Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation

Abstract

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Constitutive Arg1 expression is a conserved trait of precursor and mature ILC2s across diverse tissue sites.
Figure 2: ILC2s are a main source of Arg1 in the lung during type 2 inflammation.
Figure 3: Human ILC2s express Arg1 during chronic lung disease.
Figure 4: Absence of ILC-intrinsic Arg1 restrains ILC2 responses and dampens airway inflammation.
Figure 5: Development of airway inflammation is dependent on ILC-intrinsic, not myeloid-cell-intrinsic, expression of Arg1.
Figure 6: Cell-intrinsic Arg1 controls optimal ILC2 proliferation.
Figure 7: Inhibition of cell-intrinsic Arg1 disrupts the balance of amino acid metabolites and impairs polyamine synthesis in ILC2s.
Figure 8: Arg1 controls the maximal glycolytic capacity of activated ILC2s.

Similar content being viewed by others

References

  1. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. McKenzie, A.N., Spits, H. & Eberl, G. Innate lymphoid cells in inflammation and immunity. Immunity 41, 366–374 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Chang, Y.J. et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat. Immunol. 12, 631–638 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Barlow, J.L. et al. Innate IL-13-producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity. J. Allergy Clin. Immunol. 129, 191–198 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Halim, T.Y. et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity 40, 425–435 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Monticelli, L.A. et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 12, 1045–1054 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Buck, M.D., O'Sullivan, D. & Pearce, E.L. T cell metabolism drives immunity. J. Exp. Med. 212, 1345–1360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Vander Heiden, M.G., Cantley, L.C. & Thompson, C.B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bando, J.K., Nussbaum, J.C., Liang, H.E. & Locksley, R.M. Type 2 innate lymphoid cells constitutively express arginase-I in the naive and inflamed lung. J. Leukoc. Biol. 94, 877–884 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bando, J.K., Liang, H.E. & Locksley, R.M. Identification and distribution of developing innate lymphoid cells in the fetal mouse intestine. Nat. Immunol. 16, 153–160 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Rath, M., Muller, I., Kropf, P., Closs, E.I. & Munder, M. Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front. Immunol. 5, 532 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Thomas, A.C. & Mattila, J.T. “Of mice and men”: arginine metabolism in macrophages. Front. Immunol. 5, 479 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Murray, P.J. Amino acid auxotrophy as a system of immunological control nodes. Nat. Immunol. 17, 132–139 (2015).

    Article  CAS  Google Scholar 

  14. Raber, P., Ochoa, A.C. & Rodriguez, P.C. Metabolism of L-arginine by myeloid-derived suppressor cells in cancer: mechanisms of T cell suppression and therapeutic perspectives. Immunol. Invest. 41, 614–634 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wynn, T.A., Chawla, A. & Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 496, 445–455 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Murray, P.J. & Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 11, 723–737 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gibbons, M.A. et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am. J. Respir. Crit. Care Med. 184, 569–581 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Rodriguez, P.C., Quiceno, D.G. & Ochoa, A.C. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 109, 1568–1573 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pesce, J.T. et al. Arginase-1-expressing macrophages suppress Th2 cytokine-driven inflammation and fibrosis. PLoS Pathog. 5, e1000371 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Barron, L. et al. Role of arginase 1 from myeloid cells in th2-dominated lung inflammation. PLoS ONE 8, e61961 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cloots, R.H. et al. Ablation of Arg1 in hematopoietic cells improves respiratory function of lung parenchyma, but not that of larger airways or inflammation in asthmatic mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 305, L364–L376 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. North, M.L., Khanna, N., Marsden, P.A., Grasemann, H. & Scott, J.A. Functionally important role for arginase 1 in the airway hyperresponsiveness of asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 296, L911–L920 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, M. et al. Inhibition of arginase I activity by RNA interference attenuates IL-13-induced airways hyperresponsiveness. J. Immunol. 177, 5595–5603 (2006).

    Article  CAS  PubMed  Google Scholar 

  24. Hoyler, T. et al. The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity 37, 634–648 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lara, A. et al. Alterations of the arginine metabolome in asthma. Am. J. Respir. Crit. Care Med. 178, 673–681 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bratt, J.M., Zeki, A.A., Last, J.A. & Kenyon, N.J. Competitive metabolism of L-arginine: arginase as a therapeutic target in asthma. J. Biomed. Res. 25, 299–308 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Holguin, F. et al. An association between L-arginine/asymmetric dimethyl arginine balance, obesity, and the age of asthma onset phenotype. Am. J. Respir. Crit. Care Med. 187, 153–159 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Vonk, J.M. et al. Arginase 1 and arginase 2 variations associate with asthma, asthma severity and β2 agonist and steroid response. Pharmacogenet. Genomics 20, 179–186 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Bergeron, C. et al. Influence of cigarette smoke on the arginine pathway in asthmatic airways: increased expression of arginase I. J. Allergy Clin. Immunol. 119, 391–397 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Tadié, J.M. et al. Role of nitric oxide synthase/arginase balance in bronchial reactivity in patients with chronic obstructive pulmonary disease. Am. J. Physiol. Lung Cell. Mol. Physiol. 294, L489–L497 (2008).

    Article  PubMed  CAS  Google Scholar 

  31. Wu, G. et al. Arginine metabolism and nutrition in growth, health and disease. Amino Acids 37, 153–168 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Hams, E. et al. IL-25 and type 2 innate lymphoid cells induce pulmonary fibrosis. Proc. Natl. Acad. Sci. USA 111, 367–372 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Turner, J.E. et al. IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. J. Exp. Med. 210, 2951–2965 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wojno, E.D. et al. The prostaglandin D2 receptor CRTH2 regulates accumulation of group 2 innate lymphoid cells in the inflamed lung. Mucosal Immunol. 8, 1313–1323 (2015).

    Article  PubMed  CAS  Google Scholar 

  35. Reece, J.J., Siracusa, M.C. & Scott, A.L. Innate immune responses to lung-stage helminth infection induce alternatively activated alveolar macrophages. Infect. Immun. 74, 4970–4981 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Marsland, B.J., Kurrer, M., Reissmann, R., Harris, N.L. & Kopf, M. Nippostrongylus brasiliensis infection leads to the development of emphysema associated with the induction of alternatively activated macrophages. Eur. J. Immunol. 38, 479–488 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Fujita, M. & Nakanishi, Y. The pathogenesis of COPD: lessons learned from in vivo animal models. Med. Sci. Monit. 13, RA19–RA24 (2007).

    PubMed  Google Scholar 

  38. Demkow, U. & van Overveld, F.J. Role of elastases in the pathogenesis of chronic obstructive pulmonary disease: implications for treatment. Eur. J. Med. Res. 15, 27–35 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Sonnenberg, G.F., Monticelli, L.A., Elloso, M.M., Fouser, L.A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Knippenberg, S., Brumshagen, C., Aschenbrenner, F., Welte, T. & Maus, U.A. Arginase 1 activity worsens lung-protective immunity against Streptococcus pneumoniae infection. Eur. J. Immunol. 45, 1716–1726 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Miller-Fleming, L., Olin-Sandoval, V., Campbell, K. & Ralser, M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J. Mol. Biol. 427, 3389–3406 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Phang, J.M., Liu, W., Hancock, C.N. & Fischer, J.W. Proline metabolism and cancer: emerging links to glutamine and collagen. Curr. Opin. Clin. Nutr. Metab. Care 18, 71–77 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Millard, P., Letisse, F., Sokol, S. & Portais, J.C. IsoCor: correcting MS data in isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. van der Windt, G.J. & Pearce, E.L. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol. Rev. 249, 27–42 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Molofsky, A.B. et al. Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages. J. Exp. Med. 210, 535–549 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, D. et al. IL-33 promotes ST2-dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice. J. Allergy Clin. Immunol. 134, 1422–1432 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xue, L. et al. Prostaglandin D2 activates group 2 innate lymphoid cells through chemoattractant receptor-homologous molecule expressed on TH2 cells. J. Allergy Clin. Immunol. 133, 1184–1194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schlenner, S.M. et al. Fate mapping reveals separate origins of T cells and myeloid lineages in the thymus. Immunity 32, 426–436 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Brestoff, J.R. et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature 519, 242–246 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Osborne, L.C. et al. Virus-helminth coinfection reveals a microbiota-independent mechanism of immunomodulation. Science 345, 578–582 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the D. Artis and G.F. Sonnenberg laboratories for critical reading of this manuscript. We thank R.M. Locksley (University of California, San Francisco, San Francisco, California, USA) for generously providing the Arg1YFP mice and B. Vallance (University of British Columbia, Vancouver, British Columbia, Canada) for providing Citrobacter rodentium. This work was supported by the National Institutes of Health (NIH) (grants AI061570, AI087990, AI074878, AI083480, AI095466, AI095608, AI102942 and AI097333 to D.A.; T32-AI007532 to L.A.M.; CA181125 and AI091965 to E.L.P.; HL087115, HL081619, HL096845, HL115354 and HL114626 to J.D.C.; HL116656 to E.C.; and HL090021 and K23-HL121406 to J.M.D.), the Burroughs Wellcome Fund (D.A. and E.L.P.), the Crohn's & Colitis Foundation of America (D.A. and M.R.H.), the Edmond J. Safra Foundation/Cancer Research Institute (L.C.O.), the National Science Foundation (grant DGE-1143954 to M.D.B.), the Robert Wood Johnson Foundation (grant AMFDP 70640 to E.C.), the Thoracic Surgery Foundation (E.C.) and the German Research Foundation (DFG SFB 873–project 11 to H.-R.R.). We thank the Mucosal Immunology Studies Team (MIST) of the NIH NIAID for shared expertise and resources.

Author information

Authors and Affiliations

Authors

Contributions

L.A.M. carried out mouse experiments with technical assistance from A.-L.F., N.A.Y., L.C.O., M.R.H. and S.V.T. M.D.B. and E.L.P. carried out Seahorse metabolic assays. H.S. and J.R.C. carried out liquid chromatography–mass spectrometry experiments. J.M.D., E.C. and J.D.C. provided human tissue samples. L.A.M., S.A.S. and E.D.T.W. carried out experiments involving human tissue samples. H.-R.R. provided Il7rCre/+ mice. L.A.M. and D.A. conceived of the study, designed experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to David Artis.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Distribution of human ILCs during chronic lung disease.

Quantification of flow cytometric analysis of lung tissue from patients with COPD or IPF identifying (a) frequencies of total LinCD127+ ILCs (Lin gating includes CD3, CD5, CD56, CD19, FcεR1, CD14, CD16). Cells are pregated on live CD45+ cells. (b) Frequency of ILC subsets defined by IL-33R (ST2L) and CRTH2 expression, pregated on LinCD127+ cells. (c) Mean fluorescence intensity (MFI) of intracellular Arg1 expression in each cell subset. Data shown are from one experiment. Data are the mean ± SEM, n = 8 COPD and n = 8 IPF tissue samples.

Supplementary Figure 2 Fate-mapping analysis of IL-7Rα expression in immune cell subsets during lung inflammation.

Il7rcre/+ Rosa26floxSTOP-eYFP and C57BL/6J WT mice were treated with 30 μg papain or PBS intranasally (i.n.) for 5 days and analyzed on day 6. Representative flow cytometric histograms of eYFP expression from lung immune cell subsets, Il7rcre/+ Rosa26floxSTOP-eYFP red line; WT, gray shaded. Numbers indicate the percentage of total cells that are marked by a history of IL-7Rα expression for ILC2s (LinCD45+CD90+CD25+IL-33R+), CD4+ T cells (CD3+CD4+), B cells (CD19+CD3), NK cells (NK1.1+CD3) and eosinophils (CD11b+Siglec F+CD11c). All data are representative of two independent experiments with similar results. N = 3-4 mice per group.

Supplementary Figure 3 Analysis of Arg1 expression in immune cell subsets during lung inflammation.

Arg1YFP and C57BL/6J WT mice were treated with 30 μg papain or PBS i.n. for 5 days and analyzed on day 6. Representative flow cytometric histograms of Arg1-YFP expression from lung immune cell subsets, Arg1YFP, blue line; WT, gray shaded. Numbers indicate the percentage of total cells that express Arg1 for ILC2s (LinCD45+CD90+CD25+IL-33R+), CD4+ T cells (CD3+CD4+), B cells (CD19+CD3), NK cells (NK1.1+CD3) and eosinophils (CD11b+Siglec F+CD11c). All data are representative of two independent experiments with similar results. N = 3-4 mice per group.

Supplementary Figure 4 Loss of ILC-intrinsic Arg1 impairs ILC2 responses during chronic lung inflammation.

(a-d) Arg1fl/fl and Arg1ΔILC mice were infected with 500 L3 Nippostrongylus brasiliensis (Nb) larvae subcutaneously and assessed one month post infection. (a) Representative flow cytometric plots and (b) total frequencies of ILC2s (LinCD45+CD90+CD25+) in the lungs of naïve and Nb-infected mice. (c) mRNA expression of (c) Arg1 in lung tissue, determined by RT-PCR and expressed relative to levels in naive Arg1fl/fl mice. (d-g) Arg1fl/fl and Arg1ΔILC mice were instilled with 3 units of elastase or PBS intratracheally and assessed one month post treatment. (d) Representative flow cytometric plots and (e) total frequencies of ILC2s (Lin CD45+ CD90+ CD25+) in the lungs of PBS and elastase-treated mice. (f) mRNA expression of Arg1 in lung tissue, determined by RT-PCR and expressed relative to levels in PBS-treated Arg1fl/fl mice. (g) Representative histological sections of whole left lung lobes stained with H&E. Scale bar, 100 μm. Data are representative of two independent experiments with similar results. N = 2 mice (PBS and naïve) and n = 4 mice (Nb and elastase). Data shown are the mean ± SEM. *p < 0.05, **p < 0.01, *** p < 0.001, as determined by unpaired Student’s t test.

Supplementary Figure 5 ILC3 development and anti-bacterial immunity are independent of ILC-intrinsic Arg1.

(a) Representative flow cytometric plots and (b) frequencies of total ILCs (LinCD45+CD90+CD25+) in the gut-associated mesenteric lymph nodes (mLN) of naïve Arg1fl/fl and Arg1ΔILC mice. (c) Representative flow cytometric plots and (d) total frequencies of GATA3hi ILC2 versus RORγt+ ILC3 populations, parent gate is total ILCs as in (a,b). (e) RORc(γt)gfp/gfp, Rag2-/-Il2rg-/-, Rag1-/-, Arg1fl/fl and Arg1ΔILC mice were infected with 1 × 1010 CFU of Citrobacter rodentium (C. rodentium) in 200 µL via oral gavage. Mice were monitored for morbidity and mortality over the course of 28 days. Data are representative of two independent experiments with similar results. N = 2-5 mice per group. Data shown are the mean ± SEM. NS, not significant.

Supplementary Figure 6 ILC-intrinsic Arg1 does not influence cell survival.

(a-c) Arg1fl/fl, Arg1+/+ Il7rCre/+ and Arg1ΔILC were treated with 30 μg papain or PBS i.n. for 5 days and assessed on day 6 for survival of lung ILC2s by Annexin V and 7AAD staining. (a) Representative flow cytometric plot showing validation of gating strategy to identify dead cells by 7AAD and Annexin V co-staining. (b) Representative flow cytometric plots and (c) quantification of Annexin V+ 7AAD+ ILC2 frequencies. ILC2s gated as LinCD45+CD90+CD25+. (d) C57BL/6J WT mice were treated with 100 ng of rmIL-33 intranasally every 3 days for 2 weeks. Sort-purified lung ILC2s (LinCD45+CD90+CD127+CD25+IL-33R+) were labeled with cell trace violet and cultured for 48 h with rmIL-2, rmIL-7, rmIL-33 and DMSO or Nω-hydroxy-nor-Arginine (nor-NOHA). Cells were assessed for dilution of the cell trace dye using flow cytometry. Data are representative of two experiments (a-c) or three experiments (d) with similar results. N = 3-4 mice per group.

Supplementary Figure 7 ILC-intrinsic arginase metabolism.

(a) Pathway diagram of arginine metabolism in activated ILC2s. Green indicates positive metabolism, black indicates no significant metabolism, grey indicates metabolite not detected or not examined. Blue indicates enzymes arginase (Arg1), nitric oxide synthase (NOS) and L-Arginine:glycine amidinotransferase (AGAT). (b) Experimental schematic for liquid chromatography mass spectrometry analysis. C57BL/6J WT mice were treated with 100 ng rmIL-33 i.n. every 3 days for 2-3 weeks and sort-purified lung ILC2s (LinCD45+CD90+CD127+CD25+IL-33R+) were cultured with rmIL-2, rmIL-7 and rmIL-33 in DMSO or 500μM nor-NOHA for 24 h. Methanol-fixed cells were subjected to liquid chromatography mass spectrometry analysis.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 2731 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Monticelli, L., Buck, M., Flamar, AL. et al. Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat Immunol 17, 656–665 (2016). https://doi.org/10.1038/ni.3421

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3421

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing