Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1833 2023-12-15 05:37:50 |
2 Reference format revised. Meta information modification 1833 2023-12-18 07:32:22 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Savin, I.A.; Zenkova, M.A.; Sen’kova, A.V. Bronchial Asthma as a Precursor of Lung Fibrosis. Encyclopedia. Available online: https://encyclopedia.pub/entry/52785 (accessed on 08 July 2024).
Savin IA, Zenkova MA, Sen’kova AV. Bronchial Asthma as a Precursor of Lung Fibrosis. Encyclopedia. Available at: https://encyclopedia.pub/entry/52785. Accessed July 08, 2024.
Savin, Innokenty A., Marina A. Zenkova, Aleksandra V. Sen’kova. "Bronchial Asthma as a Precursor of Lung Fibrosis" Encyclopedia, https://encyclopedia.pub/entry/52785 (accessed July 08, 2024).
Savin, I.A., Zenkova, M.A., & Sen’kova, A.V. (2023, December 15). Bronchial Asthma as a Precursor of Lung Fibrosis. In Encyclopedia. https://encyclopedia.pub/entry/52785
Savin, Innokenty A., et al. "Bronchial Asthma as a Precursor of Lung Fibrosis." Encyclopedia. Web. 15 December, 2023.
Bronchial Asthma as a Precursor of Lung Fibrosis
Edit

Bronchial asthma is a heterogeneous disease characterized by persistent respiratory system inflammation, airway hyperreactivity, and airflow obstruction. Lung fibrosis is a common occurrence in the pathogenesis of fatal and long-term asthma, and it is associated with disease severity and resistance to therapy.

asthma lung fibrosis

1. Introduction

Asthma is the most common chronic inflammatory disease of the respiratory tract, characterized by leukocyte infiltration and tissue remodeling, with the latter generally referring to epithelial hyperplasia and collagen deposition [1]. Clinically, asthma is accompanied by airway inflammation, hyperresponsiveness, and airflow limitation, which can lead to respiratory symptoms such as coughing, wheezing, and shortness of breath [2]. The pathogenesis of asthma is complex and involves various genetic, environmental, and immunological factors [3][4].
One of the central features of asthma is airway remodeling, defined as changes in airway wall structure, including extensive epithelial damage, airway smooth muscle hypertrophy and hyperplasia, collagen deposition, subepithelial basement membrane thickening, and fibrosis [5]. Excessive proliferation of smooth muscle cells producing a wide range of pro-inflammatory and pro-fibrotic mediators may lead to amplified airflow obstruction and extracellular matrix (ECM) deposition, ultimately resulting in fibrosis in individuals affected by asthma [6][7]. Subepithelial fibrosis observed in asthma is associated with enhanced differentiation of bronchial fibroblasts into myofibroblasts—fibroblast-to-myofibroblast transition (FMT)—induced mainly by transforming growth factor-β (TGF-β) [8].
Fibrosis of alveolar structures is an important phenomenon, as it usually occurs in the pathogenesis of fatal and long-term asthma and may be associated with disease severity and resistance to therapy [9][10][11]. In asthma treatment, drugs targeting chronic inflammation and bronchodilators control asthma but have a negligible effect on the structural changes in the bronchi. Recent studies indicate that inflammation and remodeling of asthmatic bronchi can be driven independently [8][12]. Therefore, lung fibrosis can be considered a long-term and eventually irreversible consequence of asthma-induced airway inflammation and remodeling.

2. Bronchial Asthma as One of the Precursors of Lung Fibrosis: Etiology, Pathogenesis, and Morphological Characteristics

Bronchial asthma is a heterogenic disease characterized by persistent inflammation in the respiratory system, airway hyperreactivity, and reversible airflow obstruction, affecting approximately 300 million people worldwide [13][14]. Trends in asthma prevalence have fluctuated throughout the last decades; the overall number of asthma cases has remained consistent, though asthma-related deaths have decreased in recent years, reflecting improved therapeutic control. However, despite advances in modern healthcare, global asthma damage remains high, with about 450,000 asthma-related overall deaths and an economic burden that costs USD 50 billion annually [15][16]. In addition, it is one of the most widespread chronic lung pathology among pediatric patients [17].

2.1. Asthma Endotypes and Phenotypes

At the present time, asthma is considered an “umbrella” diagnosis, unifying several diseases with different clinical manifestations (phenotypes) and pathophysiological mechanisms (endotypes) [18]. According to the Global Initiative for Asthma (GINA) definition, “asthma phenotypes” are recognizable clusters of demographic, clinical, and/or pathophysiological characteristics [19], while the term “endotypes” describes a subset of asthma with distinct molecular mechanisms and treatment response [20]. Today, the best-researched type of asthma is eosinophilic asthma, the most common type of this disease, also called T2-high endotype [21]. The T2-high endotype includes the following phenotypes: early-onset atopic (responsive to steroids), late-onset non-atopic eosinophilic (refractory to steroids), and aspirin-exacerbated respiratory disease (surgical treatment, sensitive to leukotriene modifiers).
Early-onset atopic asthma is the archetypal asthma phenotype, with a well-defined early onset indicated by blood or sputum eosinophil count, serum IgE, high FeNO, and high total IgE, and is sensitive to inhaled corticosteroids (ICS) therapy. It is distinguished from T2-high non-atopic asthma by positive skin prick tests and increased IgE [22].
Late-onset eosinophilic asthma is a subset of T2-high asthma manifesting in adulthood, notable for its higher severity and steroid resistance. The majority of these patients also have comorbid chronic rhinosinusitis with nasal polyps. Generally, this phenotype is characterized by prominent blood and sputum eosinophilia refractory to ICS treatment and normal or slightly elevated serum IgE levels. Additionally, it is believed that inflammation in this phenotype is driven by the production of IL-5 and IL-13 by innate lymphoid cells. Some patients also have sputum neutrophilia, indicating that Th2/Th17 interactions are taking place [23].
Aspirin-exacerbated respiratory disease (AERD) is a subset of late-onset eosinophilic asthma, characterized by dysregulated arachidonic acid metabolism, cysteinyl leukotrienes production, elevated eosinophils in the blood and sputum, high severity from the onset, and frequent exacerbations. Aspirin is a potent cyclooxygenase inhibitor, and it shifts arachidonic acid metabolism from the cyclooxygenase to the 5-lipooxygenase pathway. This leads to the overproduction of cysteinyl leukotrienes, which are potent bronchoconstrictors responsible for the clinical symptoms and resistance to conventional therapy for AERD [24].
Non-eosinophilic asthma, also known as T2-low or non-T2, is a less understood endotype of asthma and is typically defined by the absence of T2-high asthma signs, such as eosinophilia and elevated IgE, the presence of neutrophilic or paucigranulocytic inflammation, and resistance to ICS. Mechanisms underlying the manifestation of T2-low asthma and the maintenance of neutrophilic inflammation are currently unknown, but they have been associated with chronic infection, obesity, smoking, and smooth muscle abnormalities [25]. Therapeutic options are quite limited and consist of tiotropium and macrolides [26].

2.2. Etiology and Pathogenesis of Bronchial Asthma

The etiology of bronchial asthma is currently unknown; however, there are plenty of risk factors, including genetic and environmental conditions [27]. Genetic factors include changes in the expression of several genes responsible for protein folding in the endoplasmic reticulum [27], epithelial [28], and eosinophil dysfunction [29]. Allergic airway diseases (such as allergic rhinitis) are also associated with an increased risk of asthma development [30]. Environmental factors include smoking (both active and passive) [31], air pollution (including automobile associated, such as black carbon and NO2) [32], obesity [33], and professional risk factors, such as flour dust, animal and plant enzymes, tree resins, tobacco, polyisocyanate, acids, anhydrides, and metals [34][35].
Allergic asthma is considered one of the most widespread asthma types, developing due to sensitization to environmental allergens, mostly house dust, plant pollen, and mushroom spores [36]. After sensitization, asthma symptoms usually develop during second contact with the allergen [37]. Allergic reactions, activating the IgE dependent pathways, are the most common mechanism underlying asthma.
IgE is the main effector of type 1 hypersensitivity, underlying the development of asthmatic inflammation [38]. Its synthesis occurs either by direct class-switch recombination from IgM in germinal centers or through a “sequential” switch from IgM to IgG1 and then from IgG1 to IgE outside of germinal centers. The high-affinity receptor of IgE (FcεRI) is expressed on mast cells and basophils as a tetramer and on monocytes and dendritic cells as a trimer.
During the sensitization step of asthma development, IgE focuses the allergen on the cell surface through FcεRI, leading to the procession of the antigen-IgE complex and presentation through the major histocompatibility complex class II molecules, lowering the threshold for T-cell activation during the allergen challenge [39]. During the next contact with the allergen, inflammation is initiated when the antigen contacts IgE, presenting on all mast cells and basophiles. After contact, cells degranulate, releasing such mediators as histamine, heparin, proteases, and pro-inflammatory cytokines, which are responsible for the chemotaxis of inflammatory cells.
In addition to classic IgE, there is a cytokinergic IgE that facilitates asthmatic inflammation in the absence of allergens, making allergen avoidance an ineffective therapeutic strategy [40].
CD4+ lymphocytes also take part in the development of allergic asthma. After contact with antigen, T helpers type 2 (Th2) secrete pro-inflammatory cytokines, such as IL-4, IL-5, IL-9, and IL-13, which stimulate IgE production and inflammatory cell migration [41][42][43][44][45]. In turn, T helpers type 1 (Th1) start to secrete IL-2 and IFN-γ, activating macrophages and enhancing the cell immune response. T-cell immune response is additionally controlled by IL-1, IL-4, IL-12, and IL-18, secreted by dendritic cells [46]. The cascade of the aforementioned reactions leads to persisting inflammation in the lungs.
About one-third of bronchial asthma patients are believed to have non-allergic asthma, mediated by non-Th2 cytokines, including IL-17 and TNF-α, and characterized by the absence of allergen reactions in the skin prick tests and a decreased or unaffected amount of IgE, contrary to allergic asthma [47][48]. The mechanisms of non-allergic asthma development are currently unknown, but it is supposed that there are two parts to its pathogenesis: dysregulation of the neutrophilic immune response due to lung inflammation [49], and defects in IL-17 mediated signaling pathway [50], leading to the persisting inflammation [51].
Another group of cells that play a significant role in asthma and post-asthmatic fibrosis development are innate lymphoid cells (ILCs). It is a group of loosely related lymphocytes, characterized into five subgroups based on functions, origins, transcription factors, and cytokine expression patterns: natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue-inducer cells [52]. They are abundantly present in the tissue of organs performing barrier functions such as intestines, lungs, and skin. For quite some time, ILC2s have been established as crucial mediators of lung allergy, airway inflammation, and fibrosis, thus affecting the pathogenesis and clinical course of many respiratory diseases, like, for instance, asthma, cystic fibrosis, and chronic rhinosinusitis [53]. More specifically, ILC2s are activated by the alarmin cytokines IL-22 and IL-33, produced by the lung epithelium after contact with allergens, infections, and other injurious stimuli. After activation, ILC2s start producing IL-5, IL-13, and amphiregulin, which in turn recruit and stimulate eosinophils to release profibrotic cytokines such as TGF-β, PDGF, and IL-13, promoting the fibroblast-to-myofibroblast transition [54].

2.3. Pathomorphological Changes in the Lungs during Asthma Development

Pathomorphological changes in the bronchial asthma lungs can be divided into two patterns: alterations in bronchial epithelium and smooth muscles, prominent signs of asthma exacerbations, and subepithelial fibrosis, a characteristic of long-term asthma [55]. All these pathological changes lead to bronchial obstruction, which is reversible at the early stages of the disease and irreversible at the later ones.
During acute asthma development, hyperplasia and metaplasia of the goblet and epithelial cells of the bronchial epithelium, leading to mucus hyperproduction, thickening of the airways, and bronchial obstruction, are observed [56]. Moreover, in severe asthma exacerbations, large and small airways are often obstructed by mucus plugs with an admixture of inflammatory cells (mostly eosinophils in the case of allergic asthma) [57]. An additional factor leading to the formation of mucus plugs is the dysfunction of ciliated cells due to airway inflammation, characterized by a decrease in the frequency of its fluctuations as well as dyskinesia and disorientation of the cilia [58].
Spasm of the bronchial smooth musculature—bronchoconstriction—is another factor leading to airway obstruction. Under physiological conditions, bronchial smooth muscles provide mechanical stability to the airways without cartilage. However, hyperreactivity of asthmatic airways decreases smooth muscle sensitivity threshold, following spasm and reversible airway obstruction [59]. The accumulation of smooth muscle cells due to their hypertrophy and hyperplasia is another component of asthma pathomorphological changes, leading to airway thickening [60]. Moreover, it is believed that smooth muscle cells may support airway remodeling through the secretion of pro-inflammatory mediators, matrix and cell adhesion proteins, and other stimulatory molecules, affecting the further migration and activity of inflammatory cells [61].
The major characteristic of chronic asthmatic inflammation is subepithelial airway and, in some cases, lung fibrosis, consisting of connective tissue growth in the basal membrane and submucosal area. However, changes in the airways, leading to lung fibrosis, are present even in the earliest stages of asthma [55][62].

References

  1. Alvarez, C.A.; Qian, E.; Glendenning, L.M.; Reynero, K.M.; Kukan, E.N.; Cobb, B.A. Acute and chronic lung inflammation drives changes in epithelial glycans. Front. Immunol. 2023, 14, 1167908.
  2. Salameh, L.; Mahmood, W.; Hamoudi, R.; Almazrouei, K.; Lochanan, M.; Seyhoglu, S.; Mahboub, B. The Role of Vitamin D Supplementation on Airway Remodeling in Asthma: A Systematic Review. Nutrients 2023, 15, 2477.
  3. Cha, J.; Choi, S. Gene-Smoking Interaction Analysis for the Identification of Novel Asthma-Associated Genetic Factors. Int. J. Mol. Sci. 2023, 24, 12266.
  4. Colombo, S.A.P.; Brown, S.L.; Hepworth, M.R.; Hankinson, J.; Granato, F.; Kitchen, S.J.; Hussell, T.; Simpson, A.; Cook, P.C.; MacDonald, A.S. Comparative phenotype of circulating versus tissue immune cells in human lung and blood compartments during health and disease. Discov. Immunol. 2023, 2, kyad009.
  5. Wieczfinska, J.; Pawliczak, R. Anti-fibrotic effect of ciglitazone in HRV-induced airway remodelling cell model. J. Cell. Mol. Med. 2023, 27, 1867–1879.
  6. Rimkunas, A.; Januskevicius, A.; Vasyle, E.; Palacionyte, J.; Janulaityte, I.; Miliauskas, S.; Malakauskas, K. Blood Inflammatory-like and Lung Resident-like Eosinophils Affect Migration of Airway Smooth Muscle Cells and Their ECM-Related Proliferation in Asthma. Int. J. Mol. Sci. 2023, 24, 3469.
  7. Abohalaka, R. Bronchial Epithelial and Airway Smooth Muscle Cell Interactions in Health and Disease. Heliyon 2023, 9, e19976.
  8. Paw, M.; Wnuk, D.; Madeja, Z.; Michalik, M. PPARδ Agonist GW501516 Suppresses the TGF-β-Induced Profibrotic Response of Human Bronchial Fibroblasts from Asthmatic Patients. Int. J. Mol. Sci. 2023, 24, 7721.
  9. Mauad, T.; Silva, L.F.F.; Santos, M.A.; Grinberg, L.; Bernardi, F.D.C.; Martins, M.A.; Saldiva, P.H.N.; Dolhnikoff, M. Abnormal alveolar attachments with decreased elastic fiber content in distal lung in fatal asthma. Am. J. Respir. Crit. Care Med. 2004, 170, 857–862.
  10. Pechkovsky, D.V.; Hackett, T.L.; An, S.S.; Shaheen, F.; Murray, L.A.; Knight, D.A. Human lung parenchyma but not proximal bronchi produces fibroblasts with enhanced TGF-β signaling and α-SMA expression. Am. J. Respir. Cell Mol. Biol. 2010, 43, 641–651.
  11. Tsukui, T.; Sun, K.H.; Wetter, J.B.; Wilson-Kanamori, J.R.; Hazelwood, L.A.; Henderson, N.C.; Adams, T.S.; Schupp, J.C.; Poli, S.D.; Rosas, I.O.; et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat. Commun. 2020, 11, 1920.
  12. Pascual, R.M.; Peters, S.P. The irreversible component of persistent asthma. J. Allergy Clin. Immunol. 2009, 124, 883–890.
  13. Perez-Garcia, J.; Espuela-Ortiz, A.; Lorenzo-Diaz, F.; Pino-Yanes, M. Pharmacogenetics of Pediatric Asthma: Current Perspectives. Pharmgenom. Pers. Med. 2020, 13, 89.
  14. Sethi, G.S.; Naura, A.S. Progressive increase in allergen concentration abrogates immune tolerance in ovalbumin-induced murine model of chronic asthma. Int. Immunopharmacol. 2018, 60, 121–131.
  15. Abbafati, C.; Abbas, K.M.; Abbasi-Kangevari, M.; Abd-Allah, F.; Abdelalim, A.; Abdollahi, M.; Abdollahpour, I.; Abegaz, K.H.; Abolhassani, H.; Aboyans, V.; et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222.
  16. Stern, J.; Pier, J.; Litonjua, A.A. Asthma epidemiology and risk factors. Semin. Immunopathol. 2020, 42, 5–15.
  17. Dharmage, S.C.; Perret, J.L.; Custovic, A. Epidemiology of asthma in children and adults. Front. Pediatr. 2019, 7, 246.
  18. Agache, I.; Akdis, C.A. Endotypes of allergic diseases and asthma: An important step in building blocks for the future of precision medicine. Allergol. Int. 2016, 65, 243–252.
  19. Wenzel, S.E. Asthma phenotypes: The evolution from clinical to molecular approaches. Nat. Med. 2012, 18, 716–725.
  20. Borish, L. The immunology of asthma: Asthma phenotypes and their implications for personalized treatment. Ann. Allergy Asthma Immunol. 2016, 117, 108–114.
  21. McIntyre, A.P.; Viswanathan, R.K. Phenotypes and Endotypes in Asthma. Adv. Exp. Med. Biol. 2023, 1426, 119–142.
  22. Moore, W.C.; Meyers, D.A.; Wenzel, S.E.; Teague, W.G.; Li, H.; Li, X.; D’Agostino, R.; Castro, M.; Curran-Everett, D.; Fitzpatrick, A.M.; et al. Identification of asthma phenotypes using cluster analysis in the severe asthma research program. Am. J. Respir. Crit. Care Med. 2010, 181, 315–323.
  23. Hastie, A.T.; Moore, W.C.; Meyers, D.A.; Vestal, P.L.; Li, H.; Peters, S.P.; Bleecker, E.R. Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes. J. Allergy Clin. Immunol. 2010, 125, 1028–1036.e13.
  24. Buchheit, K.M.; Cahill, K.N.; Katz, H.R.; Murphy, K.C.; Feng, C.; Lee-Sarwar, K.; Lai, J.; Bhattacharyya, N.; Israel, E.; Boyce, J.A.; et al. Thymic stromal lymphopoietin controls prostaglandin D2 generation in patients with aspirin-exacerbated respiratory disease. J. Allergy Clin. Immunol. 2016, 137, 1566–1576.e5.
  25. Carr, T.F.; Kraft, M. Chronic Infection and Severe Asthma. Immunol. Allergy Clin. 2016, 36, 483–502.
  26. Fitzpatrick, A.M.; Chipps, B.E.; Holguin, F.; Woodruff, P.G. T2-“Low” Asthma: Overview and Management Strategies. J. Allergy Clin. Immunol. Pract. 2020, 8, 452–463.
  27. Mims, J.W. Asthma: Definitions and pathophysiology. Int. Forum Allergy Rhinol. 2015, 5, S2–S6.
  28. Holloway, J.W.; Yang, I.A.; Holgate, S.T. Genetics of allergic disease. J. Allergy Clin. Immunol. 2010, 125, S81–S94.
  29. Bunyavanich, S.; Schadt, E.E. Systems biology of asthma and allergic diseases: A multiscale approach. J. Allergy Clin. Immunol. 2015, 135, 31–42.
  30. Tohidinik, H.R.; Mallah, N.; Takkouche, B. History of allergic rhinitis and risk of asthma; a systematic review and meta-analysis. World Allergy Organ. J. 2019, 12, 100069.
  31. Turek, E.M.; Cox, M.J.; Hunter, M.; Hui, J.; James, P.; Willis-Owen, S.A.G.; Cuthbertson, L.; James, A.; Musk, A.W.; Moffatt, M.F.; et al. Airway microbial communities, smoking and asthma in a general population sample. EBioMedicine 2021, 71, 103538.
  32. Pfeffer, P.E.; Mudway, I.S.; Grigg, J. Air Pollution and Asthma: Mechanisms of Harm and Considerations for Clinical Interventions. Chest 2021, 159, 1346–1355.
  33. Miethe, S.; Karsonova, A.; Karaulov, A.; Renz, H. Obesity and asthma. J. Allergy Clin. Immunol. 2020, 146, 685–693.
  34. Tan, J.; Bernstein, J.A. Occupational asthma: An overview topical collection on occupational allergies. Curr. Allergy Asthma Rep. 2014, 14, 431.
  35. Malo, J.L.; Vandenplas, O. Definitions and Classification of Work-Related Asthma. Immunol. Allergy Clin. 2011, 31, 645–662.
  36. Holgate, S.T.; Wenzel, S.; Postma, D.S.; Weiss, S.T.; Renz, H.; Sly, P.D. Asthma. Nat. Rev. Dis. Prim. 2015, 1, 15025.
  37. Valero, A.; Quirce, S.; Dávila, I.; Delgado, J.; Domínguez-Ortega, J. Allergic respiratory disease: Different allergens, different symptoms. Allergy 2017, 72, 1306–1316.
  38. Wu, L.C.; Zarrin, A.A. The production and regulation of IgE by the immune system. Nat. Rev. Immunol. 2014, 14, 247–259.
  39. Sallmann, E.; Reininger, B.; Brandt, S.; Duschek, N.; Hoflehner, E.; Garner-Spitzer, E.; Platzer, B.; Dehlink, E.; Hammer, M.; Holcmann, M.; et al. High-Affinity IgE Receptors on Dendritic Cells Exacerbate Th2-Dependent Inflammation. J. Immunol. 2011, 187, 164–171.
  40. Bax, H.J.; Keeble, A.H.; Gould, H.J. Cytokinergic IgE action in mast cell activation. Front. Immunol. 2012, 3, 229.
  41. Lambrecht, B.N.; Hammad, H. The airway epithelium in asthma. Nat. Med. 2012, 18, 684–692.
  42. Johansson, M.W. Activation states of blood eosinophils in asthma. Clin. Exp. Allergy 2014, 44, 482–498.
  43. Broide, D. New perspectives on mechanisms underlying chronic allergic inflammation and asthma in 2007. J. Allergy Clin. Immunol. 2008, 122, 475–480.
  44. Holt, P.G.; Strickland, D.H.; Bosco, A.; Jahnsen, F.L. Pathogenic mechanisms of allergic inflammation: Atopic asthma as a paradigm. Adv. Immunol. 2009, 104, 51–113.
  45. Holtzman, M.J. Asthma as a chronic disease of the innate and adaptive immune systems responding to viruses and allergens. J. Clin. Investig. 2012, 122, 2741–2748.
  46. McGee, H.S.; Agrawal, D.K. Naturally Occurring and Inducible T-Regulatory Cells Modulating Immune Response in Allergic Asthma. Am. J. Respir. Crit. Care Med. 2012, 180, 211–225.
  47. Peters, S.P. Asthma Phenotypes: Nonallergic (Intrinsic) Asthma. J. Allergy Clin. Immunol. Pract. 2014, 2, 650–652.
  48. Matucci, A.; Vultaggio, A.; Maggi, E.; Kasujee, I. Is IgE or eosinophils the key player in allergic asthma pathogenesis? Are we asking the right question? Respir. Res. 2018, 19, 113.
  49. Green, B.J.; Wiriyachaiporn, S.; Grainge, C.; Rogers, G.B.; Kehagia, V.; Lau, R.; Carroll, M.P.; Bruce, K.D.; Howarth, P.H. Potentially pathogenic airway bacteria and neutrophilic inflammation in treatment resistant severe asthma. PLoS ONE 2014, 9, e100645.
  50. Peters, M.C.; Mekonnen, Z.K.; Yuan, S.; Bhakta, N.R.; Woodruff, P.G.; Fahy, J.V. Measures of gene expression in sputum cells can identify TH2-high and TH2-low subtypes of asthma. J. Allergy Clin. Immunol. 2014, 133, 388–394.e5.
  51. Uddin, M.; Nong, G.; Ward, J.; Seumois, G.; Prince, L.R.; Wilson, S.J.; Cornelius, V.; Dent, G.; Djukanović, R. Prosurvival activity for airway neutrophils in severe asthma. Thorax 2010, 65, 684–689.
  52. Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Rard Eberl, G.; Koyasu, S.; Locksley, R.M.; Mckenzie, A.N.J.; Mebius, R.E.; et al. Leading Edge Review Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066.
  53. Wirtz, S.; Schulz-Kuhnt, A.; Neurath, M.F.; Atreya, I. Functional Contribution and Targeted Migration of Group-2 Innate Lymphoid Cells in Inflammatory Lung Diseases: Being at the Right Place at the Right Time. Front. Immunol. 2021, 12, 688879.
  54. Mikami, Y.; Takada, Y.; Hagihara, Y.; Kanai, T. Innate lymphoid cells in organ fibrosis. Cytokine Growth Factor Rev. 2018, 42, 27–36.
  55. Bittar, H.E.T.; Yousem, S.A.; Wenzel, S.E. Pathobiology of severe asthma. Annu. Rev. Pathol. Mech. Dis. 2015, 10, 511–545.
  56. Gordon, I.O.; Husain, A.N.; Charbeneau, J.; Krishnan, J.A.; Hogarth, D.K. Endobronchial biopsy: A guide for asthma therapy selection in the era of bronchial thermoplasty. J. Asthma 2013, 50, 634–641.
  57. Dunican, E.M.; Watchorn, D.C.; Fahy, J.V. Autopsy and imaging studies of mucus in asthma: Lessons learned about disease mechanisms and the role of mucus in airflow obstruction. Ann. Am. Thorac. Soc. 2018, 15, S184–S191.
  58. Thomas, B.; Rutman, A.; Hirst, R.A.; Haldar, P.; Wardlaw, A.J.; Bankart, J.; Brightling, C.E.; O’Callaghan, C. Ciliary dysfunction and ultrastructural abnormalities are features of severe asthma. J. Allergy Clin. Immunol. 2010, 126, 722–729.e2.
  59. Lam, M.; Lamanna, E.; Bourke, J.E. Regulation of Airway Smooth Muscle Contraction in Health and Disease. In Advances in Experimental Medicine and Biology; Springer New York LLC: New York, NY, USA, 2019; Volume 1124, pp. 381–422.
  60. James, A.L.; Noble, P.B.; Drew, S.A.; Mauad, T.; Bai, T.R.; Abramson, M.J.; McKay, K.O.; Green, F.H.Y.; Elliot, J.G. Airway smooth muscle proliferation and inflammation in asthma. J. Appl. Physiol. 2018, 125, 1090–1096.
  61. Liu, Y.; Li, X.; He, C.; Chen, R.; Wei, L.; Meng, L.; Zhang, C. Emodin ameliorates ovalbumin-induced airway remodeling in mice by suppressing airway smooth muscle cells proliferation. Int. Immunopharmacol. 2020, 88, 106855.
  62. Elliot, J.G.; Noble, P.B.; Mauad, T.; Bai, T.R.; Abramson, M.J.; McKay, K.O.; Green, F.H.Y.; James, A.L. Inflammation-dependent and independent airway remodelling in asthma. Respirology 2018, 23, 1138–1145.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 138
Revisions: 2 times (View History)
Update Date: 18 Dec 2023
1000/1000
Video Production Service