Korean J Physiol Pharmacol.  2018 Sep;22(5):481-491. 10.4196/kjpp.2018.22.5.481.

Influence of rutin on the effects of neonatal cigarette smoke exposure-induced exacerbated MMP-9 expression, Th17 cytokines and NF-κB/iNOS-mediated inflammatory responses in asthmatic mice model

Affiliations
  • 1Children's Medical Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R.China. lfhfuhai@hotmail.com
  • 2Department of Pathology, Shandong University of Medicine, Jinan, Shandong 250012, P.R.China.

Abstract

Allergic asthma is one of the most enduring diseases of the airway. The T-helper cells and regulatory T-cells are critically involved in inflammatory responses, mucus hypersecretion, airway remodelling and in airway hyper-responsiveness. Cigarette smoke (CS) has been found to aggravate inflammatory responses in asthma. Though currently employed drugs are effective, associated side effects demand identification and development of novel drugs with negligible or no adverse effects. Rutin, plant-derived flavonoid has been found to possess antioxidant and anti-inflammatory effects. We investigated the ability of rutin to modulate T-cells and inhibit inflammation in experimentally-induced asthma in cigarette smoke exposed mice. Separate groups of neonatal mice were exposed to CS for 10 days from post-natal days 2 to 11. After 2 weeks, the mice were sensitized and challenged with ovalbumin (OVA). Treatment group were given rutin (37.5 or 75 mg/kg body weight) during OVA sensitization and challenge. Rutin treatment was found to significantly inhibit cellular infiltration in the airways and Th2 and Th17 cytokine levels as well. Flow cytometry revealed effectively raised CD4⁺CD25⁺Fox3⁺ Treg cells and supressed Th17 cell population on rutin treatment. Airway hyper-responsiveness observed following CS and OVA challenge were inhibited by rutin. NF-κB and iNOS, chief regulators of inflammatory responses robustly activated by CS and OVA were down-regulated by rutin. Rutin also inhibited the expression of matrix metalloproteinase 9, thereby aiding in prevention of airway remodelling in asthma thereby revealing to be a potent candidate in asthma therapy.

Keyword

Asthma; Cigarette smoke; Matrix metalloproteinase; Regulatory T cells; Rutin

MeSH Terms

Airway Remodeling
Animals
Asthma
Cytokines*
Flow Cytometry
Inflammation
Matrix Metalloproteinase 9
Mice*
Mucus
Ovalbumin
Ovum
Respiratory Hypersensitivity
Rutin*
Smoke*
T-Lymphocytes
T-Lymphocytes, Regulatory
Th17 Cells
Tobacco Products*
Cytokines
Matrix Metalloproteinase 9
Ovalbumin
Rutin
Smoke

Figure

  • Fig. 1 Rutin reduced airway resistance in lung function following CS and OVA challenge. Data are given as mean±SD where n=6. *Symbolizes p<0.05 related with control as determined by one way-ANOVA.

  • Fig. 2 Rutin improved lung compliance following CS and OVA challenge. Data are given as mean±SD where n=6. *Symbolizes p<0.05 related with control as determined by one way-ANOVA.

  • Fig. 3 Effect of Rutin on cell accumulation in BALF. Rutin reduced inflammatory cell infiltration in to BALF following CS and OVA challenge. Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~gdenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.

  • Fig. 4 Rutin reduces OVA-specific IgE levels in serum and BALF. Rutin was found to significantly reduce the raised OVA-specific IgE in serum and in the BALF in mice exposed to CS and/or OVA. Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~idenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.

  • Fig. 5 Rutin caused marked decrease in the levels of Th2 cytokines and increased the levels of IFN-γ. Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~idenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.

  • Fig. 6 Rutin caused marked decrease in the levels of Th17 cytokines and increased the levels of IL-10. Data are given as mean±SD where n=6. adenotes statistical significance at p<0.05 related against control and b~idenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.

  • Fig. 7 Rutin regulates Th17 and CD4+CD25+Foxp3+ Treg cell populations. Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~hdenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.

  • Fig. 8 Rutin was found to reduce inflammatory cell infiltration as in H&E staining (A) and reduce inflammation score (B). Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~hdenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis. (a) Control, (b) OVA+Rutin (75 mg), (c) CS+OVA+Rutin (75 mg), (d) CS+OVA+DEX.

  • Fig. 9 Rutin significantly reduce mucus hypersecretion and goblet cell hyperplasia as in PAFS staining (A and B). Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~hdenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis. (a) Control, (b) OVA, (c) OVA+Rutin (75 mg), (d) CS+OVA, (e) CS+OVA+Rutin (75 mg), (f) CS+OVA+DEX.

  • Fig. 10 Rutin modulates NF-κB/iNOS-mediated signaling and reduce MMP-9 expression. (A) L1, Control; L2, OVA; L3, OVA+Rutin (37.5 mg); L4, OVA+Rutin (75 mg); L5, CS+OVA; L6, CS+OVA+Rutin (37.5 mg); L7, CS+OVA+Rutin (75 mg); L8, CS+OVA+DEX. (B~D) Data are given as mean±SD where n=6. aDenotes statistical significance at p<0.05 related against control and b~hdenotes data within the same group that differ from each other at p<0.05 as calculated by one-way ANOVA followed by DMRT analysis.


Reference

1. Galli SJ, Tsai M, Piliponsky AM. The development of allergic inflammation. Nature. 2008; 454:445–454.
Article
2. Medoff BD, Thomas SY, Luster AD. T cell trafficking in allergic asthma: the ins and outs. Annu Rev Immunol. 2008; 26:205–232.
Article
3. 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
4. Nakajima H, Hirose K. Role of IL-23 and Th17 cells in air way inflammation in asthma. Immune Netw. 2010; 10:1–4.
5. 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.
6. Schmidt-Weber CB, Akdis M, Akdis CA. TH17 cells in the big picture of immunology. J Allergy Clin Immunol. 2007; 120:247–254.
Article
7. 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
8. 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
9. Boulet LP, Turcott H, Plante S, Chakir J. Airway function, inflammation and regulatory T cell function in subjects in asthma remission. Can Respir J. 2012; 19:19–25.
Article
10. Umetsu DT, DeKruyff RH. The regulation of allergy and asthma. Immunol Rev. 2006; 212:238–255.
Article
11. Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev. 2006; 212:8–27.
12. Choi IW, Kim DK, Ko HM, Lee HK. Administration of antisense phosphorothioate oligonucleotide to the p65 subunit of NF-kappaB inhibits established asthmatic reaction in mice. Int Immunopharmacol. 2004; 4:1817–1828.
13. Gagliardo R, Chanez P, Mathieu M, Bruno A, Costanzo G, Gougat C, Vachier I, Bousquet J, Bonsignore G, Vignola AM. Persistent activation of nuclear factor-kappaB signaling pathway in severe uncontrolled asthma. Am J Respir Crit Care Med. 2003; 168:1190–1198.
14. Jeon WY, Shin IS, Shin HK, Lee MY. Samsoeum water extract attenuates allergic airway inflammation via modulation of Th1/Th2 cytokines and decrease of iNOS expression in asthmatic mice. BMC Complement Altern Med. 2015; 15:47.
Article
15. Yoo D, Guk K, Kim H, Khang G, Wu D, Lee D. Antioxidant polymeric nanoparticles as novel therapeutics for airway inflammatory diseases. Int J Pharm. 2013; 450:87–94.
Article
16. Elias JA. Airway remodeling in asthma. Unanswered questions. Am J Respir Crit Care Med. 2000; 161:S168–S171.
17. Bossé M, Chakir J, Rouabhia M, Boulet LP, Audette M, Laviolette M. Serum matrix metalloproteinase-9:Tissue inhibitor of metalloproteinase-1 ratio correlates with steroid responsiveness in moderate to severe asthma. Am J Respir Crit Care Med. 1999; 159:596–602.
Article
18. Tritar-Cherif F, Ben M'Rad S, Merai S, Djenayah F. Corticotherapy for asthma in the child. Tunis Med. 2002; 80:1–6.
19. 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
20. Li XM, Brown L. Efficacy and mechanisms of action of traditional Chinese medicines for treating asthma and allergy. J Allergy Clin Immunol. 2009; 123:297–306. quiz 307–308.
Article
21. Cho SJ, Kim HW, Kim BY, Cho SI. Sam So Eum, a herb extract, as the remedy for allergen-induced asthma in mice. Pulm Pharmacol Ther. 2008; 21:578–583.
Article
22. Metodiewa D, Kochman A, Karolczak S. Evidence for antiradical and antioxidant properties of four biologically active N,N-diethylaminoethyl ethers of flavanone oximes: a comparison with natural polyphenolic flavonoid (rutin) action. Biochem Mol Biol Int. 1997; 41:1067–1075.
23. Guardia T, Rotelli AE, Juarez AO, Pelzer LE. Anti-inflammatory properties of plant flavonoids. Effects of rutin, quercetin and hesperidin on adjuvant arthritis in rat. Farmaco. 2001; 56:683–687.
Article
24. Siroux V, Pin I, Oryszczyn MP, Le Moual N, Kauffmann F. Relationships of active smoking to asthma and asthma severity in the EGEA study. Epidemiological study on the Genetics and Environment of Asthma. Eur Respir J. 2000; 15:470–477.
25. Barrett EG, Wilder JA, March TH, Espindola T, Bice DE. Cigarette smoke-induced airway hyperresponsiveness is not dependent on elevated immunoglobulin and eosinophilic inflammation in a mouse model of allergic airway disease. Am J Respir Crit Care Med. 2002; 165:1410–1418.
Article
26. Wu ZX, Benders KB, Hunter DD, Dey RD. Early postnatal exposure of mice to side-steam tobacco smoke increases neuropeptide Y in lung. Am J Physiol Lung Cell Mol Physiol. 2012; 302:L152–L159.
Article
27. Pinkerton KE, Joad JP. The mammalian respiratory system and critical windows of exposure for children's health. Environ Health Perspect. 2000; 108:Suppl 3. 457–462.
Article
28. 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
29. 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
30. Jain VV, Kitagaki K, Businga T, Hussain I, George C, O'shaughnessy P, Kline JN. CpG-oligodeoxynucleotides inhibit airway remodeling in a murine model of chronic asthma. J Allergy Clin Immunol. 2002; 110:867–872.
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. Bao Z, Guan S, Cheng C, Wu S, Wong SH, Kemeny DM, Leung BP, Wong WS. A novel antiinflammatory role for andrographolide in asthma via inhibition of the nuclear factor-kappaB pathway. Am J Respir Crit Care Med. 2009; 179:657–665.
34. 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
35. Lee YC, Lee HB, Rhee YK, Song CH. The involvement of matrix metalloproteinase-9 in airway inflammation of patients with acute asthma. Clin Exp Allergy. 2001; 31:1623–1630.
Article
36. Busse WW, Lemanske RF Jr. Asthma. N Engl J Med. 2001; 344:350–362.
Article
37. Moerloose KB, Robays LJ, Maes T, Brusselle GG, Tournoy KG, Joos GF. Cigarette smoke exposure facilitates allergic sensitization in mice. Respir Res. 2006; 7:49–57.
Article
38. Yang SR, Chida AS, Bauter MR, Shafiq N, Seweryniak K, Maggirwar SB, Kilty I, Rahman I. Cigarette smoke induces proinflammatory cytokine release by activation of NF-kappaB and posttranslational modifications of histone deacetylase in macrophages. Am J Physiol Lung Cell Mol Physiol. 2006; 291:L46–L57.
39. Elsner J, Kapp A. Regulation and modulation of eosinophil effector functions. Allergy. 1999; 54:15–26.
Article
40. Wenzel SE. Asthma: defining of the persistent adult phenotypes. Lancet. 2006; 368:804–813.
Article
41. Li J, Zhang B. Apigenin protects ovalbumin-induced asthma through the regulation of Th17 cells. Fitoterapia. 2013; 91:298–304.
Article
42. Simon D, Braathen LR, Simon HU. Eosinophils and atopic dermatitis. Allergy. 2004; 59:561–570.
Article
43. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004; 21:467–476.
Article
44. Cockcroft DW, Davis BE. Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol. 2006; 118:551–559. quiz 560–561.
Article
45. Royer B, Varadaradjalou S, Saas P, Guillosson JJ, Kantelip JP, Arock M. Inhibition of IgE-induced activation of human mast cells by IL-10. Clin Exp Allergy. 2001; 31:694–704.
Article
46. Nembrini C, Marsland BJ, Kopf M. IL-17-producing T cells in lung immunity and inflammation. J Allergy Clin Immunol. 2009; 123:986–994. quiz 995–996.
Article
47. Siebenlist U, Brown K, Claudio E. Control of lymphocyte development by nuclear factor-kappaB. Nat Rev Immunol. 2005; 5:435–445.
48. Rahman MS, Yamasaki A, Yang J, Shan L, Halayko AJ, Gounni AS. IL-17A induces eotaxin-1/CC chemokine ligand 11 expression in human airway smooth muscle cells: role of MAPK (Erk1/2, JNK, and p38) pathways. J Immunol. 2006; 177:4064–4071.
Article
49. Suresh V, Mih JD, George SC. Measurement of IL-13-induced iNOS-derived gas phase nitric oxide in human bronchial epithelial cells. Am J Respir Cell Mol Biol. 2007; 37:97–104.
Article
50. Grzela K, Zagorska W, Krejner A, Litwiniuk M, Zawadzka-Krajewska A, Banaszkiewicz A, Kulus M, Grzela T. Prolonged treatment with inhaled corticosteroids does not normalize high activity of matrix metalloproteinase-9 in exhaled breath condensates of children with asthma. Arch Immunol Ther Exp (Warsz). 2015; 63:231–237.
Article
51. Mehra D, Sternberg DI, Jia Y, Canfield S, Lemaitre V, Nkyimbeng T, Wilder J, Sonett J, D'Armiento J. Altered lymphocyte trafficking and diminished airway reactivity in transgenic mice expressing human MMP-9 in a mouse model of asthma. Am J Physiol Lung Cell Mol Physiol. 2010; 298:L189–L196.
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