NLRP3 inflammasome activation by mitochondrial ROS in bronchial epithelial cells is required for allergic inflammation

S R Kim, D I Kim, S H Kim, H Lee, K S Lee, S H Cho, Y C Lee, S R Kim, D I Kim, S H Kim, H Lee, K S Lee, S H Cho, Y C Lee

Abstract

Abnormality in mitochondria has been suggested to be associated with development of allergic airway disorders. In this study, to evaluate the relationship between mitochondrial reactive oxygen species (ROS) and NLRP3 inflammasome activation in allergic asthma, we used a newly developed mitochondrial ROS inhibitor, NecroX-5. NecroX-5 reduced the increase of mitochondrial ROS generation in airway inflammatory cells, as well as bronchial epithelial cells, NLRP3 inflammasome activation, the nuclear translocation of nuclear factor-κB, increased expression of various inflammatory mediators and pathophysiological features of allergic asthma in mice. Finally, blockade of IL-1β substantially reduced airway inflammation and hyperresponsiveness in the asthmatic mice. These findings suggest that mitochondrial ROS have a critical role in the pathogenesis of allergic airway inflammation through the modulation of NLRP3 inflammasome activation, providing a novel role of airway epithelial cells expressing NLRP3 inflammasome as an immune responder.

Figures

Figure 1
Figure 1
Generation of Intracellular ROS and mitochondrial ROS in BAL cells and primary cultured tracheal epithelial cells from OVALPS-OVA mice. BAL cells were obtained at 48 h after the challenge in SAL-SAL mice administered with drug vehicle (SV), OVALPS-OVA mice administered with drug vehicle (OLV), OVALPS-OVA mice administered with 3 mg/kg Necrox-5 (OLN 3) or OVALPS-OVA mice administered with 30 mg/kg Necrox-5 (OLN 30). The experiment was performed with the primary cultured tracheal epithelial cells from SAL-SAL mice without any treatment (Control), OVALPS-OVA mice with no treatment (OL), or OVALPS-OVA mice at 90 min after the treatment with drug vehicle (OLV) or 10 μM Necrox-5 (OLN 10). Representative confocal laser immunofluorescence photomicrography of BAL cells (a) and primary cultured tracheal epithelial cells (e) showed the localization of mitochondrial ROS (the center panels, red fluorescence views) in the cells. The blue fluorescent DAPI stain was used for nuclear localization. The left and the right panels presented phase contrast views and the merger views, respectively. Bars indicate scale of 10 μm (a) or 20 μm (e). Histogram analysis for ROS generation in BAL cells (b) and primary cultured tracheal epithelial cells (f). (c) DCF fluorescence intensity of intracelluar ROS in BAL cells. (d) MitoTracker fluorescence intensity of mitochondrial ROS in BAL cells. The ROS levels are presented as the relative ratio of values in experimental groups to those in SV. The relative ratio of ROS levels in the BAL cells of SV arbitrarily presented as 100. Bars represent mean±S.E.M. from seven mice per group. #P<0.05 versus SV or control; *P<0.05 versus OLV
Figure 2
Figure 2
Effects of NecroX-5 on the integrity and content of mtDNA in lung tissues of OVALPS-OVA mice. Sampling was performed at 48 h after the last challenge in SV, OLV, OLN 3 or OLN 30. (a) Representative PCR analyses for both a long (8642-bp) mtDNA fragment and a short (316-bp) mtDNA fragment from the lung tissues of OVALPS-OVA mice. (b) The long fragment/short mtDNA fragment hybridization ratio in the lung tissues of OVALPS-OVA mice. The ratio of SV is arbitrarily presented as 1. (c) Changes of the content of mtDNA in the lung tissues of OVALPS-OVA mice. The levels of mtDNA were presented as ng/1 μg of protein/1 g of lung tissues. Data represent mean±S.E.M. from six mice per group. #P<0.05 versus SV; *P<0.05 versus OLV
Figure 3
Figure 3
Effects of mitochondrial ROS inhibitor, Necrox-5 on the protein levels of NLRP3, caspase-1 and IL-1β in primary cultured tracheal epithelial cells and lung tissues of OVALPS-OVA mice. Representative western blot analysis of NLRP3 (a), caspase-1 (b) and IL-1β (c) in primary cultured tracheal epithelial cells of OVALPS-OVA mice. Representative western blots of NLRP3 (d), caspase-1 (e) and IL-1β (f) in lung tissues of OVALPS-OVA mice. Densitometric analyses of the bands on films are presented as the relative ratio of NLRP3 (g), caspase-1 (h) or IL-1β (i) to actin. The relative ratio of each protein in the lung tissues of SV is arbitrarily presented as 1. Representative western blots of NLRP3 (j) and caspase-1 (k) in human BAL fluids from asthmatic patients and healthy subjects (n=3 per group). (l) Enzyme immunoassay of IL-1β in BAL fluids of OVALPS-OVA mice. Bars represent mean±S.E.M. from seven mice per group. #P<0.05 versus SV; *P<0.05 versus OLV. (m) Representative confocal laser immunofluorescence photomicrography of BAL cells of OVALPS-OVA mice showed the expression of IL-1β. The blue fluorescent DAPI stain was used for nuclear localization. The left and the right panels presented phase contrast views and the merger views, respectively. Bars indicate scale of 20 μm
Figure 4
Figure 4
Effects of Necrox-5 on total and differential cell counts in BAL fluids, histological changes, MPO activity, airway hyperresponsiveness and serum levels of OVA-specific immunoglobulins in OVALPS-OVA mice. All parameters were measured at 48 h after the last challenge in SV, OLV, OLN 3 and OLN 30. (a) Cellular changes in BAL fluids of OVALPS-OVA mice. Bars represent mean±S.E.M. from seven mice per group. (be) Representative H&E-stained sections of the lungs isolated from SV (b), OLV (c), OLN 3 (d) and OLN 30 (e). Bars indicate scale of 20 μm. (f) MPO activity in lung tissue homogenates of OVALPS-OVA mice. Airway responsiveness in OVALPS-OVA mice was assessed by both noninvasive (g, %Penh) and invasive (h, Rrs) measurements. (ik) Changes of the serum levels of OVA-specific IgE (i), OVA-specific IgG1 (j) and OVA-specific IgG2a (k). Bars represent mean±S.E.M. from six or seven mice per group. ND, not detected, #P<0.05 versus SV; *P<0.05 versus OLV
Figure 5
Figure 5
Effects of NecroX-5 on the protein levels of various inflammatory mediators and the activation of NF-κB/IκBα in lung tissues of OVALPS-OVA mice. Sampling was performed at 48 h after the last challenge in SV, OLV, OLN 3 and OLN 30. Representative western blots of IL-4 (a), IL-5 (c), IL-13 (e), TNF-α (g), IFN-γ (i), IL-17 (k) and KC (m) in lung tissues. (b, d, f, h, j, l, n) Densitometric analysis of the bands on films is presented as the relative ratio of each protein to actin. (o) Representative western blot of NF-κB p65 in lung tissues. (p) Densitometric analyses of the bands on films are presented as the relative ratio of NF-κB p65 level in OLV, OLN 3 or OLN 30 to the level in SV. The relative ratio of each protein in the lung tissues of SV is arbitrarily presented as 1. Bars represent mean±S.E.M. from seven mice per group. #P<0.05 versus SV; *P<0.05 versus OLV
Figure 6
Figure 6
Effects of NecroX-5 on total and differential cell counts in BAL fluids, histological changes, airway hyperresponsiveness, protein levels of NLRP3, IL-1β and caspase-1, and generation of mitochondrial ROS in the lung of HDM-instilled mice. All parameters were measured at 48 h after the last challenge in saline-instilled mice administered drug vehicle (SV), HDM-instilled mice administered drug vehicle (HV), HDM-instilled mice administered 3 mg/kg of NecroX-5 (HN 3) and HDM-instilled mice administered 30 mg/kg of NecroX-5 (HN 30). (a) Cellular changes in BAL fluids of HDM-instilled mice. Bars represent mean±S.E.M. from six mice per group. (be) Representative H&E-stained sections of the lungs isolated from SV (b), HV (c), HN 3 (d) and HN 30 (e). Bars indicate scale of 20 μm. (f) Airway responsiveness in HDM-instilled mice was assessed by invasive measurement (Rrs). (gl) Representative western blots for NLRP3 (g), caspase-1 (i) and IL-1β (k) in lung tissues and densitometric analysis. Bars represent mean±S.E.M. from six mice per group. (m) Representative confocal laser immunofluorescence photomicrography of BAL cells showed the localization of mitochondrial ROS (red fluorescence views) in the cells. The blue fluorescent DAPI stain was used for nuclear localization. The left and the right panels presented phase contrast views and the merger views, respectively. Bars indicate scale of 20 μm. #P<0.05 versus SV; *P<0.05 versus HV
Figure 7
Figure 7
Effects of blockade of IL-1β on total and differential cell counts in BAL fluids, histological changes and airway hyperresponsiveness in the lung of OVALPS-OVA mice and OVALPS-OVA IL-1R KO mice. All parameters were measured at 48 h after the last challenge in SAL-SAL mice administered with PBS (SP), OVALPS-OVA mice administered with isotype control antibody (OLC-Ab), OVALPS-OVA mice administered with an anti-IL-1β-neutralizing antibody (OLA-IL-1β Ab), SAL-SAL WT mice (WT CON), OVALPS-OVA WT mice (WT OL), SAL-SAL IL-1R KO mice (IL-1R KO CON) and OVALPS-OVA IL-1R KO mice (IL-1R KO OL). (a) Cellular changes in BAL fluids of OVALPS-OVA mice. Bars represent mean±S.E.M. from seven mice per group. (b-d) Representative H&E-stained sections of the lungs isolated from SP (b), OLC-Ab (c) and OLA-IL-1β Ab (d). Bars indicate scale of 20 μm. (e and f) Airway responsiveness in OVALPS-OVA mice was assessed by both noninvasive (e, %Penh) and invasive (f, Rrs) measurements. Bars represent mean±S.E.M. from seven mice per group. (g) Cellular changes in BAL fluids of IL-1R KO or WT mice. Bars represent mean±S.E.M. from five mice per group. (hk) Representative H&E-stained sections of the lungs isolated from WT CON (h), WT OL (i), IL-1R KO CON (j) and IL-1R KO OL (k). Bars indicate scale of 50 μm. (l) Airway responsiveness in IL-1R KO or WT mice was assessed by invasive measurement (Rrs). Bars represent mean±S.E.M. from five mice per group. #P<0.05 versus SP or WT CON; *P<0.05 versus OLC-Ab or WT OL
Figure 8
Figure 8
Effects of blockade of IL-1β on the protein levels of various inflammatory mediators in lung tissues of OVALPS-OVA mice and OVALPS-OVA IL-1R KO mice. Sampling was performed at 48 h after the last challenge in SP, OLC-Ab, OLA-IL-1β Ab, WT CON, WT OL, IL-1R KO CON,and IL-1R KO OL. Representative western blots of IL-4 (a and o), IL-5 (c and q), IL-13 (e and s), TNF-α (g), IFN-γ (i), IL-17 (k and u) and KC (m and w) in lung tissues. (b, d, f, h, j, l, n, p, r, t, v, x) Densitometric analysis of the bands on films is presented as the relative ratio of each protein to actin. The relative ratio of each protein in the lung tissues of SP or WT CON is arbitrarily presented as 1. Bars represent mean±S.E.M. from five or seven mice per group. #P<0.05 versus SP or WT CON; *P<0.05 versus OLC-Ab or WT OL
Figure 9
Figure 9
Structure of NecroX-5

References

    1. 1Kim 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.
    1. 2Riedl MA, Nel AE. Importance of oxidative stress in the pathogenesis and treatment of asthma. Curr Opin Allergy Clin Immunol 2008; 8: 49–56.
    1. 3Ciencewicki J, Trivedi S, Kleeberger SR. Oxidants and the pathogenesis of lung diseases. J Allergy Clin Immunol 2008; 122: 456–468.
    1. 4Barnes PJ. Reactive oxygen species in asthma. Eur Respir Rev 2000; 10: 240–243.
    1. 5Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 2004; 4: 181–189.
    1. 6Emelyanov VV. Mitochondrial connection to the origin of the eukaryotic cell. Eur J Biochem 2003; 270: 1599–1618.
    1. 7Cloonan SM, Choi AM. Mitochondria: commanders of innate immunity and disease? Curr Opin Immunol 2012; 24: 32–40.
    1. 8Hopps E, Noto D, Caimi G, Averna MR. A novel component of the metabolic syndrome: the oxidative stress. Nutr Metab Cardiovasc Dis 2010; 20: 72–77.
    1. 9Sullivan PG, Bueler H. Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1-deficient mice. PLoS One 2011; 6: e16038.
    1. 10d'Avila JC, Santiago AP, Amâncio RT, Galina A, Oliveira MF, Bozza FA. Sepsis induces brain mitochondrial dysfunction. Crit Care Med 2008; 36: 1925–1932.
    1. 11Heinzmann A, Thoma C, Dietrich H, Deichmann KA. Identification of common polymorphisms in the mitochondrial genome. Allergy 2003; 58: 830–831.
    1. 12Raby BA, Klanderman B, Murphy A, Mazza S, Camargo CA Jr, Silverman EK et al. A common mitochondrial haplogroup is associated with elevated total serum IgE levels. J Allergy Clin Immunol 2007; 120: 351–358.
    1. 13Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 2011; 12: 222–230.
    1. 14Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011; 469: 221–225.
    1. 15Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008; 320: 674–677.
    1. 16Petrilli V, Papin S, Dostert C, Mayor A, Martinon F, Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ 2007; 14: 1583–1589.
    1. 17Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006; 440: 237–241.
    1. 18Besnard AG, Guillou N, Tschopp J, Erard F, Couillin I, Iwakura Y et al. NLRP3 inflammasome is required in murine asthma in the absence of aluminum adjuvant. Allergy 2011; 66: 1047–1057.
    1. 19Kool M, Pétrilli V, De Smedt T, Rolaz A, Hammad H, van Nimwegen M et al. Cutting edge: alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J Immunol 2008; 181: 3755–3759.
    1. 20Gregory LG, Lloyd CM. Orchestrating house dust mite-associated allergy in the lung. Trends Immunol 2011; 32: 402–411.
    1. 21Lamblin C, Gosset P, Tillie-Leblond I, Saulnier F, Marquette CH, Wallaert B et al. Bronchial neutrophilia in patients with noninfectious status asthmaticus. Am J Respir Crit Care Med 1998; 157: 394–402.
    1. 22Thu VT, Kim HK, Long le T, Lee SR, Hanh TM, Ko TH et al. NecroX-5 prevents hypoxia/reoxygenation injury by inhibiting the mitochondrial calcium uniporter. Cardiovasc Res 2012; 94: 342–350.
    1. 23Dashdorj A, Jyothi KR, Lim S, Jo A, Nguyen MN, Ha J et al. Mitochondria-targeted antioxidant MitoQ ameliorates experimental mouse colitis by suppressing NLRP3 inflammasome-mediated inflammatory cytokines. BMC Med 2013; 11: 178.
    1. 24Roca FJ, Ramakrishnan L. TNF dually mediates resistance and susceptibility to mycobacteria via mitochondrial reactive oxygen species. Cell 2013; 153: 521–534.
    1. 25Mansouri A, Gaou I, De Kerguenec C, Amsellem S, Haouzi D, Berson A et al. An alcoholic binge causes massive degradation of hepatic mitochondrial DNA in mice. Gastroenterology 1999; 117: 181–190.
    1. 26Teramoto S, Shu CY, Ouchi Y, Fukuchi Y. Increased spontaneous production and generation of superoxide anion by blood neutrophils in patients with asthma. J Asthma 1996; 33: 149–155.
    1. 27Rochelle LG, Fischer BM, Adler KB. Concurrent production of reactive oxygen and nitrogen species by airway epithelial cells in vitro. Free Radic Biol Med 1998; 24: 863–868.
    1. 28Chihara J, Hayashi N, Kakazu T, Yamamoto T, Kurachi D, Nakajima S. RANTES augments radical oxygen products from eosinophils. Int Arch Allergy Immunol 1994; 104: 52–53.
    1. 29Tenscher K, Metzner B, Schopf E, Norgauer J, Czech W. Recombinant human eotaxin induces oxygen radical production, up-regulation in human eosinophils via a pertussis toxin sensitive heterotrimeric guanine nucleotide-binding protein. Blood 1996; 88: 3195–3199.
    1. 30Nagata M, Sedgwick JB, Vrtis R, Busse WW. Endothelial cells upregulate eosinophil superoxide generation via VCAM-1 expression. Clin Exp Allergy 1998; 29: 550–561.
    1. 31Lee YC, Lee KS, Park SJ, Park HS, Lim JS, Park KH et al. Blockade of airway hyperresponsiveness and inflammation in a murine model of asthma by a prodrug of cysteine, L-2-oxothiazolidine-4-carboxylic acid. FASEB J 2004; 18: 1917–1919.
    1. 32Park HS, Kim SR, Lee YC. Impact of oxidative stress on lung diseases. Respirology 2009; 14: 27–38.
    1. 33Kagan VE, Serbinova EA, Stoyanovsky DA, Khwaja S, Packer L. Assay of ubiquinones and ubiquinols as antioxidants. Methods Enzymol 1994; 234: 343–354.
    1. 34Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN et al. Emerging role of damageassociated molecular patterns derived from mitochondria in inflammation. Trends Immunol 2011; 32: 157–164.
    1. 35Davis BK, Wen H, Ting JP. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol 2011; 29: 707–735.
    1. 36Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 2009; 458: 514–518.
    1. 37Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science 2009; 323: 1057–1060.
    1. 38Cruz CM, Rinna A, Forman HJ, Ventura AL, Persechini PM, Ojcius DM. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem 2007; 282: 2871–2879.
    1. 39Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 2008; 9: 847–856.
    1. 40Meissner F, Molawi K, Zychlinsky A. Superoxide dismutase 1 regulates caspase-1 and endotoxic shock. Nat Immunol 2008; 9: 866–872.
    1. 41Schumacker PT, Gillespie MN, Nakahira K, Choi AM, Crouser ED, Piantadosi CA et al. Mitochondria in lung biology and pathology: more than just a powerhouse. Am J Physiol Lung Cell Mol Physiol 2014; 306: L962–L974.
    1. 42Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 2011; 12: 222–230.
    1. 43Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 2009; 27: 519–550.
    1. 44Hultner L, Kölsch S, Stassen M, Kaspers U, Kremer JP, Mailhammer R et al. In activated mast cells, IL-1 up-regulates the production of several Th2-related cytokines including IL-9. J Immunol 2000; 164: 5556–5563.
    1. 45Broide DH, Lotz M, Cuomo AJ, Coburn DA, Federman EC, Wasserman SI. Cytokines in symptomatic asthma airways. J Allergy Clin Immunol 1992; 8: 958–967.
    1. 46Hirota JA, Hirota SA, Warner SM, Stefanowicz D, Shaheen F, Beck PL et al. The airway epithelium nucleotide-binding domain and leucine-rich repeat protein 3 inflammasome is activated by urban particulate matter. J Allergy Clin Immunol 2012; 129: 1116–1125.
    1. 47Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997; 336: 1066–1071.
    1. 48Henderson WR Jr, Chi EY, Teo JL, Nguyen C, Kahn M. A small molecule inhibitor of redox-regulated NF-kappa B and activator protein-1 transcription blocks allergic airway inflammation in a mouse asthma model. J Immunol 2002; 169: 5294–5299.
    1. 49Rahman I, MacNee W. Role of transcription factors in inflammatory lung diseases. Thorax 1998; 53: 601–612.
    1. 50Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science 1995; 270: 296–299.
    1. 51Cho JY, Miller M, Baek KJ, Han JW, Nayar J, Lee SY et al. Inhibition of airway remodeling in IL-5-deficient mice. J Clin Invest 2004; 113: 551–560.
    1. 52Kwak 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.
    1. 53Hamelmann E, Schwarze J, Takeda K, Oshiba A, Larsen GL, Irvin CG et al. Noninvasive measurement of airway responsiveness in allergic mice using barometric plethysmography. Am J Respir Crit Care Med 1997; 156: 766–775.
    1. 54Takeda K, Hamelmann E, Joetham A, Shultz LD, Larsen GL, Irvin CG et al. Development of eosinophilic airway inflammation and airway hyperresponsiveness in mast cell-deficient mice. J Exp Med 1997; 186: 449–454.

Source: PubMed

3
Abonnieren