Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity

Hye Young Kim, Hyun Jun Lee, Ya-Jen Chang, Muriel Pichavant, Stephanie A Shore, Katherine A Fitzgerald, Yoichiro Iwakura, Elliot Israel, Kenneth Bolger, John Faul, Rosemarie H DeKruyff, Dale T Umetsu, Hye Young Kim, Hyun Jun Lee, Ya-Jen Chang, Muriel Pichavant, Stephanie A Shore, Katherine A Fitzgerald, Yoichiro Iwakura, Elliot Israel, Kenneth Bolger, John Faul, Rosemarie H DeKruyff, Dale T Umetsu

Abstract

Obesity is associated with the development of asthma, which is often difficult to control. To understand the immunological pathways that lead to obesity-associated asthma, we fed mice a high-fat diet for 12 weeks, which resulted in obesity and the development of airway hyperreactivity (AHR), a cardinal feature of asthma. This AHR was independent of adaptive immunity, as it occurred in obese Rag1(-/-) mice, which lack B and T cells, and was dependent on interleukin-17A (IL-17A) and the NLRP3 inflammasome, as it did not develop in obese Il17a(-/-) or Nlrp3(-/-) mice. AHR was also associated with the expansion of CCR6(+) type 3 innate lymphoid cells (ILCs) producing IL-17A (ILC3 cells) in the lung, which could by themselves mediate AHR when adoptively transferred into Rag2(-/-); Il2rg(-/-) mice treated with recombinant IL-1β. Macrophage-derived IL-1β production was induced by HFD and expanded the number of lung ILC3 cells. Blockade of IL-1β with an IL-1 receptor antagonist abolished obesity-induced AHR and reduced the number of ILC3 cells. As we found ILC3-like cells in the bronchoalveolar lavage fluid of individuals with asthma, we suggest that obesity-associated asthma is facilitated by inflammation mediated by NLRP3, IL-1β and ILC3 cells.

Figures

Figure 1. High fat diet feeding induces…
Figure 1. High fat diet feeding induces AHR in wildtype mice
(a–c). Wild type mice fed normal chow or 60% HFD, starting at weaning for 12–14 weeks. a. Comparison of bodyweight gain in wild-type C57BL/6 mice on normal chow or the HFD. ***p<0.001, HFD group compared to chow fed group. (Student’s t-test) b. Representative picture of mice and organs after chow or HFD feeding. Scale bar indicates 5mm c. Wild-type mice develop AHR, as assessed by the response to increasing doses of methacholine. Results represent the changes in lung resistance (RL) as a measure of AHR. **p<0.01, and ***p<0.001, compared to chow fed group. (Two-way ANOVA). Data are representative of at least three independent experiments. Error bars represent SEM. d. 7 wks old ob/ob mice fed on normal chow diet develop AHR. Results represent the changes in lung resistance (RL) as a measure of AHR. *p<0.05, and **p<0.01, compared to wild type mice. (Two-way ANOVA).
Figure 2. HFD induced AHR requires the…
Figure 2. HFD induced AHR requires the presence of IL-17A
a. Cells were isolated from lungs of mice fed on normal chow or on the HFD, and supernatants were analyzed for IL-17A by ELISA. b. Comparison of bodyweight gain between WT and Il17−/− mice on either chow or HFD. NS, WT mice on HFD vsIl17−/− mice on HFD (Student’s t-test). c. AHR was measured using mice represented in Fig. 2b. Graph represents the changes in lung resistance (RL). *p<0.05, and **p<0.01, Il17−/− mice on HFD compared to WT mice on HFD (Two-way ANOVA). d. Total lung cells from mice fed normal chow or HFD were sorted for CD4+ or CD4− cell fraction and analyzed for Il17A mRNA expression by qRT-PCR. ***p<0.001 (chow CD4− vs HFD CD4− fraction), and *p<0.05 (chow CD4+ vs HFD CD4+ fraction), (Student’s t-test). e. ILC3 cells (CD45+Lineage−IL-17A+cells) from HFD fed WT mice (upper panel) and normal chow fed ob/ob mice (lower panel). Graph (right) represents total number of ILC3 cells in the lungs (upper panel; wild type chow vs HFD, ***p<0.001, lower panel; chow fed wild type vsob/ob, **p<0.01). f. ILC3 cells in Figure 2e were further characterized. Grey histograms represent isolype control staining. g. The changes in lung resistance (RL) as a measure of AHR in Rag1−/− mice fed a high fat diet. *p<0.05, and **p<0.01, comparing normal chow fed and high diet fed groups (Two-way ANOVA). h. ILC3 cells in the lungs of Rag1−/− mice. Graph (right) represents total number of ILC3 cells (chow vs HFD groups, ***p<0.001).
Figure 3. IL-1β production and M1 macrophages…
Figure 3. IL-1β production and M1 macrophages are increased in the lungs of obese mice
(a–f). Total RNA was extracted from the lung and adipose tissue for qRT-PCR. Fold induction of Il1b (a,d), Il6 (b,e), and Il23 (c,f) were calculated based on GAPDH expression. *p<0.05, and **p<0.01, mRNA expression from HFD group was compared to chow group. g. Expression of M1/M2 macrophage markers, CD206 (M2) and CD11c (M1), were analyzed after gating on of CD45+F4/80+ cells. Graph represents the total numbers of macrophages in the lung (left, *p<0.05 and ***p<0001) and adipose tissue (right, ***p<0.001). (Student’s t-test) h. IL-1β production from M1/M2 macrophages. Cells were taken from lung or adipose tissue and IL-1β producing M1 (CD11c+, left panel) or M2 (CD206+, right panel) macrophages were assessed by flow cytometry.
Figure 4. HFD increases NLRP3, which is…
Figure 4. HFD increases NLRP3, which is required for AHR
a. Total RNA was extracted from the lung, liver and adipose tissue and assessed for Nlrp3. Fold induction of Nlrp3 mRNA was calculated based on GAPDH expression. **p<0.01 and ***p<0.001, mRNA level from mice fed on HFD compared to chow group. (Student’s t-test) b. Graph represent grams of weight gain in WT or Nlrp3−/− mice. **p<0.01, WT mice on HFD compared to Nlrp3−/− mice on HFD group. (Student’s t-test) c. WT and Nlrp3−/− mice 3 months after HFD (left). Representative pictures of lung, liver and epididymal adipose tissue from Nlrp3−/− mice fed on chow or HFD (right). Scale bar indicates 5mm d. Development of obesity induced AHR was compared in WT and Nlrp3−/− mice. Graph represents the changes in lung resistance (RL). *p<0.05, Nlrp3−/− mice on HFD were compared to WT mice on HFD group (Two-way ANOVA). e. IL-1β levels in the culture supernatant of lung cells from WT and Nlrp3−/− mice. The data shown are means ± SEM. **p<0.01, supernatants from HFD group were compared to chow group. (Student’s t-test) f. Lungs were taken from chow or HFD fed mice and stimulated with PMA/ionomycin for 5 hr. −IL-17A+ ILC3 cells in WT mice (upper panel) or Nlrp3−/− mice (lower panel) were assessed by FACS. Graph represents the total number of ILC3 cells in the lung. ***p<0.001, ILC3 cells in WT mice were compared to Nlrp3−/− mice. (Student’s t-test).
Figure 5. IL-17 producing innate lymphoid cells…
Figure 5. IL-17 producing innate lymphoid cells are required for the development of AHR
Graphs represent the changes in lung resistance (RL) (upper panels) and BAL fluid cell counts (lower panels) after rIL-1β (a) or rIL-1α (b) treatment. For AHR, **p<0.05, and ***p<0.001, rIL-1β or rIL-1α treated mice were compared to saline treated mice (Two-way ANOVA). For BAL fluid, **p<0.01, and ***p<0.001, rIL-1β vs saline (a), **p<0.01, rIL-1α vs saline (b) (Student’s t-test). c. Changes in lung resistance (RL) (upper panel) and BAL fluid cell counts (lower panel) after depletion of ILCs. For AHR, **p<0.01, rIL-1β treated mice were compared to anti-Thy1.2 mAb treated mice (Two way ANOVA). For BAL fluid, **p<0.01, and ***p<0.001, rIL-1β vs anti-Thy1.2 mAb+rIL-1β (Student’s t-test). d. ILC3 cells in the lung (left panel), and total number of ILC3 cells in each group (right panel). **p<0.01, rIL-1β treated mice were compared to anti-Thy1.2 mAb treated mice (Student’s t-test). e. Changes in lung resistance (RL) in Rag2−/−Il2rγ−/− mice after adoptive transfer of ILC3 cells (Lin−CCR6+). ***p<0.001, comparing Rag2−/−Il2rγ−/− mice treated with rIL-1β receiving or not receiving ILC3 cells (Two-way ANOVA). Cells in BAL fluid (right panel). *p<0.05, and ***p<0.001, ILC3 cells +rIL-1β vs rIL-1β group (Student’s t-test). f. IL-17A and IL-13 production from adoptively transferred ILC3 cells. g. Changes in lung resistance (RL) in Il4/Il13−/− and Il17−/− mice after rIL-1β treatment. *p<0.05, and **p<0.01, rIL-1β treated Il4/Il13−/− mice were compared to rIL-1β treated Il17−/− mice (Two way- ANOVA). Cells in BAL fluid (right panel). ** p<0.01, and ***p<0.001, rIL-1β treated Il4/Il13−/− mice were compared to rIL-1β treated Il17−/− mice (Student’s t-test). h. IL-1R expression on ILC3 cells. Lin−IL-17+ cells (red) express higher level of IL-1R compared to Lin−IL-17− cells (blue). Shaded histogram: isotype control. i. Cells were taken from rIL-1β treated Rag2−/− mice then cultured in vitro to induce ILC3 cells.
Figure 6. Treatment of obese mice with…
Figure 6. Treatment of obese mice with anti-IL-1, prevents AHR
a. Schedule of feeding and anti-IL1R antagonist treatment. To block IL-1β (and IL-1α), obese mice were treated with IL1R antagonist (50 mg/kg) subcutaneously for 7 days before measuring AHR. b. AHR was measured 24hr after last IL1R antagonist treatment. Graph represents the changes in lung resistance (RL). ***p<0.001, IL1R antagonist treated obese mice were compared to HFD fed obese mice. (Two-way ANOVA). c. ILC3 cells were analyzed after PMA/Ionomycin stimulation (left panel). Graph represents the percentage of IL-17A producing ILC3 cells of the total lung lymphocytes in each group (right panel). d. Cells from BAL fluid were cultured in vitro in the presence of rIL-2, rIL-7 and rIL-1β for 72hr. To measure IL-17 production from Lin− population, cells were re-stimulated with PMA and ionomycin for 5hr. Graph represents the percentage of IL-17 producing CD45+Lin− lymphocytes in each group (bottom), *p<0.05 (Student’s T-test). e. Possible mechanisms of obesity induced AHR. High fat diet results in the activation of the NLRP3 inflammasome, through fatty acids or cholesterol crystals in macrophages in adipose tissue and in the lungs. This results in IL-1β production, which drives the development of ILC3 cells in the lungs. Treatment with IL-1R antagonist blocks the effects of IL-1β (and IL-1α), and blocks the development of pulmonary ILC3 cells.

References

    1. Flegal KM, Carroll MD, Kit BK, Ogden CL. Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA. 2012;307:491–497.
    1. Osborn O, Olefsky JM. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 2012;18:363–374.
    1. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18:716–725.
    1. Holguin F, et al. Obesity and asthma: an association modified by age of asthma onset. J Allergy Clin Immunol. 2011;127:1486–1493. e1482.
    1. Camargo CA, Jr., Weiss ST, Zhang S, Willett WC, Speizer FE. Prospective study of body mass index, weight change, and risk of adult-onset asthma in women. Arch Intern Med. 1999;159:2582–2588.
    1. Dixon AE, et al. Effects of obesity and bariatric surgery on airway hyperresponsiveness, asthma control, and inflammation. J Allergy Clin Immunol. 2011;128:508–515. e501–e502.
    1. Sutherland ER, Lehman EB, Teodorescu M, Wechsler ME. Body mass index and phenotype in subjects with mild-to-moderate persistent asthma. J Allergy Clin Immunol. 2009;123:1328–1334. e1321.
    1. Shore SA. Obesity, airway hyperresponsiveness, and inflammation. J Appl Physiol. 2010;108:735–743.
    1. Farah CS, Salome CM. Asthma and obesity: a known association but unknown mechanism. Respirology. 2012;17:412–421.
    1. Kanneganti TD, Dixit VD. Immunological complications of obesity. Nat Immunol. 2012;13:707–712.
    1. Feuerer M, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930–939.
    1. Nishimura S, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914–920.
    1. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246.
    1. Scott HA, Gibson PG, Garg ML, Wood LG. Airway inflammation is augmented by obesity and fatty acids in asthma. Eur Respir J. 2011;38:594–602.
    1. Chen Y, Dales R, Jiang Y. The association between obesity and asthma is stronger in nonallergic than allergic adults. Chest. 2006;130:890–895.
    1. Moore WC, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med. 2010;181:315–323.
    1. Lu FL, et al. Increased pulmonary responses to acute ozone exposure in obese db/db mice. Am J Physiol Lung Cell Mol Physiol. 2006;290:L856–L865.
    1. Arteaga-Solis E, et al. Inhibition of leptin regulation of parasympathetic signaling as a cause of extreme body weight-associated asthma. Cell Metab. 2013;17:35–48.
    1. Buonocore S, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature. 2010;464:1371–1375.
    1. Geremia A, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med. 2011;208:1127–1133.
    1. Coccia M, et al. IL-1beta mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4(+) Th17 cells. J Exp Med. 2012;209:1595–1609.
    1. Klose CS, et al. A T-bet gradient controls the fate and function of CCR6-RORgammat+ innate lymphoid cells. Nature. 2013;494:261–265.
    1. Sun Z, et al. Requirement for RORgamma in thymocyte survival and lymphoid organ development. Science. 2000;288:2369–2373.
    1. Sawa S, et al. RORgammat+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol. 2011;12:320–326.
    1. Cupedo T, et al. Human fetal lymphoid tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cells. Nat Immunol. 2009;10:66–74.
    1. Sanos SL, et al. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol. 2009;10:83–91.
    1. Luci C, et al. Influence of the transcription factor RORgammat on the development of NKp46+ cell populations in gut and skin. Nat Immunol. 2009;10:75–82.
    1. Chang Y-J, et al. Innate lymphoid cells mediate influenza-induced airway hyperreactivity independent of adaptive immunity. Nature Immunol. 2011;12:631–638.
    1. Halim TY, Krauss RH, Sun AC, Takei F. Lung natural helper cells are a critical source of th2 cell-type cytokines in protease allergen-induced airway inflammation. Immunity. 2012;36:451–463.
    1. Klein Wolterink RG, et al. Pulmonary innate lymphoid cells are major producers of IL-5 and IL-13 in murine models of allergic asthma. Eur J Immunol. 2012;42:1106–1116.
    1. Spits H, et al. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13:145–149.
    1. Winer S, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15:921–929.
    1. Ivanov II, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–498.
    1. Gaboriau-Routhiau V, et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity. 2009;31:677–689.
    1. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189.
    1. Lynch L, et al. Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity. 2012;37:574–587.
    1. Wen H, et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol. 2011;12:408–415.
    1. Duewell P, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464:1357–1361.
    1. Larsen CM, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356:1517–1526.
    1. Vandanmagsar B, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17:179–188.
    1. Johnston RA, et al. Diet-induced obesity causes innate airway hyperresponsiveness to methacholine and enhances ozone-induced pulmonary inflammation. J Appl Physiol. 2008;104:1727–1735.
    1. Williams AS, et al. Obesity and airway responsiveness: Role of TNFR2. Pulmonary pharmacology & therapeutics. 2012
    1. Johnston RA, et al. Allergic airway responses in obese mice. Am J Respir Crit Care Med. 2007;176:650–658.
    1. Calixto MC, et al. Obesity enhances eosinophilic inflammation in a murine model of allergic asthma. British journal of pharmacology. 2010;159:617–625.
    1. Kudo M, et al. IL-17A produced by alphabeta T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med. 2012;18:547–554.
    1. McKinley L, et al. TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J Immunol. 2008;181:4089–4097.
    1. Wakashin H, et al. IL-23 and Th17 cells enhance Th2-cell-mediated eosinophilic airway inflammation in mice. Am J Respir Crit Care Med. 2008;178:1023–1032.
    1. Lajoie S, et al. Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma. Nat Immunol. 2010;11:928–935.
    1. Murdoch JR, Lloyd CM. Resolution of allergic airway inflammation and airway hyperreactivity is mediated by IL-17-producing {gamma}{delta}T cells. Am J Respir Crit Care Med. 2010;182:464–476.
    1. Naik S, et al. Compartmentalized control of skin immunity by resident commensals. Science. 2012;337:1115–1119.
    1. Gross O, Thomas CJ, Guarda G, Tschopp J. The inflammasome: an integrated view. Immunol Rev. 2011;243:136–151.
    1. Eisenbarth SC, Colegio OR, O'Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122–1126.
    1. Besnard AG, et al. NLRP3 inflammasome is required in murine asthma in the absence of aluminum adjuvant. Allergy. 2011;66:1047–1057.
    1. Allen IC, et al. Analysis of NLRP3 in the development of allergic airway disease in mice. J Immunol. 2012;188:2884–2893.
    1. Kool M, et al. An unexpected role for uric acid as an inducer of T helper 2 cell immunity to inhaled antigens and inflammatory mediator of allergic asthma. Immunity. 2011;34:527–540.
    1. Marichal T, et al. DNA released from dying host cells mediates aluminum adjuvant activity. Nat Med. 2011;17:996–1002.
    1. Crellin NK, et al. Regulation of cytokine secretion in human CD127(+) LTi-like innate lymphoid cells by Toll-like receptor 2. Immunity. 2010;33:752–764.
    1. Winer S, et al. Obesity predisposes to Th17 bias. Eur J Immunol. 2009;39:2629–2635.
    1. Fantuzzi G. Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 2005;115:911–919. quiz 920.
    1. Kim HY, DeKruyff RH, Umetsu DT. The many paths to asthma: phenotype shaped by innate and adaptive immunity. Nat Immunol. 2010;11:577–584.
    1. Nakae S, et al. Antigen-specific T cell sensitization is impaired in IL-17-deficient mice, causing suppression of allergic cellular and humoral responses. Immunity. 2002;17:375–387.
    1. Sutterwala FS, et al. Critical role for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1. Immunity. 2006;24:317–327.
    1. McKenzie GJ, Fallon PG, Emson CL, Grencis RK, McKenzie AN. Simultaneous disruption of interleukin (IL)-4 and IL-13 defines individual roles in T helper cell type 2-mediated responses. J. Exp. Med. 1999;189:1565.
    1. Akbari O, et al. Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity. Nature Medicine. 2003;9:582–588.

Source: PubMed

3
구독하다