Korean J Physiol Pharmacol.  2019 Jul;23(4):251-261. 10.4196/kjpp.2019.23.4.251.

Magnolol exerts anti-asthmatic effects by regulating Janus kinase-signal transduction and activation of transcription and Notch signaling pathways and modulating Th1/Th2/Th17 cytokines in ovalbumin-sensitized asthmatic mice

Affiliations
  • 1Department of Gerontology, Wujiang Hospital Affiliated to Nantong University, Suzhou, Jiangsu 215505, China. ISwoggerisits@yahoo.com

Abstract

Allergic asthma, is a common chronic inflammatory disease of the airway presenting with airway hyperresponsiveness and airway remodelling. T helper cells-derived cytokines are critically associated with asthma pathogenesis. Janus kinase-signal transduction and activation of transcription (JAK/STAT) signaling is found to be involved in asthma. Magnolol is a plant-derived bioactive compound with several pharmacological effects. The study aimed to assess the effects of magnolol in ovalbumin (OVA)-induced asthmatic model. BALB/c mice were sensitized and challenged with OVA. Magnolol (12.5, 25, or 50 mg/kg body weight) was administered to separate groups of animals. Dexamethasone was used as the positive control. Cellular infiltration into the bronchoalveolar lavage fluid (BALF) were reduced on magnolol treatment. The levels of Th2 and Th17 cytokines were reduced with noticeably raised levels of interferon gamma. Lung function was improved effectively along with restoration of bronchial tissue architecture. OVA-specific immunoglobulin E levels in serum and BALF were decreased by magnolol. Magnolol reduced Th17 cell population and effectively modulated the JAK-STAT and Notch 1 signaling. The results suggest the promising use of magnolol in therapy for allergic asthma.

Keyword

Asthma; Cytokines; JAK-STAT pathway; Magnolol

MeSH Terms

Airway Remodeling
Animals
Asthma
Bronchoalveolar Lavage Fluid
Cytokines*
Dexamethasone
Immunoglobulin E
Immunoglobulins
Interferons
Lung
Mice*
Ovalbumin
Ovum
Th17 Cells
Cytokines
Dexamethasone
Immunoglobulin E
Immunoglobulins
Interferons
Ovalbumin

Figure

  • Fig. 1 Effect of magnolol on lung function. (A) Airway resistance and (B) lung compliance of mice exposed to ovalbumin (OVA). Values are represented as mean ± standard deviation; n = 6. *p < 0.05 v.s. control as determined by one way-ANOVA followed by Duncan's multiple range test analysis. Dex, dexamethasone.

  • Fig. 2 Effect of magnolol on cell accumulation in bronchoalveolar lavage fluid (BALF). Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against ovalbumin (OVA) alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. Dex, dexamethasone.

  • Fig. 3 Magnolol reduced levels of ovalbumin (OVA)-specific immunoglobulin E (IgE). Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against OVA alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. BALF, bronchoalveolar lavage fluid; Dex, dexamethasone.

  • Fig. 4 Effect of magnolol on cytokines. Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against ovalbumin (OVA) alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. IL, interleukin; Dex, dexamethasone.

  • Fig. 5 Magnolol regulates T helper 17 (Th17) cell populations in bronchoalveolar lavage fluid (BALF). Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against ovalbumin (OVA) alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. Dex, dexamethasone.

  • Fig. 6 Magnolol reduced inflammation score following ovalbumin (OVA) induction. Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against OVA alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. Dex, dexamethasone.

  • Fig. 7 Magnolol activated the Janus kinase (JAK)/signal transduction and activation of transcription (STAT) pathway following ovalbumin (OVA) challenge. L1, control; L2, OVA; L3, OVA + magnolol (12.5 mg/kg); L4, OVA + magnolol (25 mg/kg); L5, OVA + magnolol (50 mg/kg); L6, OVA + dexamethasone (2 mg/kg).

  • Fig. 8 Effect of magnolol on Notch signaling. (A) Representative immunoblot. (B) Relative expressions of proteins. Values are represented as mean ± standard deviation; n = 6. *p < 0.05 compared against control; #p < 0.05 compared against ovalbumin (OVA) alone group; a–eSignificant difference (p < 0.05) between mean values within the groups as determined by one-way ANOVA followed by Duncan's multiple range test analysis. L1, control; L2, OVA; L3, OVA + magnolol (12.5 mg/kg); L4, OVA + magnolol (25 mg/kg); L5, OVA + magnolol (50 mg/kg); L6, OVA + dexamethasone (2 mg/kg).

  • Fig. 9 Graphical representation of the protective effects exerted by magnolol. OVA, ovalbumin; Th, T helper; IL, interleukin; IFN, interferon; BALF, bronchoalveolar lavage fluid; IgE, immunoglobulin E; JAK, Janus kinase; STAT, signal transduction and activation of transcription.


Reference

1. Pawankar R, Canonica GW, Holgate ST, Lockey RF, Blaiss M. WAO white book on allergy [Internet]. Milwaukee: World Allergy Organization;2013. cited 2017 Oct 16. Available from: http://www.worldallergy.org/UserFiles/file/ExecSummary-2013-v6-hires.pdf.
2. Galli SJ, Tsai M, Piliponsky AM. The development of allergic inflammation. Nature. 2008; 454:445–454.
Article
3. Kim MS, Cho KA, Cho YJ, Woo SY. Effects of interleukin-9 blockade on chronic airway inflammation in murine asthma models. Allergy Asthma Immunol Res. 2013; 5:197–206.
Article
4. Wegmann M. Th2 cells as targets for therapeutic intervention in allergic bronchial asthma. Expert Rev Mol Diagn. 2009; 9:85–100.
Article
5. Bosnjak B, Stelzmueller B, Erb KJ, Epstein MM. Treatment of allergic asthma: modulation of Th2 cells and their responses. Respir Res. 2011; 12:114.
Article
6. Simon D, Braathen LR, Simon HU. Eosinophils and atopic dermatitis. Allergy. 2004; 59:561–570.
Article
7. Nakajima H, Hirose K. Role of IL-23 and Th17 cells in airway inflammation in asthma. Immune Netw. 2010; 10:1–4.
Article
8. Brewer JM, Conacher M, Hunter CA, Mohrs M, Brombacher F, Alexander J. Aluminium hydroxide adjuvant initiates strong antigen-specific Th2 responses in the absence of IL-4- or IL-13-mediated signaling. J Immunol. 1999; 163:6448–6454.
9. Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, Overbergh L, Mathieu C, Ceuppens JL. Interleukin-17 orchestrates the granulocyte influx into airways after allergen inhalation in a mouse model of allergic asthma. Am J Respir Cell Mol Biol. 2003; 28:42–50.
Article
10. Schmidt-Weber CB, Akdis M, Akdis CA. TH17 cells in the big picture of immunology. J Allergy Clin Immunol. 2007; 120:247–254.
Article
11. Sergejeva S, Ivanov S, Lötvall J, Lindén A. Interleukin-17 as a recruitment and survival factor for airway macrophages in allergic airway inflammation. Am J Respir Cell Mol Biol. 2005; 33:248–253.
Article
12. Ashino S, Takeda K, Li H, Taylor V, Joetham A, Pine PR, Gelfand EW. Janus kinase 1/3 signaling pathways are key initiators of TH2 differentiation and lung allergic responses. J Allergy Clin Immunol. 2014; 133:1162–1174.
Article
13. Li RF, Wang GF. JAK/STAT5 signaling pathway inhibitor ruxolitinib reduces airway inflammation of neutrophilic asthma in mice model. Eur Rev Med Pharmacol Sci. 2018; 22:835–843.
14. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. 2017; 77:521–546.
15. Hsieh YY, Chang CC, Hsu CM, Wan L, Chen SY, Lin WH, Tsai FJ. JAK-1 rs2780895 C-related genotype and allele but not JAK-1 rs10789166, rs4916008, rs2780885, rs17127114, and rs3806277 are associated with higher susceptibility to asthma. Genet Test Mol Biomarkers. 2011; 15:841–847.
Article
16. Shen Y, Liu Y, Ke X, Kang HY, Hu GH, Hong SL. Association between JAK1 gene polymorphisms and susceptibility to allergic rhinitis. Asian Pac J Allergy Immunol. 2016; 34:124–129.
Article
17. Gandhi NA, Bennett BL, Graham NM, Pirozzi G, Stahl N, Yancopoulos GD. Targeting key proximal drivers of type 2 inflammation in disease. Nat Rev Drug Discov. 2016; 15:35–50.
Article
18. Hu C, Li Z, Feng J, Tang Y, Qin L, Hu X, Zhang Y, He R. Glucocorticoids modulate Th1 and Th2 responses in asthmatic mouse models by inhibition of Notch1 signaling. Int Arch Allergy Immunol. 2018; 175:44–52.
Article
19. Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nat Rev Immunol. 2009; 9:116–124.
Article
20. Kang JH, Kim BS, Uhm TG, Lee SH, Lee GR, Park CS, Chung IY. Gamma-secretase inhibitor reduces allergic pulmonary inflammation by modulating Th1 and Th2 responses. Am J Respir Crit Care Med. 2009; 179:875–882.
21. Guo XJ, Zhou M, Ren LP, Yang M, Huang SG, Xu WG. Small interfering RNA-mediated knockdown of Notch1 in lung. Chin Med J (Engl). 2009; 122:2647–2651.
22. Vafeiadou K, Vauzour D, Lee HY, Rodriguez-Mateos A, Williams RJ, Spencer JP. The citrus flavanone naringenin inhibits inflammatory signalling in glial cells and protects against neuroinflammatory injury. Arch Biochem Biophys. 2009; 484:100–109.
Article
23. Lim H, Park H, Kim HP. Effects of flavonoids on matrix metalloproteinase-13 expression of interleukin-1β-treated articular chondrocytes and their cellular mechanisms: inhibition of c-Fos/AP-1 and JAK/STAT signaling pathways. J Pharmacol Sci. 2011; 116:221–231.
Article
24. Cheng YC, Tsao MJ, Chiu CY, Kan PC, Chen Y. Magnolol inhibits human glioblastoma cell migration by regulating N-cadherin. J Neuropathol Exp Neurol. 2018; 77:426–436.
Article
25. Ranaware AM, Banik K, Deshpande V, Padmavathi G, Roy NK, Sethi G, Fan L, Kumar AP, Kunnumakkara AB. Magnolol: a neolignan from the magnolia family for the prevention and treatment of cancer. Int J Mol Sci. 2018; 19:2362.
Article
26. Amorati R, Zotova J, Baschieri A, Valgimigli L. Antioxidant activity of magnolol and honokiol: kinetic and mechanistic investigations of their reaction with peroxyl radicals. J Org Chem. 2015; 80:10651–10659.
Article
27. Huang SY, Tai SH, Chang CC, Tu YF, Chang CH, Lee EJ. Magnolol protects against ischemic-reperfusion brain damage following oxygen-glucose deprivation and transient focal cerebral ischemia. Int J Mol Med. 2018; 41:2252–2262.
Article
28. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the care and use of laboratory animals. 8th ed. Washington: National Academies Press;2011.
29. Oh SW, Pae CI, Lee DK, Jones F, Chiang GK, Kim HO, Moon SH, Cao B, Ogbu C, Jeong KW, Kozu G, Nakanishi H, Kahn M, Chi EY, Henderson WR Jr. Tryptase inhibition blocks airway inflammation in a mouse asthma model. J Immunol. 2002; 168:1992–2000.
Article
30. Djukanović R, Roche WR, Wilson JW, Beasley CR, Twentyman OP, Howarth RH, Holgate ST. Mucosal inflammation in asthma. Am Rev Respir Dis. 1990; 142:434–457.
Article
31. Pichavant M, Goya S, Hamelmann E, Gelfand EW, Umetsu DT. Animal models of airway sensitization. Curr Protoc Immunol. 2007; Chapter 15:Unit 15.18.
Article
32. Glaab T, Daser A, Braun A, Neuhaus-Steinmetz U, Fabel H, Alarie Y, Renz H. Tidal midexpiratory flow as a measure of airway hyperresponsiveness in allergic mice. Am J Physiol Lung Cell Mol Physiol. 2001; 280:L565–L573.
Article
33. Duan W, Chan JH, Wong CH, Leung BP, Wong WS. Anti-inflammatory effects of mitogen-activated protein kinase kinase inhibitor U0126 in an asthma mouse model. J Immunol. 2004; 172:7053–7059.
Article
34. Tabatabaian F, Ledford DK. Omalizumab for severe asthma: toward personalized treatment based on biomarker profile and clinical history. J Asthma Allergy. 2018; 11:53–61.
Article
35. Holgate ST, Wenzel S, Postma DS, Weiss ST, Renz H, Sly PD. Asthma. Nat Rev Dis Primers. 2015; 1:15025.
Article
36. Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015; 16:45–56.
Article
37. Tritar-Cherif F, Ben M'Rad S, Merai S, Djenayah F. Corticotherapy for asthma in the child. Tunis Med. 2002; 80:1–6. French.
38. Southworth T, Mason S, Bell A, Ramis I, Calbet M, Domenech A, Prats N, Miralpeix M, Singh D. PI3K, p38 and JAK/STAT signalling in bronchial tissue from patients with asthma following allergen challenge. Biomark Res. 2018; 6:14.
Article
39. Kleiman A, Tuckermann JP. Glucocorticoid receptor action in beneficial and side effects of steroid therapy: lessons from conditional knockout mice. Mol Cell Endocrinol. 2007; 275:98–108.
Article
40. Hocaoglu AB, Karaman O, Erge DO, Erbil G, Yilmaz O, Bagriyanik A, Uzuner N. Glycyrrhizin and long-term histopathologic changes in a murine model of asthma. Curr Ther Res Clin Exp. 2011; 72:250–261.
Article
41. Oh SW, Cha JY, Jung JE, Chang BC, Kwon HJ, Lee BR, Kim DY. Curcumin attenuates allergic airway inflammation and hyper-responsiveness in mice through NF-κB inhibition. J Ethnopharmacol. 2011; 136:414–421.
Article
42. Zhang W, Zhang X, Sheng A, Weng C, Zhu T, Zhao W, Li C. γ-Secretase inhibitor alleviates acute airway inflammation of allergic asthma in mice by downregulating Th17 cell differentiation. Mediators Inflamm. 2015; 2015:258168.
Article
43. Deshmukh R, Kaundal M, Bansal V, Samardeep . Caffeic acid attenuates oxidative stress, learning and memory deficit in intra-cerebroventricular streptozotocin induced experimental dementia in rats. Biomed Pharmacother. 2016; 81:56–62.
Article
44. Wenzel SE. Asthma: defining of the persistent adult phenotypes. Lancet. 2006; 368:804–813.
Article
45. Turner DJ, Mulholland MW. Calcium signaling pathways in the enteric nervous system. Int J Surg Investig. 1999; 1:87–97.
46. Elsner J, Kapp A. Regulation and modulation of eosinophil effector functions. Allergy. 1999; 54:15–26.
Article
47. Wei M, Chu X, Guan M, Yang X, Xie X, Liu F, Chen C, Deng X. Protocatechuic acid suppresses ovalbumin-induced airway inflammation in a mouse allergic asthma model. Int Immunopharmacol. 2013; 15:780–788.
Article
48. Lee MY, Seo CS, Lee JA, Lee NH, Kim JH, Ha H, Zheng MS, Son JK, Shin HK. Anti-asthmatic effects of Angelica dahurica against ovalbumin-induced airway inflammation via upregulation of heme oxygenase-1. Food Chem Toxicol. 2011; 49:829–837.
Article
49. Busse WW, Lemanske RF Jr. Asthma. N Engl J Med. 2001; 344:350–362.
Article
50. Busse WW, Coffman RL, Gelfand EW, Kay AB, Rosenwasser LJ. Mechanisms of persistent airway inflammation in asthma. A role for T cells and T-cell products. Am J Respir Crit Care Med. 1995; 152:388–393.
Article
51. Cohn L, Tepper JS, Bottomly K. IL-4-independent induction of airway hyperresponsiveness by Th2, but not Th1, cells. J Immunol. 1998; 161:3813–3816.
52. Bisset LR, Schmid-Grendelmeier P. Chemokines and their receptors in the pathogenesis of allergic asthma: progress and perspective. Curr Opin Pulm Med. 2005; 11:35–42.
Article
53. Ngoc PL, Gold DR, Tzianabos AO, Weiss ST, Celedón JC. Cytokines, allergy, and asthma. Curr Opin Allergy Clin Immunol. 2005; 5:161–166.
Article
54. Desai D, Brightling C. Cytokines and cytokine-specific therapy in asthma. Adv Clin Chem. 2012; 57:57–97.
Article
55. Fish SC, Donaldson DD, Goldman SJ, Williams CM, Kasaian MT. IgE generation and mast cell effector function in mice deficient in IL-4 and IL-13. J Immunol. 2005; 174:7716–7724.
Article
56. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004; 21:467–476.
Article
57. Wakashin H, Hirose K, Maezawa Y, Kagami S, Suto A, Watanabe N, Saito Y, Hatano M, Tokuhisa T, Iwakura Y, Puccetti P, Iwamoto I, Nakajima H. IL-23 and Th17 cells enhance Th2-cell-mediated eosinophilic airway inflammation in mice. Am J Respir Crit Care Med. 2008; 178:1023–1032.
Article
58. Li J, Zhang B. Apigenin protects ovalbumin-induced asthma through the regulation of Th17 cells. Fitoterapia. 2013; 91:298–304.
Article
59. Sun YC, Zhou QT, Yao WZ. Sputum interleukin-17 is increased and associated with airway neutrophilia in patients with severe asthma. Chin Med J (Engl). 2005; 118:953–956.
60. Wilson RH, Whitehead GS, Nakano H, Free ME, Kolls JK, Cook DN. Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness. Am J Respir Crit Care Med. 2009; 180:720–730.
Article
61. Ma C, Ma Z, Fu Q, Ma S. Curcumin attenuates allergic airway inflammation by regulation of CD4+CD25+ regulatory T cells (Tregs)/Th17 balance in ovalbumin-sensitized mice. Fitoterapia. 2013; 87:57–64.
Article
62. Ji X, Han M, Yun Y, Li G, Sang N. Acute nitrogen dioxide (NO2) exposure enhances airway inflammation via modulating Th1/Th2 differentiation and activating JAK-STAT pathway. Chemosphere. 2015; 120:722–728.
Article
63. Mao X, Ren Z, Parker GN, Sondermann H, Pastorello MA, Wang W, McMurray JS, Demeler B, Darnell JE Jr, Chen X. Structural bases of unphosphorylated STAT1 association and receptor binding. Mol Cell. 2005; 17:761–771.
Article
64. O'Shea JJ, Kontzias A, Yamaoka K, Tanaka Y, Laurence A. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis. 2013; 72:Suppl 2. ii111–ii115.
65. O'Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med. 2013; 368:161–170.
66. Zhong Z, Wen Z, Darnell JE Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994; 264:95–98.
Article
67. Abroun S, Saki N, Ahmadvand M, Asghari F, Salari F, Rahim F. STATs: an old story, yet mesmerizing. Cell J. 2015; 17:395–411.
68. Walford HH, Doherty TA. STAT6 and lung inflammation. JAKSTAT. 2013; 2:e25301.
Article
69. Dallman MJ, Smith E, Benson RA, Lamb JR. Notch: control of lymphocyte differentiation in the periphery. Curr Opin Immunol. 2005; 17:259–266.
Article
70. Osborne BA, Minter LM. Notch signalling during peripheral T-cell activation and differentiation. Nat Rev Immunol. 2007; 7:64–75.
Article
71. Laky K, Fowlkes BJ. Notch signaling in CD4 and CD8 T cell development. Curr Opin Immunol. 2008; 20:197–202.
Article
72. Zhang W, Nie Y, Chong L, Cai X, Zhang H, Lin B, Liang Y, Li C. PI3K and Notch signal pathways coordinately regulate the activation and proliferation of T lymphocytes in asthma. Life Sci. 2013; 92:890–895.
Article
Full Text Links
  • KJPP
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