Allergy Asthma Immunol Res.  2018 Mar;10(2):106-120. 10.4168/aair.2018.10.2.106.

Can Controlling Endoplasmic Reticulum Dysfunction Treat Allergic Inflammation in Severe Asthma With Fungal Sensitization?

Affiliations
  • 1Department of Internal Medicine, Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, Korea. leeyc@jbnu.ac.kr
  • 2Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Medical School, Jeonju, Korea.

Abstract

Severe asthma is a heterogeneous disease entity to which diverse cellular components and pathogenetic mechanisms contribute. Current asthma therapies, including new biologic agents, are mainly targeting T helper type 2 cell-dominant inflammation, so that they are often unsatisfactory in the treatment of severe asthma. Respiratory fungal exposure has long been regarded as a precipitating factor for severe asthma phenotype. Moreover, as seen in clinical definitions of allergic bronchopulmonary aspergillosis (ABPA) and severe asthma with fungal sensitization (SAFS), fungal allergy-associated severe asthma phenotype is increasingly thought to have distinct pathobiologic mechanisms requiring different therapeutic approaches other than conventional treatment. However, there are still many unanswered questions on the direct causality of fungal sensitization in inducing severe allergic inflammation in SAFS. Recently, growing evidence suggests that stress response from the largest organelle, endoplasmic reticulum (ER), is closely interconnected to diverse cellular immune/inflammatory platforms, thereby being implicated in severe allergic lung inflammation. Interestingly, a recent study on this issue has suggested that ER stress responses and several associated molecular platforms, including phosphoinositide 3-kinase-δ and mitochondria, may be crucial players in the development of severe allergic inflammation in the SAFS. Defining emerging roles of ER and associated cellular platforms in SAFS may offer promising therapeutic options in the near future.

Keyword

Severe asthma; fungi; SAFS; ER stress; subcellular organelles; innate immunity

MeSH Terms

Aspergillosis, Allergic Bronchopulmonary
Asthma*
Biological Factors
Endoplasmic Reticulum*
Fungi
Immunity, Innate
Inflammation*
Mitochondria
Organelles
Phenotype
Pneumonia
Precipitating Factors
Biological Factors

Figure

  • Fig. 1 Fungal exposure leads to the activation of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) in the lung. Inhaled fungi possess a wide array of ligands that activate pattern recognition receptors (PRRs) expressed on structural cells (e.g. airway epithelial cells) and dendritic cells (DCs). Fungi can also produce large amounts of secreted enzymes, such as proteases, which disrupt tight junctions of airway epithelium. Initial recognition of fungi is followed by allergic sensitization and eosinophilic airway inflammation through close interactions between various facets of host immunity (not presented here). During this process, various cell types, including frontline cells (e.g. airway epithelial cells, DCs, and alveolar macrophages) and adaptive T and B cells, produce large amounts of cytokines/chemokines as well as host defensive molecules. Increased protein folding demand in these cells results in ER stress and triggers UPR. UPR signaling is orchestrated by 3 ER-localized sensors, namely, PERK, IRE1, and ATF6. These adaptive responses together reduce protein folding demand, increase enzymes and chaperones involved in protein folding, and facilitate protein degradation pathway. However, when cells fail to resolve ER stress, UPR mediates ER stress-induced apoptosis.

  • Fig. 2 Cytoplasmic interactions involving endoplasmic reticulum (ER), mitochondria, and NLRP3 inflammasome may contribute to fungi-induced severe eosinophilic allergic inflammation in the lung. Initial fungal recognition activates the cell membrane-associated phosphoinositide 3-kinase delta (PI3K-δ) signaling pathway in various structural cells and immune cells. This process is followed by the downstream cascade of protein interactions and phosphorylation, leading to diverse biological consequences in these cells. Particularly, PI3K-δ modulates fungi-induced ER stress and UPR activation, especially in airway epithelial cells. Furthermore, mitochondrial generation of reactive oxygen species (mtROS) and NLRP3 inflammasome activation in response to respiratory fungal exposure are closely related to this process. The net result of these associations may play a key role in the pathogenesis of fungi-induced severe eosinophilic allergic inflammation in the lung.


Reference

1. Gauthier M, Ray A, Wenzel SE. Evolving concepts of asthma. Am J Respir Crit Care Med. 2015; 192:660–668.
Article
2. Barnes PJ. Severe asthma: advances in current management and future therapy. J Allergy Clin Immunol. 2012; 129:48–59.
Article
3. Moore WC, Fitzpatrick AM, Li X, Hastie AT, Li H, Meyers DA, et al. Clinical heterogeneity in the severe asthma research program. Ann Am Thorac Soc. 2013; 10:S118–S124.
Article
4. Lötvall J, Akdis CA, Bacharier LB, Bjermer L, Casale TB, Custovic A, et al. Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol. 2011; 127:355–360.
5. Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014; 43:343–373.
6. Twaroch TE, Curin M, Valenta R, Swoboda I. Mold allergens in respiratory allergy: from structure to therapy. Allergy Asthma Immunol Res. 2015; 7:205–220.
Article
7. Denning DW, O’Driscoll BR, Hogaboam CM, Bowyer P, Niven RM. The link between fungi and severe asthma: a summary of the evidence. Eur Respir J. 2006; 27:615–626.
Article
8. Knutsen AP, Bush RK, Demain JG, Denning DW, Dixit A, Fairs A, et al. Fungi and allergic lower respiratory tract diseases. J Allergy Clin Immunol. 2012; 129:280–291.
Article
9. Harley KG, Macher JM, Lipsett M, Duramad P, Holland NT, Prager SS, et al. Fungi and pollen exposure in the first months of life and risk of early childhood wheezing. Thorax. 2009; 64:353–358.
Article
10. Delfino RJ, Zeiger RS, Seltzer JM, Street DH, Matteucci RM, Anderson PR, et al. The effect of outdoor fungal spore concentrations on daily asthma severity. Environ Health Perspect. 1997; 105:622–635.
Article
11. Lin WR, Chen YH, Lee MF, Hsu LY, Tien CJ, Shih FM, et al. Does spore count matter in fungal allergy?: the role of allergenic fungal species. Allergy Asthma Immunol Res. 2016; 8:404–411.
Article
12. Agarwal R. Severe asthma with fungal sensitization. Curr Allergy Asthma Rep. 2011; 11:403–413.
Article
13. Millien VO, Lu W, Shaw J, Yuan X, Mak G, Roberts L, et al. Cleavage of fibrinogen by proteinases elicits allergic responses through Toll-like receptor 4. Science. 2013; 341:792–796.
Article
14. Zureik M, Neukirch C, Leynaert B, Liard R, Bousquet J, Neukirch F, et al. Sensitisation to airborne moulds and severity of asthma: cross sectional study from European Community respiratory health survey. BMJ. 2002; 325:411–414.
Article
15. O'Driscoll BR, Hopkinson LC, Denning DW. Mold sensitization is common amongst patients with severe asthma requiring multiple hospital admissions. BMC Pulm Med. 2005; 5:4.
16. Denning DW, O’Driscoll BR, Powell G, Chew F, Atherton GT, Vyas A, et al. 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. 2009; 179:11–18.
17. Agbetile J, Bourne M, Fairs A, Hargadon B, Desai D, Broad C, et al. Effectiveness of voriconazole in the treatment of Aspergillus fumigatus-associated asthma (EVITA3 study). J Allergy Clin Immunol. 2014; 134:33–39.
Article
18. Arensdorf AM, Diedrichs D, Rutkowski DT. Regulation of the transcriptome by ER stress: non-canonical mechanisms and physiological consequences. Front Genet. 2013; 4:256.
Article
19. Hosoi T, Ozawa K. Endoplasmic reticulum stress in disease: mechanisms and therapeutic opportunities. Clin Sci (Lond). 2009; 118:19–29.
Article
20. Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature. 2008; 454:455–462.
Article
21. Kim SR, Kim DI, Kang MR, Lee KS, Park SY, Jeong JS, et al. Endoplasmic reticulum stress influences bronchial asthma pathogenesis by modulating nuclear factor κB activation. J Allergy Clin Immunol. 2013; 132:1397–1408.
Article
22. Osorio F, Lambrecht B, Janssens S. The UPR and lung disease. Semin Immunopathol. 2013; 35:293–306.
Article
23. Ribeiro CM, O'Neal WK. Endoplasmic reticulum stress in chronic obstructive lung diseases. Curr Mol Med. 2012; 12:872–882.
24. Lawson WE, Cheng DS, Degryse AL, Tanjore H, Polosukhin VV, Xu XC, et al. Endoplasmic reticulum stress enhances fibrotic remodeling in the lungs. Proc Natl Acad Sci U S A. 2011; 108:10562–10567.
Article
25. Kim HJ, Jeong JS, Kim SR, Park SY, Chae HJ, Lee YC. Inhibition of endoplasmic reticulum stress alleviates lipopolysaccharide-induced lung inflammation through modulation of NF-κB/HIF-1α signaling pathway. Sci Rep. 2013; 3:1142.
Article
26. Denning DW, Pashley C, Hartl D, Wardlaw A, Godet C, Del Giacco S, et al. Fungal allergy in asthma-state of the art and research needs. Clin Transl Allergy. 2014; 4:14.
Article
27. Agarwal R. Allergic bronchopulmonary aspergillosis. Chest. 2009; 135:805–826.
Article
28. Chishimba L, Niven RM, Cooley J, Denning DW. Voriconazole and posaconazole improve asthma severity in allergic bronchopulmonary aspergillosis and severe asthma with fungal sensitization. J Asthma. 2012; 49:423–433.
Article
29. Hardison SE, Brown GD. C-type lectin receptors orchestrate antifungal immunity. Nat Immunol. 2012; 13:817–822.
Article
30. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140:805–820.
Article
31. Iossifova Y, Reponen T, Sucharew H, Succop P, Vesper S. Use of (1–3)-beta-d-glucan concentrations in dust as a surrogate method for estimating specific fungal exposures. Indoor Air. 2008; 18:225–232.
32. Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med. 2009; 361:1760–1767.
Article
33. Glocker EO, Hennigs A, Nabavi M, Schäffer AA, Woellner C, Salzer U, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med. 2009; 361:1727–1735.
34. Fogelmark B, Thorn J, Rylander R. Inhalation of (1→3)-beta-Dglucan causes airway eosinophilia. Mediators Inflamm. 2001; 10:13–19.
35. Douwes J, Zuidhof A, Doekes G, van der Zee SC, Wouters I, Boezen MH, et al. (1→3)-beta-D-glucan and endotoxin in house dust and peak flow variability in children. Am J Respir Crit Care Med. 2000; 162:1348–1354.
36. Bourgeois C, Kuchler K. Fungal pathogens-a sweet and sour treat for Toll-like receptors. Front Cell Infect Microbiol. 2012; 2:142.
Article
37. Meier A, Kirschning CJ, Nikolaus T, Wagner H, Heesemann J, Ebel F. Toll-like receptor (TLR) 2 and TLR4 are essential for Aspergillus-induced activation of murine macrophages. Cell Microbiol. 2003; 5:561–570.
Article
38. Tessarolli V, Gasparoto TH, Lima HR, Figueira EA, Garlet TP, Torres SA, et al. Absence of TLR2 influences survival of neutrophils after infection with Candida albicans. Med Mycol. 2010; 48:129–140.
Article
39. Gasparoto TH, Tessarolli V, Garlet TP, Torres SA, Garlet GP, da Silva JS, et al. Absence of functional TLR4 impairs response of macrophages after Candida albicans infection. Med Mycol. 2010; 48:1009–1017.
40. Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, et al. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature. 2009; 459:433–436.
Article
41. Franchi L, Muñoz-Planillo R, Núñez G. Sensing and reacting to microbes through the inflammasomes. Nat Immunol. 2012; 13:325–332.
Article
42. Knight DA, Lim S, Scaffidi AK, Roche N, Chung KF, Stewart GA, et al. Protease-activated receptors in human airways: upregulation of PAR-2 in respiratory epithelium from patients with asthma. J Allergy Clin Immunol. 2001; 108:797–803.
Article
43. Homma T, Kato A, Bhushan B, Norton JE, Suh LA, Carter RG, et al. Role of Aspergillus fumigatus in triggering protease-activated receptor-2 in airway epithelial cells and skewing the cells toward a T-helper 2 bias. Am J Respir Cell Mol Biol. 2016; 54:60–70.
44. Kheradmand F, Kiss A, Xu J, Lee SH, Kolattukudy PE, Corry DB. A protease-activated pathway underlying Th cell type 2 activation and allergic lung disease. J Immunol. 2002; 169:5904–5911.
Article
45. Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med. 2012; 18:684–692.
Article
46. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med. 2009; 15:410–416.
Article
47. Bartemes KR, Iijima K, Kobayashi T, Kephart GM, McKenzie AN, Kita H. IL-33-responsive lineage- CD25+ CD44(hi) lymphoid cells mediate innate type 2 immunity and allergic inflammation in the lungs. J Immunol. 2012; 188:1503–1513.
48. Van Dyken SJ, Mohapatra A, Nussbaum JC, Molofsky AB, Thornton EE, Ziegler SF, et al. Chitin activates parallel immune modules that direct distinct inflammatory responses via innate lymphoid type 2 and γδ T cells. Immunity. 2014; 40:414–424.
Article
49. Halim TY, Steer CA, Mathä L, Gold MJ, Martinez-Gonzalez I, McNagny KM, et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity. 2014; 40:425–435.
Article
50. Snelgrove RJ, Gregory LG, Peiró T, Akthar S, Campbell GA, Walker SA, et al. Alternaria-derived serine protease activity drives IL-33-mediated asthma exacerbations. J Allergy Clin Immunol. 2014; 134:583–592.e6.
Article
51. Saglani S, Lui S, Ullmann N, Campbell GA, Sherburn RT, Mathie SA, et al. IL-33 promotes airway remodeling in pediatric patients with severe steroid-resistant asthma. J Allergy Clin Immunol. 2013; 132:676–685.e13.
Article
52. Castanhinha S, Sherburn R, Walker S, Gupta A, Bossley CJ, Buckley J, et al. Pediatric severe asthma with fungal sensitization is mediated by steroid-resistant IL-33. J Allergy Clin Immunol. 2015; 136:312–322.e7.
Article
53. Underhill DM, Pearlman E. Immune interactions with pathogenic and commensal fungi: a two-way street. Immunity. 2015; 43:845–858.
Article
54. Knutsen AP, Vijay HM, Kumar V, Kariuki B, Santiago LA, Graff R, et al. Mold-sensitivity in children with moderate-severe asthma is associated with HLA-DR and HLA-DQ. Allergy. 2010; 65:1367–1375.
Article
55. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010; 363:1211–1221.
Article
56. Vannuvel K, Renard P, Raes M, Arnould T. Functional and morphological impact of ER stress on mitochondria. J Cell Physiol. 2013; 228:1802–1818.
Article
57. Gardner BM, Walter P. Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science. 2011; 333:1891–1894.
Article
58. Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012; 13:89–102.
Article
59. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003; 11:619–633.
Article
60. Wu J, Rutkowski DT, Dubois M, Swathirajan J, Saunders T, Wang J, et al. ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev Cell. 2007; 13:351–364.
61. Yamamoto K, Sato T, Matsui T, Sato M, Okada T, Yoshida H, et al. Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev Cell. 2007; 13:365–376.
62. Lee AH, Scapa EF, Cohen DE, Glimcher LH. Regulation of hepatic lipogenesis by the transcription factor XBP1. Science. 2008; 320:1492–1496.
Article
63. Martinon F, Chen X, Lee AH, Glimcher LH. TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol. 2010; 11:411–418.
Article
64. Acosta-Alvear D, Zhou Y, Blais A, Tsikitis M, Lents NH, Arias C, et al. XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks. Mol Cell. 2007; 27:53–66.
Article
65. Reimold AM, Etkin A, Clauss I, Perkins A, Friend DS, Zhang J, et al. An essential role in liver development for transcription factor XBP-1. Genes Dev. 2000; 14:152–157.
66. Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, Szomolanyi-Tsuda E, Gravallese EM, et al. Plasma cell differentiation requires the transcription factor XBP-1. Nature. 2001; 412:300–307.
Article
67. Zhang K, Wong HN, Song B, Miller CN, Scheuner D, Kaufman RJ. The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis. J Clin Invest. 2005; 115:268–281.
68. Hollien J, Weissman JS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science. 2006; 313:104–107.
Article
69. Maurel M, Chevet E, Tavernier J, Gerlo S. Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem Sci. 2014; 39:245–254.
Article
70. Ventoso I, Kochetov A, Montaner D, Dopazo J, Santoyo J. Extensive translatome remodeling during ER stress response in mammalian cells. PLoS One. 2012; 7:e35915.
Article
71. Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009; 1:a001651.
72. Deng J, Lu PD, Zhang Y, Scheuner D, Kaufman RJ, Sonenberg N, et al. Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol Cell Biol. 2004; 24:10161–10168.
Article
73. Yaman I, Fernandez J, Liu H, Caprara M, Komar AA, Koromilas AE, et al. The zipper model of translational control: a small upstream ORF is the switch that controls structural remodeling of an mRNA leader. Cell. 2003; 113:519–531.
74. Zhou D, Palam LR, Jiang L, Narasimhan J, Staschke KA, Wek RC. Phosphorylation of eIF2 directs ATF5 translational control in response to diverse stress conditions. J Biol Chem. 2008; 283:7064–7073.
Article
75. Korennykh AV, Egea PF, Korostelev AA, Finer-Moore J, Zhang C, Shokat KM, et al. The unfolded protein response signals through high-order assembly of Ire1. Nature. 2009; 457:687–693.
Article
76. Li H, Korennykh AV, Behrman SL, Walter P. Mammalian endoplasmic reticulum stress sensor IRE1 signals by dynamic clustering. Proc Natl Acad Sci U S A. 2010; 107:16113–16118.
Article
77. Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell. 2000; 103:239–252.
Article
78. Hu P, Han Z, Couvillon AD, Kaufman RJ, Exton JH. Autocrine tumor necrosis factor alpha links endoplasmic reticulum stress to the membrane death receptor pathway through IRE1alpha-mediated NF-kappaB activation and down-regulation of TRAF2 expression. Mol Cell Biol. 2006; 26:3071–3084.
79. Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000; 287:664–666.
Article
80. Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ, Diehl JA. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol Cell Biol. 2003; 23:7198–7209.
Article
81. Vinson C, Acharya A, Taparowsky EJ. Deciphering B-ZIP transcription factor interactions in vitro and in vivo. Biochim Biophys Acta. 2006; 1759:4–12.
Article
82. Ramji DP, Foka P. CCAAT/enhancer-binding proteins: structure, function and regulation. Biochem J. 2002; 365:561–575.
Article
83. Wang Y, Vera L, Fischer WH, Montminy M. The CREB coactivator CRTC2 links hepatic ER stress and fasting gluconeogenesis. Nature. 2009; 460:534–537.
Article
84. Valencia-Sanchez MA, Liu J, Hannon GJ, Parker R. Control of translation and mRNA degradation by miRNAs and siRNAs. Genes Dev. 2006; 20:515–524.
Article
85. Martino ME, Olsen JC, Fulcher NB, Wolfgang MC, O’Neal WK, Ribeiro CM. Airway epithelial inflammation-induced endoplasmic reticulum Ca2+ store expansion is mediated by X-box binding protein-1. J Biol Chem. 2009; 284:14904–14913.
Article
86. Bettigole SE, Glimcher LH. Endoplasmic reticulum stress in immunity. Annu Rev Immunol. 2015; 33:107–138.
Article
87. Laing S, Wang G, Briazova T, Zhang C, Wang A, Zheng Z, et al. Airborne particulate matter selectively activates endoplasmic reticulum stress response in the lung and liver tissues. Am J Physiol Cell Physiol. 2010; 299:C736–C749.
Article
88. Jorgensen E, Stinson A, Shan L, Yang J, Gietl D, Albino AP. Cigarette smoke induces endoplasmic reticulum stress and the unfolded protein response in normal and malignant human lung cells. BMC Cancer. 2008; 8:229.
Article
89. Hengstermann A, Müller T. Endoplasmic reticulum stress induced by aqueous extracts of cigarette smoke in 3T3 cells activates the unfolded-protein-response-dependent PERK pathway of cell survival. Free Radic Biol Med. 2008; 44:1097–1107.
Article
90. Wang S, Kaufman RJ. The impact of the unfolded protein response on human disease. J Cell Biol. 2012; 197:857–867.
Article
91. Voeltz GK, Prinz WA, Shibata Y, Rist JM, Rapoport TA. A class of membrane proteins shaping the tubular endoplasmic reticulum. Cell. 2006; 124:573–586.
Article
92. Wright PL, Yu J, Di YP, Homer RJ, Chupp G, Elias JA, et al. Epithelial reticulon 4B (Nogo-B) is an endogenous regulator of Th2-driven lung inflammation. J Exp Med. 2010; 207:2595–2607.
Article
93. Schroeder BW, Verhaeghe C, Park SW, Nguyenvu LT, Huang X, Zhen G, et al. AGR2 is induced in asthma and promotes allergen-induced mucin overproduction. Am J Respir Cell Mol Biol. 2012; 47:178–185.
Article
94. Martino MB, Jones L, Brighton B, Ehre C, Abdulah L, Davis CW, et al. The ER stress transducer IRE1β is required for airway epithelial mucin production. Mucosal Immunol. 2013; 6:639–654.
Article
95. Breslow DK, Collins SR, Bodenmiller B, Aebersold R, Simons K, Shevchenko A, et al. Orm family proteins mediate sphingolipid homeostasis. Nature. 2010; 463:1048–1053.
Article
96. Han S, Lone MA, Schneiter R, Chang A. Orm1 and Orm2 are conserved endoplasmic reticulum membrane proteins regulating lipid homeostasis and protein quality control. Proc Natl Acad Sci U S A. 2010; 107:5851–5856.
Article
97. Miller M, Tam AB, Cho JY, Doherty TA, Pham A, Khorram N, et al. ORMDL3 is an inducible lung epithelial gene regulating metalloproteases, chemokines, OAS, and ATF6. Proc Natl Acad Sci U S A. 2012; 109:16648–16653.
Article
98. Kim SR, Lee YC. Endoplasmic reticulum stress and the related signaling networks in severe asthma. Allergy Asthma Immunol Res. 2015; 7:106–117.
Article
99. Hoffman SM, Chapman DG, Lahue KG, Cahoon JM, Rattu GK, Daphtary N, et al. Protein disulfide isomerase-endoplasmic reticulum resident protein 57 regulates allergen-induced airways inflammation, fibrosis, and hyperresponsiveness. J Allergy Clin Immunol. 2016; 137:822–832.e7.
Article
100. Lee KS, Jeong JS, Kim SR, Cho SH, Kolliputi N, Ko YH, et al. Phosphoinositide 3-kinase-δ regulates fungus-induced allergic lung inflammation through endoplasmic reticulum stress. Thorax. 2016; 71:52–63.
Article
101. Rommel C, Camps M, Ji H. PI3K delta and PI3K gamma: partners in crime in inflammation in rheumatoid arthritis and beyond? Nat Rev Immunol. 2007; 7:191–201.
102. Vanhaesebroeck B, Ali K, Bilancio A, Geering B, Foukas LC. Signalling by PI3K isoforms: insights from gene-targeted mice. Trends Biochem Sci. 2005; 30:194–204.
Article
103. Kwak YG, Song CH, Yi HK, Hwang PH, Kim JS, Lee KS, et al. Involvement of PTEN in airway hyperresponsiveness and inflammation in bronchial asthma. J Clin Invest. 2003; 111:1083–1092.
Article
104. Lee KS, Lee HK, Hayflick JS, Lee YC, Puri KD. Inhibition of phosphoinositide 3-kinase delta attenuates allergic airway inflammation and hyperresponsiveness in murine asthma model. FASEB J. 2006; 20:455–465.
105. Barnes PJ. Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2013; 131:636–645.
Article
106. Park SW, Zhou Y, Lee J, Lu A, Sun C, Chung J, et al. The regulatory subunits of PI3K, p85alpha and p85beta, interact with XBP-1 and increase its nuclear translocation. Nat Med. 2010; 16:429–437.
107. Zhang W, Neo SP, Gunaratne J, Poulsen A, Boping L, Ong EH, et al. Feedback regulation on PTEN/AKT pathway by the ER stress kinase PERK mediated by interaction with the Vault complex. Cell Signal. 2015; 27:436–442.
Article
108. Koga T, Suico MA, Shimasaki S, Watanabe E, Kai Y, Koyama K, et al. Endoplasmic reticulum (ER) stress induces sirtuin 1 (SIRT1) expression via the PI3K-Akt-GSK3β signaling pathway and promotes hepatocellular injury. J Biol Chem. 2015; 290:30366–30374.
Article
109. Ciencewicki J, Trivedi S, Kleeberger SR. Oxidants and the pathogenesis of lung diseases. J Allergy Clin Immunol. 2008; 122:456–468.
Article
110. Rahman I, Morrison D, Donaldson K, MacNee W. Systemic oxidative stress in asthma, COPD, and smokers. Am J Respir Crit Care Med. 1996; 154:1055–1060.
Article
111. Kim SR, Lee KS, Park SJ, Min KH, Lee MH, Lee KA, et al. A novel dithiol amide CB3 attenuates allergic airway disease through negative regulation of p38 mitogen-activated protein kinase. Am J Respir Crit Care Med. 2011; 183:1015–1024.
Article
112. Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell. 2012; 48:158–167.
Article
113. Martínez-Reyes I, Diebold LP, Kong H, Schieber M, Huang H, Hensley CT, et al. TCA cycle and mitochondrial membrane potential are necessary for diverse biological functions. Mol Cell. 2016; 61:199–209.
Article
114. Malhotra JD, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid Redox Signal. 2007; 9:2277–2293.
Article
115. Malhotra JD, Miao H, Zhang K, Wolfson A, Pennathur S, Pipe SW, et al. Antioxidants reduce endoplasmic reticulum stress and improve protein secretion. Proc Natl Acad Sci U S A. 2008; 105:18525–18530.
Article
116. Deniaud A, Sharaf el dein O, Maillier E, Poncet D, Kroemer G, Lemaire C, et al. Endoplasmic reticulum stress induces calcium-dependent permeability transition, mitochondrial outer membrane permeabilization and apoptosis. Oncogene. 2008; 27:285–299.
Article
117. Viner RI, Williams TD, Schöneich C. Nitric oxide-dependent modification of the sarcoplasmic reticulum Ca-ATPase: localization of cysteine target sites. Free Radic Biol Med. 2000; 29:489–496.
Article
118. Kim SR, Kim DI, Kim SH, Lee H, Lee KS, Cho SH, et al. NLRP3 inflammasome activation by mitochondrial ROS in bronchial epithelial cells is required for allergic inflammation. Cell Death Dis. 2014; 5:e1498.
Article
119. Kim HJ, Koo SY, Ahn BH, Park O, Park DH, Seo DO, et al. NecroX as a novel class of mitochondrial reactive oxygen species and ONOO scavenger. Arch Pharm Res. 2010; 33:1813–1823.
120. Bronner DN, Abuaita BH, Chen X, Fitzgerald KA, Nuñez G, He Y, et al. Endoplasmic reticulum stress activates the inflammasome via NLRP3- and Caspase-2-driven mitochondrial damage. Immunity. 2015; 43:451–462.
Article
121. Simpson JL, Phipps S, Baines KJ, Oreo KM, Gunawardhana L, Gibson PG. Elevated expression of the NLRP3 inflammasome in neutrophilic asthma. Eur Respir J. 2014; 43:1067–1076.
Article
122. Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015; 21:248–255.
Article
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr