Quercetin prevents progression of disease in elastase/LPS-exposed mice by negatively regulating MMP expression

Shyamala Ganesan, Andrea N Faris, Adam T Comstock, Sangbrita S Chattoraj, Asamanja Chattoraj, John R Burgess, Jeffrey L Curtis, Fernando J Martinez, Suzanna Zick, Marc B Hershenson, Uma S Sajjan, Shyamala Ganesan, Andrea N Faris, Adam T Comstock, Sangbrita S Chattoraj, Asamanja Chattoraj, John R Burgess, Jeffrey L Curtis, Fernando J Martinez, Suzanna Zick, Marc B Hershenson, Uma S Sajjan

Abstract

Background: Chronic obstructive pulmonary disease (COPD) is characterized by chronic bronchitis, emphysema and irreversible airflow limitation. These changes are thought to be due to oxidative stress and an imbalance of proteases and antiproteases. Quercetin, a plant flavonoid, is a potent antioxidant and anti-inflammatory agent. We hypothesized that quercetin reduces lung inflammation and improves lung function in elastase/lipopolysaccharide (LPS)-exposed mice which show typical features of COPD, including airways inflammation, goblet cell metaplasia, and emphysema.

Methods: Mice treated with elastase and LPS once a week for 4 weeks were subsequently administered 0.5 mg of quercetin dihydrate or 50% propylene glycol (vehicle) by gavage for 10 days. Lungs were examined for elastance, oxidative stress, inflammation, and matrix metalloproteinase (MMP) activity. Effects of quercetin on MMP transcription and activity were examined in LPS-exposed murine macrophages.

Results: Quercetin-treated, elastase/LPS-exposed mice showed improved elastic recoil and decreased alveolar chord length compared to vehicle-treated controls. Quercetin-treated mice showed decreased levels of thiobarbituric acid reactive substances, a measure of lipid peroxidation caused by oxidative stress. Quercetin also reduced lung inflammation, goblet cell metaplasia, and mRNA expression of pro-inflammatory cytokines and muc5AC. Quercetin treatment decreased the expression and activity of MMP9 and MMP12 in vivo and in vitro, while increasing expression of the histone deacetylase Sirt-1 and suppressing MMP promoter H4 acetylation. Finally, co-treatment with the Sirt-1 inhibitor sirtinol blocked the effects of quercetin on the lung phenotype.

Conclusions: Quercetin prevents progression of emphysema in elastase/LPS-treated mice by reducing oxidative stress, lung inflammation and expression of MMP9 and MMP12.

Figures

Figure 1
Figure 1
Quercetin partially improves lung function in elastase-LPS exposed mice. Mice were anesthetized and pressure-volume relationships, compliance and elastance were measured using flexivent system. Chord length was determined by morphometry. Seven days after the last exposure to elastase/LPS, untreated mice showed a leftward and upward shift in the pressure-volume (PV)-loop (A) compared to PBS-exposed vehicle treated mice, indicative of a loss of elastic recoil. Seventeen days after the last exposure to elastase/LPS, this curve shifted further, indicating progression in the loss of elastic recoil. Quercetin treatment for 10 days prevented progression in loss of elastic recoil in elastase/LPS-exposed mice but had no effect on PBS-exposed mice (B). Each of these mice was examined 17 days after the last elastase/LPS treatment. Elastase/LPS-exposed mice treated with quercetin also showed increased elastance (C), decreased compliance (D) and decreased alveolar chord length (E) compared to vehicle treated mice. Representative PV curves from 5 to 6 mice from each group are shown in A and B. Data in C, D and E represent mean and SD calculated from 6 animals per group (*different from PBS group, p≤0.05 one-way ANOVA; † different from vehicle treated mice, p≤0.05 Mann-Whitney test).
Figure 2
Figure 2
Quercetin treatment reduces TBARS and increases Hmox-1 expression in elastase/LPS exposed mice. Mice exposed to PBS or elastase/LPS were orally gavaged with 0.2 mg of quercetin or vehicle daily for 10 days and sacrificed on the last day of quercetin treatment. A to D. Elastase/LPS exposed mice show increased levels of TBARS, increased iNOS mRNA levels, decreased Hmox-1 mRNA and increased ratio of iNOS/Hmox-1 compared to PBS exposed mice treated with either vehicle or quercetin. Quercetin treatment reduces TBARS, increases Hmox-1 mRNA and decreases ratio of iNOS/Hmox-1 in elastase/LPS-exposed mice. Data represent mean and SEM (n = 10-14, *different from PBS/vehicle and PBS/quercetin group, p≤0.05; † different from vehicle treated elastase/LPS-exposed mice, p≤0.05 one-way ANOVA).
Figure 3
Figure 3
Quercetin treatment decreases chemokine and cytokine levels in elastase/LPS exposed mice. Mice exposed to elastase/LPS were orally gavaged with vehicle or quercetin as described. A to F. Elastase/LPS exposed mice show increased levels of KC, MIP2, MCP-1, IL-1β, IL-12p40 and MIP-1β compared to control naïve mice. Quercetin treatment reduces all the examined cytokines and chemokines. Data represent mean and SEM (n = 10, *different from PBS/vehicle group, p≤0.05; † different from vehicle-treated elastase/LPS exposed mice, p≤0.05 one-way ANOVA).
Figure 4
Figure 4
Quercetin treatment reduces lung inflammation and reverses goblet cell metaplasia. Lung sections from elastase/LPS exposed mice were stained with H & E or immunostained with an antibody to Muc5AC. A and B. Mice treated with vehicle show mild-to-moderate wide-spread lung inflammation, emphysema and goblet cell metaplasia. C and D. Mice treated with quercetin show less emphysema with very mild inflammation and a complete reduction in MUC5AC producing goblet cells. Asterisks in A and C represent emphysema. Arrows in B indicate MUC5AC- producing goblet cells. Images are representative of 6 mice per group. E. Examination of BAL fluid reveals increased numbers of total cells, macrophages and neutrophils in elastase/LPS treated mice, which were almost completely reversed by quercetin treatment. F. qPCR analysis of total lung RNA shows increased Muc5AC transcript levels in elastase/LPS-exposed mice, and this was reduced in quercetin treated mice. Data represent mean and SEM (n = 10, *different from PBS/vehicle group, p≤0.05; † different from vehicle treated elastase/LPS exposed mice, p≤0.05 one-way ANOVA).
Figure 5
Figure 5
Quercetin treatment decreases levels of MMP9 and MMP12 and increases expression of SIRT1 in elastase/LPS-exposed mice. mRNA expression of Mmp9, Mmp12, and Sirt1 was measured by qPCR. MMP activity was determined by gelatin zymography. Sirt1 protein level was measured in the lung homogenates by Western blot analysis. A and B. Quercetin treated elastase/LPS exposed mice show significantly reduced Mmp9 and Mmp12 mRNA levels compared to mice treated with vehicle. C Quercetin treatment completely reduces MMP9 and MMP12 activities in elastase/LPS exposed mice. D and E. Quercetin increases Sirt1 mRNA and protein levels in elastase/LPS exposed mice. F. Ratio of Sirt1 protein/β-actin normalized to control mice calculated from 6 mice per group. Data represent mean and SEM (n = 10, *different from PBS/vehicle group, p≤0.05; † different from vehicle-treated, elastase/LPS-exposed mice, p≤0.05 one-way ANOVA). Images in C and E are representative of 4 to 6 animals per group.
Figure 6
Figure 6
Inhibition of Sirt1 activity in quercetin-treated mice attenuates querecetin-induced changes in MMP expression and emphysema progression. Elastase/LPS-exposed mice were treated with vehicle or quercetin along with sirtinol or PBS. A and B. Mice treated with quercetin and sirtinol did not show reduced MMP9 or MMP12 mRNA expression. Results represent range of data with median (n = 4-6, *different from all other groups, p≤0.05; ANOVA on ranks). C-E. Sirtinol also inhibited quercetin's effects on elastic recoil (C), compliance (D) and elastance (E). Data represent mean and SEM (n = 4-6, *different from all other groups, p≤0.05; one-way ANOVA). C. Representative of 4-6 animals per group.
Figure 7
Figure 7
Quercetin decreases MMP9 and MMP12 and increases Sirt1 in LPS-treated alveolar macrophages. Mouse alveolar macrophages were treated with LPS (1 ng/ml) for 3 days and treated with DMSO (vehicle) or 25 µM quercetin for 16 h. A and B. LPS treatment induced mRNA expression of MMP9 and MMP12 which are partially reversed by quercetin. C and E. Quercetin increases Sirt1 mRNA and protein expression. D. MMP9 activity in LPS-exposed was abrogated by quercetin treatment. E. A representative Western blot showing Sirt1 and β-actin expression. F. Ratio of Sirt1 protein/β-actin normalized to untreated cells calculated from 3 independent experiments. G and H. Quercetin decreased LPS-induced histone H4 acetylation at the NF-κB binding sites in the promoter regions of both Mmp9 and Mmp12. Zymogram and immunoblot images are representative of three independent experiments. Data represent mean and SEM calculated from 3 independent experiments performed in duplicates or triplicates (*different from macrophages exposed to media in the absence of quercetin, p≤0.05; †different from LPS-exposed macrophages which are not treated with quercetin, p≤0.05 one-way ANOVA).

References

    1. Rahman I, Adcock IM. Oxidative stress and redox regulation of lung inflammation in COPD. Eur Respir J. 2006;28:219–242. doi: 10.1183/09031936.06.00053805.
    1. MacNee W, Rahman I. Is oxidative stress central to the pathogenesis of chronic obstructive pulmonary disease? Trends Mol Med. 2001;7:55–62. doi: 10.1016/S1471-4914(01)01912-8.
    1. Sydbom A, Blomberg A, Parnia S, Stenfors N, Sandstrom T, Dahlen SE. Health effects of diesel exhaust emissions. Eur Respir J. 2001;17:733–746. doi: 10.1183/09031936.01.17407330.
    1. Nowak D, Kasielski M, Antczak A, Pietras T, Bialasiewicz P. Increased content of thiobarbituric acid-reactive substances and hydrogen peroxide in the expired breath condensate of patients with stable chronic obstructive pulmonary disease: no significant effect of cigarette smoking. Respir Med. 1999;93:389–396. doi: 10.1053/rmed.1999.0574.
    1. Chung KF, Adcock IM. Multifaceted mechanisms in COPD: inflammation, immunity, and tissue repair and destruction. Eur Respir J. 2008;31:1334–1356. doi: 10.1183/09031936.00018908.
    1. Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM, Barczyk A, Hayashi S, Adcock IM, Hogg JC. et al.Decreased histone deacetylase activity in chronic obstructive pulmonary disease. N Engl J Med. 2005;352:1967–1976. doi: 10.1056/NEJMoa041892.
    1. Nakamaru Y, Vuppusetty C, Wada H, Milne JC, Ito M, Rossios C, Elliot M, Hogg J, Kharitonov S, Goto H. et al.A protein deacetylase SIRT1 is a negative regulator of metalloproteinase-9. FASEB J. 2009;23:2810–2819. doi: 10.1096/fj.08-125468.
    1. Hasday JD, Bascom R, Costa JJ, Fitzgerald T, Dubin W. Bacterial endotoxin is an active component of cigarette smoke. Chest. 1999;115:829–835. doi: 10.1378/chest.115.3.829.
    1. Larsson L, Szponar B, Pehrson C. Tobacco smoking increases dramatically air concentrations of endotoxin. Indoor Air. 2004;14:421–424. doi: 10.1111/j.1600-0668.2004.00290.x.
    1. Sebastian A, Pehrson C, Larsson L. Elevated concentrations of endotoxin in indoor air due to cigarette smoking. J Environ Monit. 2006;8:519–522. doi: 10.1039/b600706f.
    1. Jagielo PJ, Thorne PS, Watt JL, Frees KL, Quinn TJ, Schwartz DA. Grain dust and endotoxin inhalation challenges produce similar inflammatory responses in normal subjects. Chest. 1996;110:263–270. doi: 10.1378/chest.110.1.263.
    1. Michel O, Duchateau J, Plat G, Cantinieaux B, Hotimsky A, Gerain J, Sergysels R. Blood inflammatory response to inhaled endotoxin in normal subjects. Clin Exp Allergy. 1995;25:73–79. doi: 10.1111/j.1365-2222.1995.tb01005.x.
    1. Kaneko Y, Takashima K, Suzuki N, Yamana K. Effects of theophylline on chronic inflammatory lung injury induced by LPS exposure in Guinea pigs. Allergol Int. 2007;56:445–456. doi: 10.2332/allergolint.O-07-490.
    1. Vernooy JH, Dentener MA, van Suylen RJ, Buurman WA, Wouters EF. Long-term intratracheal lipopolysaccharide exposure in mice results in chronic lung inflammation and persistent pathology. Am J Respir Cell Mol Biol. 2002;26:152–159.
    1. Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, Zhao Y, Goldsmith AM, Sonstein J, Linn MJ. et al.Elastase- and LPS-exposed mice display altered responses to rhinovirus infection. Am J Physiol Lung Cell Mol Physiol. 2009;297:L931–944. doi: 10.1152/ajplung.00150.2009.
    1. Kandaswami C, Middleton E Jr. Free radical scavenging and antioxidant activity of plant flavonoids. Adv Exp Med Biol. 1994;366:351–376.
    1. Robak J, Gryglewski RJ. Flavonoids are scavengers of superoxide anions. Biochem Pharmacol. 1988;37:837–841. doi: 10.1016/0006-2952(88)90169-4.
    1. Walker EH, Pacold ME, Perisic O, Stephens L, Hawkins PT, Wymann MP, Williams RL. Structural determinants of phosphoinositide 3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and staurosporine. Mol Cell. 2000;6:909–919. doi: 10.1016/S1097-2765(05)00089-4.
    1. Nanua S, Zick SM, Andrade JE, Sajjan US, Burgess JR, Lukacs NW, Hershenson MB. Quercetin blocks airway epithelial cell chemokine expression. Am J Respir Cell Mol Biol. 2006;35:602–610. doi: 10.1165/rcmb.2006-0149OC.
    1. Rogerio AP, Kanashiro A, Fontanari C, da Silva EV, Lucisano-Valim YM, Soares EG, Faccioli LH. Anti-inflammatory activity of quercetin and isoquercitrin in experimental murine allergic asthma. Inflamm Res. 2007;56:402–408. doi: 10.1007/s00011-007-7005-6.
    1. Hwang MK, Song NR, Kang NJ, Lee KW, Lee HJ. Activation of phosphatidylinositol 3-kinase is required for tumor necrosis factor-alpha-induced upregulation of matrix metalloproteinase-9: its direct inhibition by quercetin. Int J Biochem Cell Biol. 2009;41:1592–1600. doi: 10.1016/j.biocel.2009.01.014.
    1. Newcomb DC, Sajjan US, Nagarkar DR, Wang Q, Nanua S, Zhou Y, McHenry CL, Hennrick KT, Tsai WC, Bentley JK. et al.Human rhinovirus 1B exposure induces phosphatidylinositol 3-kinase-dependent airway inflammation in mice. Am J Respir Crit Care Med. 2008;177:1111–1121. doi: 10.1164/rccm.200708-1243OC.
    1. Mercer PF, Shute JK, Bhowmik A, Donaldson GC, Wedzicha JA, Warner JA. MMP-9, TIMP-1 and inflammatory cells in sputum from COPD patients during exacerbation. Respir Res. 2005;6:151. doi: 10.1186/1465-9921-6-151.
    1. Louhelainen N, Rytila P, Haahtela T, Kinnula VL, Djukanovic R. Persistence of oxidant and protease burden in the airways after smoking cessation. BMC Pulm Med. 2009;9:25. doi: 10.1186/1471-2466-9-25.
    1. Finlay GA, O'Driscoll LR, Russell KJ, D'Arcy EM, Masterson JB, FitzGerald MX, O'Connor CM. Matrix metalloproteinase expression and production by alveolar macrophages in emphysema. Am J Respir Crit Care Med. 1997;156:240–247.
    1. Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Becerril C, Selman M. Upregulation of gelatinases A and B, collagenases 1 and 2, and increased parenchymal cell death in COPD. Chest. 2000;117:684–694. doi: 10.1378/chest.117.3.684.
    1. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJ Jr, Chapman HA Jr, Shapiro SD, Elias JA. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest. 2000;106:1081–1093. doi: 10.1172/JCI10458.
    1. Hwang JW, Chung S, Sundar IK, Yao H, Arunachalam G, McBurney MW, Rahman I. Cigarette smoke-induced autophagy is regulated by SIRT1-PARP-1-dependent mechanism: Implication in pathogenesis of COPD. Aarch Biochem Biophy. 2010;500:203–209. doi: 10.1016/j.abb.2010.05.013.
    1. Tabak C, Arts IC, Smit HA, Heederik D, Kromhout D. Chronic obstructive pulmonary disease and intake of catechins, flavonols, and flavones: the MORGEN Study. Am J Respir Crit Care Med. 2001;164:61–64.
    1. Santus P, Sola A, Carlucci P, Fumagalli F, Di Gennaro A, Mondoni M, Carnini C, Centanni S, Sala A. Lipid peroxidation and 5-lipoxygenase activity in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2005;171:838–843. doi: 10.1164/rccm.200404-558OC.
    1. Walda IC, Tabak C, Smit HA, Rasanen L, Fidanza F, Menotti A, Nissinen A, Feskens EJ, Kromhout D. Diet and 20-year chronic obstructive pulmonary disease mortality in middle-aged men from three European countries. Eur J Clin Nutr. 2002;56:638–643. doi: 10.1038/sj.ejcn.1601370.
    1. Culpitt SV, Rogers DF, Fenwick PS, Shah P, De Matos C, Russell RE, Barnes PJ, Donnelly LE. Inhibition by red wine extract, resveratrol, of cytokine release by alveolar macrophages in COPD. Thorax. 2003;58:942–946. doi: 10.1136/thorax.58.11.942.
    1. Kode A, Rajendrasozhan S, Caito S, Yang SR, Megson IL, Rahman I. Resveratrol induces glutathione synthesis by activation of Nrf2 and protects against cigarette smoke-mediated oxidative stress in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2008;294:L478–488. doi: 10.1152/ajplung.00361.2007.
    1. Biswas SK, McClure D, Jimenez LA, Megson IL, Rahman I. Curcumin induces glutathione biosynthesis and inhibits NF-kappaB activation and interleukin-8 release in alveolar epithelial cells: mechanism of free radical scavenging activity. Antioxidants & redox signaling. 2005;7:32–41.
    1. Meja KK, Rajendrasozhan S, Adenuga D, Biswas SK, Sundar IK, Spooner G, Marwick JA, Chakravarty P, Fletcher D, Whittaker P. et al.Curcumin restores corticosteroid function in monocytes exposed to oxidants by maintaining HDAC2. Am J Respir Cell Mol Biol. 2008;39:312–323. doi: 10.1165/rcmb.2008-0012OC.
    1. Shishodia S, Potdar P, Gairola CG, Aggarwal BB. Curcumin (diferuloylmethane) down-regulates cigarette smoke-induced NF-kappaB activation through inhibition of IkappaBalpha kinase in human lung epithelial cells: correlation with suppression of COX-2, MMP-9 and cyclin D1. Carcinogenesis. 2003;24:1269–1279. doi: 10.1093/carcin/bgg078.
    1. Suzuki M, Betsuyaku T, Ito Y, Nagai K, Odajima N, Moriyama C, Nasuhara Y, Nishimura M. Curcumin attenuates elastase- and cigarette smoke-induced pulmonary emphysema in mice. Am J Physiol Lung Cell Mol Physiol. 2009;296:L614–623. doi: 10.1152/ajplung.90443.2008.
    1. Kasielski M, Nowak D. Long-term administration of N-acetylcysteine decreases hydrogen peroxide exhalation in subjects with chronic obstructive pulmonary disease. Respir Med. 2001;95:448–456. doi: 10.1053/rmed.2001.1066.
    1. Maestrelli P, Paska C, Saetta M, Turato G, Nowicki Y, Monti S, Formichi B, Miniati M, Fabbri LM. Decreased haem oxygenase-1 and increased inducible nitric oxide synthase in the lung of severe COPD patients. Eur Respir J. 2003;21:971–976. doi: 10.1183/09031936.03.00098203.
    1. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase in reactive nitrogen species production in chronic obstructive pulmonary disease airways. Am J Respir Crit Care Med. 2000;162:701–706.
    1. Shinohara T, Kaneko T, Nagashima Y, Ueda A, Tagawa A, Ishigatsubo Y. Adenovirus-mediated transfer and overexpression of heme oxygenase 1 cDNA in lungs attenuates elastase-induced pulmonary emphysema in mice. Hum Gene Ther. 2005;16:318–327. doi: 10.1089/hum.2005.16.318.
    1. Chen JC, Ho FM, Pei-Dawn Lee C, Chen CP, Jeng KC, Hsu HB, Lee ST, Wen Tung W, Lin WW. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IkappaB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur J Pharmacol. 2005;521:9–20. doi: 10.1016/j.ejphar.2005.08.005.
    1. Comalada M, Ballester I, Bailon E, Sierra S, Xaus J, Galvez J, de Medina FS, Zarzuelo A. Inhibition of pro-inflammatory markers in primary bone marrow-derived mouse macrophages by naturally occurring flavonoids: analysis of the structure-activity relationship. Biochem Pharmacol. 2006;72:1010–1021. doi: 10.1016/j.bcp.2006.07.016.
    1. Mu MM, Chakravortty D, Sugiyama T, Koide N, Takahashi K, Mori I, Yoshida T, Yokochi T. The inhibitory action of quercetin on lipopolysaccharide-induced nitric oxide production in RAW 264.7 macrophage cells. J Endotoxin Res. 2001;7:431–438. doi: 10.1179/096805101101533034.
    1. Minamoto K, Harada H, Lama VN, Fedarau MA, Pinsky DJ. Reciprocal regulation of airway rejection by the inducible gas-forming enzymes heme oxygenase and nitric oxide synthase. J Exp Med. 2005;202:283–294. doi: 10.1084/jem.20050377.
    1. Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105. doi: 10.1042/0264-6021:3510095.
    1. Agullo G, Gamet-Payrastre L, Manenti S, Viala C, Remesy C, Chap H, Payrastre B. Relationship between flavonoid structure and inhibition of phosphatidylinositol 3-kinase: A comparison with tyrosine kinase and protein kinase C inhibition. Biochem Pharmocol. 1997;53:1649–1657. doi: 10.1016/S0006-2952(97)82453-7.
    1. Huang Y-T, Hwang J-J, Lee P-P, Ke F-C, Huang J-H, Huang C-J, Kandaswami C, Middleton E Jr, Lee M-T. Effects of luteolin and quercetin, inhibitors of tyrosine kinase, on cell growth and metastasis-associated properties in A431 cells overexpressing epidermal growth factor receptor. Br J Pharmacol. 1999;128:999–1010. doi: 10.1038/sj.bjp.0702879.
    1. Peet GW, Li J. IkappaB kinases alpha and beta show a random sequential kinetic mechanism and are inhibited by staurosporine and quercetin. J Biol Chem. 1999;274:32655–32661. doi: 10.1074/jbc.274.46.32655.
    1. Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002) J Biol Chem. 1994;269:5241–5248.
    1. Babusyte A, Stravinskaite K, Jeroch J, Lotvall J, Sakalauskas R, Sitkauskiene B. Patterns of airway inflammation and MMP-12 expression in smokers and ex-smokers with COPD. Respir Res. 2007;8:81. doi: 10.1186/1465-9921-8-81.
    1. Demedts IK, Morel-Montero A, Lebecque S, Pacheco Y, Cataldo D, Joos GF, Pauwels RA, Brusselle GG. Elevated MMP-12 protein levels in induced sputum from patients with COPD. Thorax. 2006;61:196–201. doi: 10.1136/thx.2005.042432.
    1. Ohnishi K, Takagi M, Kurokawa Y, Satomi S, Konttinen YT. Matrix metalloproteinase-mediated extracellular matrix protein degradation in human pulmonary emphysema. Lab Invest. 1998;78:1077–1087.
    1. Deshmukh HS, McLachlan A, Atkinson JJ, Hardie WD, Korfhagen TR, Dietsch M, Liu Y, Di PY, Wesselkamper SC, Borchers MT. et al.Matrix metalloproteinase-14 mediates a phenotypic shift in the airways to increase mucin production. Am J Respir Crit Care Med. 2009;180:834–845. doi: 10.1164/rccm.200903-0328OC.
    1. Deshmukh HS, Shaver C, Case LM, Dietsch M, Wesselkamper SC, Hardie WD, Korfhagen TR, Corradi M, Nadel JA, Borchers MT. et al.Acrolein-activated matrix metalloproteinase 9 contributes to persistent mucin production. Am J Respir Cell Mol Biol. 2008;38:446–454. doi: 10.1165/rcmb.2006-0339OC.
    1. Park HJ, Lee CM, Jung ID, Lee JS, Jeong YI, Chang JH, Chun SH, Kim MJ, Choi IW, Ahn SC. et al.Quercetin regulates Th1/Th2 balance in a murine model of asthma. Internat Immunopharmacol. 2009;9:261–267. doi: 10.1016/j.intimp.2008.10.021.
    1. Conquer JA, Maiani G, Azzini E, Raguzzini A, Holub BJ. Supplementation with quercetin markedly increases plasma quercetin concentration without effect on selected risk factors for heart disease in healthy subjects. J Nutr. 1998;128:593–597.
    1. Hollman PC, van Trijp JM, Mengelers MJ, de Vries JH, Katan MB. Bioavailability of the dietary antioxidant flavonol quercetin in man. Cancer Lett. 1997;114:139–140. doi: 10.1016/S0304-3835(97)04644-2.
    1. Harwood M, Danielewska-Nikiel B, Borzelleca JF, Flamm GW, Williams GM, Lines TC. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem Toxicol. 2007;45:2179–2205. doi: 10.1016/j.fct.2007.05.015.
    1. Boots AW, Li H, Schins RP, Duffin R, Heemskerk JW, Bast A, Haenen GR. The quercetin paradox. Toxicol Appl Pharmacol. 2007;222:89–96. doi: 10.1016/j.taap.2007.04.004.
    1. Biasutto L, Sassi N, Mattarei A, Marotta E, Cattelan P, Toninello A, Garbisa S, Zoratti M, Paradisi C. Impact of mitochondriotropic quercetin derivatives on mitochondria. Biochim Biophys Acta. 2010;1797:189–196. doi: 10.1016/j.bbabio.2009.10.001.
    1. Papi A, Bellettato CM, Braccioni F, Romagnoli M, Casolari P, Caramori G, Fabbri LM, Johnston SL. Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med. 2006;173:1114–1121. doi: 10.1164/rccm.200506-859OC.
    1. Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177:861–870. doi: 10.1164/rccm.200708-1269OC.
    1. Mahadeva R, Shapiro SD. Chronic obstructive pulmonary disease 3: Experimental animal models of pulmonary emphysema. Thorax. 2002;57:908–914. doi: 10.1136/thorax.57.10.908.

Source: PubMed

3
Subscribe