with neutrophil inflammation by release of neutrophil chemo-attractants and were glucocorticoid resistant
    Airway smooth muscle cells Paucigranulocytic Major effector cells of bronchoconstriction in asthma and contribute to the inflammatory process by secreting pro-inflammatory cytokines
    Airway epithelial cells Paucigranulocytic Recognizing microbes, parasites, and allergens as well as alarmins/damage-associated molecular patterns; promoting release of pro-inflammatory cytokines/chemokines, including IL-6, IL-8, CCL20, CCL17, TSLP, IL25, IL-33, and GM-CSF

     | Show Table
    DownLoad: CSV

    The airway epithelial cells (AECs) constitute an efficient physical barrier in human lungs. The cells are the first line of defence against allergens, airborne irritants, and microorganisms. The most prominent character of AECs is that the cells express various important receptors in order to recognise dangers and foreign invading proteins. These receptors include toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), pattern recognition receptors (PRRs), nucleotide-binding oligomerisation domain (NOD)-like receptors (NLRs), C-type lectin receptors (CLRs), protease activated receptors (PAR)-2, and purinergic receptors [53]. These receptors recognise pathogen-associated molecular patterns (PAMPs) from inhaled allergens, microbe parasites, alarmins, and damage-associated molecular patterns (DAMPs) from death cells. After recognition, AECs can activate downstream signalling to release inflammatory chemokines and cytokines including IL-6, IL-8, IL-25, IL-33, CCL17, CCL20, TSLP, IL25, and GM-CSF [54]. Genome wide association studies (GWASs) have identified a few genes that express in human airway epithelium and associated with SA such as ORMDL3, GSDMB, and TSLP [55]. ORMDL3 mediates sphingolipid synthesis, cell stress, glycolysis, and viral infections. One of the functional roles of GSDMB in asthma path-physiology is to regulate cell pyroptosis, a specific type of cell death through GSDMB's N terminals to form pores on the epithelial cell membrane. TLSP works as an important driver of type 2 cytokine response in SA [56],[57].

    The major endotypes of cells in SA are listed in Table 2.

    The triggers of SA come from viral, bacterial, and fungal infections, allergens, smoking, and pollutants.

    Most asthma exacerbations are associated with viral respiratory infections. Viral infections are closely associated with wheezing illnesses in children. The major viruses that trigger SA are human rhinovirus (HRV), respiratory syncytial virus (RSV), influenza virus, and SARS-CoV-2 virus.

    HRV: HRVs are positive-sense, single-stranded-RNA (ssRNA) viruses that belong to the family Picornaviridae and the genus Enterovirus. Based on phylogenetic sequence, HRVs are classified as three species: HRV-A, HRV-B, and HRV-C. HRV-A and HRV-B species can be cultured in normal culture. HRV-C strains cannot be cultured in standard culture medium, although HRV-C strains have a genomic organisation like that of HRV-A and HRV-B [57]. Approximately 60% of asthma exacerbations have been linked with HRV infections [58]. HRV-C infections are responsible for the majority of asthma attacks in children that result in hospitalisation [59]. Viral upper respiratory tract infections with HRV in adults are the most common cause of asthma exacerbation [60]. Most of its serotypes enter epithelial cells in the respiratory tract via binding to ICAM-1 receptor [61][63] while a minority of them use the low-density lipoprotein (LDL) receptor for virus entry. It normally infects the upper respiratory tract, causing the common cold. However, in patients with asthma, their asthma symptoms get worse, and it could also infect the lower respiratory tract [64]. In fact, the genetic variants of ORMDL3 are linked with significant increases of wheezing episodes induced by HRV infection in children. In asthma patients, elevated levels of ICAM-1 expression increase their susceptibility to HRV infection as ICAM-1 promotes binding of HRV and entry of viral components into the host cells.

    RSV: Human RSV is an enveloped, non-segmented negative-stranded RNA virus of the family Paramyxoviridae. RSV can induce wheezing illnesses during infancy. The effect of the infection can influence respiratory health for years [65]. RSV infections can damage human airways and cause obstruction and recurrent wheezing. Palivizumab is an anti-RSV monoclonal antibody that can decrease the risk of severe illness and wheezing induced by RSV [66]. Investigating the mechanisms of severe viral illnesses caused by RSV might lead to new treatments for viral wheezing illnesses and may reduce the risk for SA [67].

    Influenza virus: Influenza viruses are members of the family Orthomyxoviridae. This family represents enveloped viruses consisting of negative-sense single-strand RNA segments in the genome [68]. In 2009, an influenza virus pandemic was highly transmissible and caused severe respiratory disease. The influenza pandemic was co-morbid with asthma, and this resulted in hospitalisations. Animal studies showed pandemic H1N1 infection had characteristics of allergic airway disease and had signs of asthma exacerbation. It was thought that although the immune response caused exacerbations, it occurred actually to protect the host from severe outcomes caused by influenza [69].

    SARS-CoV-2 virus: SARS-CoV-2 is a positive-sense single-stranded RNA virus [70]. SARS-CoV-2 virus invades lung cells through the angiotensin-converting enzyme 2 (ACE2) receptor. As IL-13 can decrease ACE2 expression, asthma would not be a significant risk factor for severe COVID-19. However, several SA-associated risk factors might occur in both SA and COVID-19. Although the respiratory symptoms of asthma related to COVID-19 were mild to moderate, some patients had chronic worsening of their asthma symptoms and required an increment in their medications to control the conditions [71],[72].

    In clinical practice, bacterial organisms are also contributors to asthma exacerbations, but they have received much less attention than viruses for SA. Asthma exacerbations can be caused by Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, and Tropheryma whipplei [73],[74].

    Streptococcus pneumoniae: S. pneumoniae is commonly found in the nasopharynx [75] in what is usually the initial step of infection. Carriage of S. pneumoniae is most prevalent in children [76], and it declines with increasing age [77]. This may be due to acquired immunity from exposure. A Prospective Study on Asthma in Childhood (PSAC) found that the infants colonised with S. pneumoniae had an increased risk of a first wheezy episode. These infants can develop persistent wheeze or asthma during follow-up. Early nasopharyngeal carriage of S. pneumoniae was also linked to increased eosinophil counts in blood, higher total IgE in serum, airway reversibility, and subsequent asthma [78][81].

    Haemophilus influenzae: Nontypeable Haemophilus influenzae (NTHi) is a respiratory tract pathobiont that can chronically colonise the airways of patients. It is often associated with neutrophilic SA [82]. In SA patients, NTHi persistence with pulmonary macrophages can induce TH17 response and cause chronic airway inflammation, leading to decreased lung function and reduced effectiveness of steroid treatments. Reducing the burden of NTHi could improve the outcomes of managing SA patients in clinic [83].

    Moraxella catarrhalis: Moraxella catarrhalis is also more abundant in patients with severe neutrophilic asthma. It has been found that specific genes have been linked to inflammasome and neutrophil activation in SA patients with Moraxella catarrhalis infections [84].

    Tropheryma whipplei: Tropheryma whipplei belongs to Actinobacteria but has reduced size in genome. The abundance of Tropheryma whipplei is associated with gene expression of IL-13 type 2 and ILC2 signatures and correlate positively with sputum eosinophils in SA. How the bacterial species drive the inflammatory response in SA needs further evaluation [84].

    Pseudomonas aeruginosa (P. aeruginosa) is a ubiquitous and opportunistic microorganism and is one of the most significant pathogens that produce chronic colonisation and infection of the lower respiratory tract, especially in people with chronic inflammatory airway diseases such as asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), and bronchiectasis. The role of P. aeruginosa in SA also requires further investigation [85].

    Many airborne fungi can also trigger SA. These fungi include species of Alternaria, Aspergillus, Cladosporium, and Penicillium [86]. Some mould allergens can produce more severe airway disease than other common allergens. In daily human life, airway exposure to airborne fungal spores is almost constant. C. herbarum, A. alternata, and A. fumigatus are the most airborne fungi. Comparing to allergens such as house dust mites, grass pollen, and cat dander, fungi cause disease not only as sources of allergenic proteins but also by having the additional ability to colonise the respiratory tract and on the host skin. Therefore, fungi have a much greater impact on human SA for triggering host defences and producing nonallergenic toxins and enzymes. Asthma patients with severe disease who are sensitized to one or more fungi can be defined as having SA with fungal sensitization (SAFS) [87]. SAFS is a relatively new classification of allergic asthmatic subjects, but it is gradually becoming more recognised [88]. Identifying fungal cases of SA is important, and treatment with imidazole antifungals can provide significant benefit. Antifungal treatment is a key part of the successful management of SAFS [89].

    Allergens can induce SA through IgE mediated allergic response. Inhalant allergens can cause chronic SA. Recent studies found that high titre IgE allergic response, particularly to dust mites, was associated with asthma exacerbations with rhinovirus infection. As we discussed above, it is now well recognised that the fungus Aspergillus can colonise the human airway and cause SA in some patients [89].

    Cigarette smoking can be a predictor of poor asthma control and asthma severity [67].Cigarette smoking exposure can impact asthma development in children [90]. Cigarette smoking may induce a neutrophil-predominant inflammation of the airways [91], which may make patients resistant to asthma treatment [92]. Persistent cigarette smoking induces both allergic TH2-driven inflammation [93] and TH1-mediated inflammatory responses in SA patients [94]. E-cigarette use can have significant associations with asthma in adolescents [95]. However, how vaping and e-cigarette use influence SA needs to be investigated further.

    In a French epidemiological study on the relation between SA and air pollutants, the relationships between air nitrogen dioxide (NO2), sulfur dioxide (SO2), and ozone (O3) concentrations and SA were investigated. Asthma severity was significantly related to O3 concentrations, while it was unrelated to NO2. SO2 was associated with asthma severity only for the model-based assignment of exposure [96]. Inhaled O3 could play a part for both acute and chronic inflammation in the lungs. O3 could induce TH2 pattern response [97] and eosinophilic airway inflammation [98]. Volatile organic compounds (VOCs) are ubiquitous domestic pollutants, but their role in asthma exacerbations is uncertain. In a systematic analysis of 11 databases and three repositories for the relationships between VOC exposure and exacerbations of asthma, the results were inconsistent [99].

    The triggers and their possible roles in SA are listed in Table 3.

    Table 3.  Most common triggers for severe asthma.
    Triggers Roles in severe asthma
    Virus
    Human rhino virus Regulating ICAM-1 expression in airway epithelial cells in asthma patients increasing their susceptibility to HRV infection as ICAM-1 promotes binding of HRV and entry of viral components into the host cells
    Respiratory syncytial virus Inducing bronchiolitis to damage the airways, promoting airway obstruction, and recurrent wheezing
    Influenzas virus Governing immune responses that cause exacerbations
    SARS-CoV-2 virus Subgroup of these asthmatic patients evolved with a chronic worsening of their asthma requiring an increment in asthma medication to control the disease when having COVID-19
    Bacteria
    Streptococcus pneumoniae: Increasing blood eosinophil count, higher total serum IgE, and airway reversibility
    Hemophilus influenzae: Contributing to chronic airway inflammation and TH17 responses in severe asthma, which can lead to decreased lung function and reduced steroid responsiveness.
    Moraxella catarrhalis: Abundant in severe neutrophilic asthma and genes linked to inflammasome and neutrophil activation
    Tropheryma whipplei Associating with TH2 response in SA
    Fungi
    Alternaria, Aspergillus, Cladosporium and Penicillium Acting as allergens, also having the additional ability to actively germinate and infect the host skin or attempt to colonise the respiratory tract to induce SA
    Allergens IgE mediates severe asthma
    Smoking Induce a neutrophil-predominant inflammation of the airways
    Pollutants O3 induces a TH2 pattern response and eosinophilic airway inflammation

     | Show Table
    DownLoad: CSV

    A majority of biological therapies for SA are antibody treatments against IgE and type 2 airway inflammation. These biologics include omalizumab against IgE, dupilumab against IL-4Rα, mepolizumab and reslizumab against IL-5, benralizumab against IL-5Rα, tezepelumab against TSLP, REGN3500 against IL-33, and fevipiprant against PGD2 [100]. Biologics have some interesting results and could be the potential treatment for uncontrolled asthma. Optimizing the standard therapies of biologics is currently ongoing and the appropriate using biologics can decrease asthma-related morbidity [101]. Although current biologics have effects in the control of SA through improving FEV1 to decrease eosinophil counts, SA patients with type 2 low or paucigranulocytic disease are still very hard to control.

    Multi-omics refers to applying modern technologies such as next generation sequencing, systemic protein profiling, and high output measurements of small molecules to carry out cutting-edge approaches that combine data from different biomolecular levels, such as DNA, RNA, proteins, metabolites, and epigenetic markers, in order to obtain a holistic view of how living systems work and interact [102]. In the review, we only report the progresses in genomics approaches, transcriptomics approaches, proteomics approaches, metabolomics approaches, and metagenomics approaches for SA.

    Genomics approaches to SA have been achieved by GWASs. More than ten genes were identified to have association with SA worldwide. The loci include THSD4, IL33 [103], MUC5AC [104], FLJ22447 [105], ORMDL3/GSDMB, and IL1RL1/IL18R1 [106]. THSD4 (thrombospondin type 1 domain containing 4) is an extracellular matrix protein and has roles in microfibril formation to maintain the structural integrity of human lungs [107]. MUC5AC has pathogenic roles in airway hyper-responsiveness and mucus plugging during asthma exacerbation in human lungs [108]. IL-33, ORMDL3, GSDMB, and IL1RL1/IL18R1 are also reported to have associations with asthma through previous GWAS studies [109],[110]. The most potential therapeutic targets are ORMDL3 and GSDMB. ORMDL3 medicates HRV infection through regulating ICAM-1 receptor to induce SA [57]. GSDMB works as a key molecule in pyroptosis to regulate cell inflammation [111]. Ongoing study into the function and inhibition for products of these genes would play a valuable role in discovering new therapeutic targets in the pathways that are involved in the pathophysiology of SA.

    The transcriptomic approaches for SA include sequencing RNA samples from blood, sputum, and brushed cells from the lungs.

    Circulation: Expression of RAR-related orphan receptor A (RORA) is significantly higher in patients with SA. SA patients have decreased glucocorticoid receptor signalling and increased activity of mitogen-activated protein kinase (MAPK) and Jun kinase [112]. SA is found to be linked with the activation of circulating CD8+ T cells. The response is correlated with the downregulation of miR-146a/b and miR-28-5p [113]. TH2-biomarker transcriptomic signature was found in SA patients, but with no signature in association with TH2-biomarker-low patients [114].

    Sputum: TH2-driven eosinophilic and non-TH2 phenotypes of both neutrophilia or non-inflammatory paucigranulocytic patterns have been identified in sputum transcriptomics [115]. A signature of six-gene expression biomarkers (CLC, CPA3, DNASE1L3, IL1B, ALPL and CXCR2) in sputum can predict response to corticosteroid managements, and the six gene expression levels predict SA better than sputum and blood eosinophils [116]. In a separate cohort of SA patients, an enrichment of genes was shown to be involved in the p38 signalling pathway in SA. Phosphorylation of p38 was increased in some patients and can associate with neutrophilic airway inflammation. p38 activation may have roles in steroid-resistant inflammation, and it can continue recruitment of neutrophils to the airways in SA [117].

    Bronchial brush samples: High baseline expression levels of calcium-activated family member 1 (CLCA1), periostin, and serpinB2 are found to be linked with a better clinical response to corticosteroids. Meanwhile, high expression levels of FK506-binding protein (FKBP51) are linked with a poor response to corticosteroids. IL-13 increases expressions of CLCA1, periostin, and serpinB2. Corticosteroids induce expression of FKBP51 [118]. Transcript analysis from bronchial brush samples of SA patients identified that the dysregulation of neural signals, apoptosis, REDOX, and O-glycan process are linked to disease severity. In non-TH2 subtype SA patients, the neural signals and IL-26-related co-expression module are significantly dysregulated. These genes encode proteins related to transmembrane transport, the apoptotic process, O-glycan processing, and amino acid biosynthetic process [119]. Genes associated with SA were enriched in co-expression modules for NK cell-mediated cytotoxicity and interleukin production. In an integrated analysis of systemic and local airway transcriptomes, NKG7 and perforin (PRF1) are identified as key drivers in SA. Nasal genes are enriched in the tricarboxylic acid (TCA) cycle module in SA patients. Network analyses identified G3BP stress granule assembly factor 1 (G3BP1) and InaD-like protein (INADL) are key nasal drivers for SA [120].

    Proteomic analysis involves systemically investigating the identification and quantification of the complete compositions of proteins in a biological system (e.g., cell, tissue, and organ). Most samples for proteomic approaches in SA are from plasma and sputum.

    Plasma: SA patients are found to be distinguished from each other by 365 different protein abundancies and the plasma proteome defines distinct patients' endotypes within SA. These proteins included HSP90A, PTPN11, CCL5, IL4, CD86, FGF9, IDG15, NME2, IFNG, and C3. Some groups of proteins are significantly enriched in the exosomal markers of immune and inflammatory cells [121].

    Sputum: In SA patients, the protein levels of colony-stimulating factor (CSF) 2 are higher in current smoker sputum supernatants; the levels of azurocidin 1, neutrophil elastase, and CXCL8 are found to be higher in ex-smokers [122]. In another experiment in proteomic screening, a significant increase of 23 proteins is observed in the sputum in SA patients. These proteins include A2M, APOA2, ELANE, GPI, S100A8, S100A9, and S100A12. These proteins have roles in multiple biological processes, including immunity, inflammatory, transport, protease activity, protease inhibition, metabolism, hydrolase activity, and vasculogenesis [123].

    Metabolomics systematically studies low molecular weight bio-chemicals in a given biological system [124]. In SA, metabolites can play an important role in disease and homeostasis from redox balance, oxidative stress, signalling, apoptosis, and inflammation [125]. All cell types involved in SA can produce and secrete exosomes that contain proteins, nucleic acids, lipids, cytokines, growth factors, and co-stimulatory molecules [126]. Amino acids such as glycine, glutamine, and glutamate may have protective effects for SA, whereas phenylalanine can have adverse effects [127]. High FENO is demonstrated in most of the asthma population, and it is associated with greater arginine metabolism in severe and reactive asthma patients [128]. In animal models with iNOS deletion, eosinophilic inflammation is regulated by arginine metabolism. Arginine inhibits inflammation, while iNOS metabolism promotes airway inflammation [128]. Systemic IL-6 inflammation is demonstrated in obese asthma patients and also in a small subset of non-obese patients. These patents were found to have more severe asthma. IL-6 inhibitors may reduce metabolic dysfunction in some SA patients [129].

    Table 4.  The potential biologic targets identified by omics for severe asthma.
    Genes or molecules Roles in severe asthma
    Genomics THSD4, IL33, MUC5AC, FLJ22447, ORMDL3/GSDMB, and IL1RL1/IL18R1 Extracellular matrix protein that is involved in microfibril formation, Cytokine alarm, airway hyper-responsiveness and mucus plugging, HRV infection, and pyroptosis
    Transcriptomics
    Circulation RORA: CD8+ T2 signature Glucocorticoid receptor signalling activity of the mitogen-activated protein kinase and Jun kinase cascades; T2 response
    Sputum A six-gene expression biomarker signature (CLC, CPA3, DNASE1L3, IL1B, ALPL and CXCR2) Predicts response to oral corticosteroids in SA
    Bronchial brush FKBP51, CLCA1, periostin, serpinB2, and IL-26 Neural signal, REDOX, apoptosis, and O-glycan process in SA
    Proteomic
    Plasma 365 proteins including HSP90A, PTPN11, CCL5, IL4, CD86, FGF9, IDG15, NME2, and IFNG Immune-inflammatory; exosomal markers
    Sputum 23 proteins including A2M, APOA2, ELANE, GPI, S100A8, S100A9, and S100A12 Immune-inflammatory: immunity, inflammatory, chemokines, protease, protease inhibitor, metabolism, transport
    Metabolomic Glycine, glutamine, glutamine
    Phenylalanine
    Arginine
    IL-6
    Protective effects in SA
    Adverse effects in SA
    Regulating FENO
    Obese SA
    Metagenomics Actinobacteria
    Proteobacteria
    Klebsiella
    Indicator of steroid responsiveness
    Regulating expression of TH17-related genes
    Indicating asthma severity

     | Show Table
    DownLoad: CSV

    Next-generation sequencing (NGS) techniques perform a more accurate microbial characterization based on genetic contents. Targeted sequencing of the 16S ribosomal RNA (16S rRNA) gene has been routed for the application of investigating the bacterial abundance and diversity of the microbiota [130]. A change in the nasopharyngeal or the salivary microbiome has been found to be associated with different phenotypes of SA [131]. As we discussed above, respiratory pathogens can be triggers for asthma exacerbations due to airway inflammation [132],[133].

    In patients with SA, the composition of bronchial bacteria is associated with several disease-related features. These features include body mass index, bronchial biopsy eosinophil values and sputum total leukocyte count. Actinobacteria is associated with improving asthma scores and bronchial epithelial gene expression of FK506 binding protein (FKBP5), an indicator of steroid treatment. Proteobacteria has negative correlations with biopsy eosinophil values. It is also associated with expression of the TH17-related genes. Specific microbiotas are associated with and may regulate inflammatory processes in SA patients and associated phenotypes [134].

    The novel genes and molecules identified by transcriptomic, proteomic, metabolomic, and metagenomic approaches for SA are listed in Table 4.

    Management of severe asthma, clinically, is a challenge due to the heterogeneity of the conditions. Cytokines, inflammatory cells, and various triggers can interact together in disease developments. The current biologics available show great advantages for treating the condition, although there is still a necessity to optimise the criteria of how and when they should be applied in clinic. The multi-omics approaches for SA identify many novel molecules and genes that could be biomarkers and therapeutic targets for the condition. The next stage for research on SA will be investigating the mechanisms for these novel genes and molecules, integrating the data from multi-omics approaches, and validating the value of biomarkers and treatment targets. Investigations into the pathways of novel genes and molecules in SA would be of great help in developing personal medicine to treat SA cases in the future.


    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(1894) PDF downloads(130) Cited by(0)

Figures and Tables

Tables(4)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog