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:

Metabolic rewiring promotes anti-inflammatory effects of glucocorticoids

Abstract

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.

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

Fig. 1: GCs promote mitochondrial reprogramming of inflammatory macrophages.
Fig. 2: GCs control pyruvate metabolism through non-genomic mechanisms.
Fig. 3: Anti-inflammatory effects of GCs require TCA cycle integrity.
Fig. 4: Amplification of itaconate production mediates the anti-inflammatory effects of GCs in vitro.
Fig. 5: Therapeutic properties of GC-based therapies rely on itaconate production.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available on reasonable request from the corresponding author or J.-P.A. The data are not publicly available as they comprise information that could compromise research participant privacy, although source data are provided with this paper. Bulk RNA-seq datasets generated in this study are available at the GEO under accession numbers GSE250273 and GSE250274. The following databases were used in this study: the Broad Institute sgRNA design tool (https://portals.broadinstitute.org/gppx/crispick/public), Ensembl mouse reference genome GRCm38 (http://www.ensembl.org/Mus_musculus/Info/Index), mzcloud and mzVault (https://www.mzcloud.org/) and ChemSpider (http://www.chemspider.com/). Source data are provided with this paper.

References

  1. Hardy, R. S., Raza, K. & Cooper, M. S. Therapeutic glucocorticoids: mechanisms of actions in rheumatic diseases. Nat. Rev. Rheumatol. 16, 133–144 (2020).

    Article  PubMed  Google Scholar 

  2. Buckley, L. & Humphrey, M. B. Glucocorticoid-induced osteoporosis. N. Engl. J. Med. 379, 2547–2556 (2018).

    Article  PubMed  Google Scholar 

  3. Lillegraven, S. et al. Immunosuppressive treatment and the risk of diabetes in rheumatoid arthritis. PLoS ONE 14, e0210459 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vettorazzi, S., Nalbantoglu, D., Gebhardt, J. C. M. & Tuckermann, J. A guide to changing paradigms of glucocorticoid receptor function-a model system for genome regulation and physiology. FEBS J. 289, 5718–5743 (2021).

    Article  PubMed  Google Scholar 

  5. Glass, C. K. & Saijo, K. Nuclear receptor transrepression pathways that regulate inflammation in macrophages and T cells. Nat. Rev. Immunol. 10, 365–376 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Hübner, S., Dejager, L., Libert, C. & Tuckermann, J. P. The glucocorticoid receptor in inflammatory processes: transrepression is not enough. Biol. Chem. 396, 1223–1231 (2015).

    Article  PubMed  Google Scholar 

  7. O’Neill, L. A., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Li, J. X. & Cummins, C. L. Fresh insights into glucocorticoid-induced diabetes mellitus and new therapeutic directions. Nat. Rev. Endocrinol. https://doi.org/10.1038/s41574-022-00683-6 (2022).

  9. Jones, C. G., Hothi, S. K. & Titheradge, M. A. Effect of dexamethasone on gluconeogenesis, pyruvate kinase, pyruvate carboxylase and pyruvate dehydrogenase flux in isolated hepatocytes. Biochem. J. 289, 821–828 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Martin, A. D., Allan, E. H. & Titheradge, M. A. The stimulation of mitochondrial pyruvate carboxylation after dexamethasone treatment of rats. Biochem. J. 219, 107–115 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sistare, F. D. & Haynes, R. C. Jr. Acute stimulation by glucocorticoids of gluconeogenesis from lactate/pyruvate in isolated hepatocytes from normal and adrenalectomized rats. J. Biol. Chem. 260, 12754–12760 (1985).

    Article  CAS  PubMed  Google Scholar 

  12. Karra, A. G. et al. Proteomic analysis of the mitochondrial glucocorticoid receptor interacting proteins reveals pyruvate dehydrogenase and mitochondrial 60 kDa heat shock protein as potent binding partners. J. Proteom. 257, 104509 (2022).

    Article  CAS  Google Scholar 

  13. Santos, J. H. Mitochondria signaling to the epigenome: a novel role for an old organelle. Free Radic. Biol. Med. 170, 59–69 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Ryan, D. G. & O’Neill, L. A. J. Krebs cycle reborn in macrophage immunometabolism. Annu. Rev. Immunol. 38, 289–313 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Hooftman, A. & O’Neill, L. A. J. The immunomodulatory potential of the metabolite itaconate. Trends Immunol. 40, 687–698 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Peace, C. G. & O’Neill, L. A. The role of itaconate in host defense and inflammation. J. Clin. Invest. https://doi.org/10.1172/jci148548 (2022).

  17. Mills, E. L. et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556, 113–117 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bambouskova, M. et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis. Nature 556, 501–504 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. O’Neill, L. A. J. & Artyomov, M. N. Itaconate: the poster child of metabolic reprogramming in macrophage function. Nat. Rev. Immunol. 19, 273–281 (2019).

    Article  PubMed  Google Scholar 

  20. Christensen, A. D., Haase, C., Cook, A. D. & Hamilton, J. A. K/BxN serum-transfer arthritis as a model for human inflammatory arthritis. Front. Immunol. 7, 213 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Aun, M. V., Bonamichi-Santos, R., Arantes-Costa, F. M., Kalil, J. & Giavina-Bianchi, P. Animal models of asthma: utility and limitations. J. Asthma Allergy 10, 293–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Finotto, S. et al. Treatment of allergic airway inflammation and hyperresponsiveness by antisense-induced local blockade of GATA-3 expression. J. Exp. Med. 193, 1247–1260 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Buck, M. D., Sowell, R. T., Kaech, S. M. & Pearce, E. L. Metabolic instruction of immunity. Cell 169, 570–586 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ip, W. K. E., Hoshi, N., Shouval, D. S., Snapper, S. & Medzhitov, R. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science 356, 513–519 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cheng, S. C. et al. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345, 1250684 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Stifel, U. et al. Glucocorticoids coordinate macrophage metabolism through the regulation of the tricarboxylic acid cycle. Mol. Metab. 57, 101424 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Clark, A. R. Anti-inflammatory functions of glucocorticoid-induced genes. Mol. Cell. Endocrinol. 275, 79–97 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Lampropoulou, V. et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 24, 158–166 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bigenzahn, J. W. et al. LZTR1 is a regulator of RAS ubiquitination and signaling. Science 362, 1171–1177 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Aletaha, D. et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. 69, 1580–1588 (2010).

    Article  Google Scholar 

  31. Savory, J. G. et al. Discrimination between NL1- and NL2-mediated nuclear localization of the glucocorticoid receptor. Mol. Cell. Biol. 19, 1025–1037 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hofmann, J., Börnke, F., Schmiedl, A., Kleine, T. & Sonnewald, U. Detecting functional groups of Arabidopsis mutants by metabolic profiling and evaluation of pleiotropic responses. Front. Plant Sci. 2, 82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank W. Baum who helped in generating the K/BxN serum; A. Klej, R. Weinkam and R. Mancuso for technical assistance; and A. Papathanassiu (Ergon Pharmaceuticals) for providing the ACOD1 inhibitor ERG344. This work was supported by the Deutsche Forschungsgemeinschaft (DFG, CRC1181-A03/A01/B08 to G.K., G. Schett and S.F.; FOR2886 PANDORA, B01/A03/B05/Z01, to G.K., G. Schett, S.U. and A.K.; CRC1149, 251293561, C02 and CRC 1506, 450627322, C05 to J.T.; Tu220/25-1, 505870049 and Major Research Instrumentation grant 441730715), the Emerging Field Initiative (EFI) of the Friedrich-Alexander University Erlangen-Nürnberg (FAU) (EFI_Verbund_Med_05_MIRACLE to G.K., U. Sonnewald and J.H.), the Interdisziplinäres Zentrum für Klinische Forschung (IZKF) of the Friedrich-Alexander University Erlangen-Nürnberg (IZKF J91 to J.-P.A.), the Bundesministerium für Bildung und Forschung (BMBF) (MASCARA to G.K. and G.S and MelAutim to G.K.), the Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis (to G. Schabbauer, S.B., M.K. and M.H.), the FWF Sonderforschungsbereich SFB F83 (to G.S.) and the EU (Horizon 2020 ERC-2014-StG 640087, SOS and Horizon 2020 ERC-2020-CoG 101001866, INSPIRE to G.K.; Horizon Europe ERC-2021-StG 101039438, NEXUS, to S.U.; and Horizon 2020 ERC-2018-SyG nanoSCOPE and RTCure to G.S.). J.-P.A. received additional financial support from the Canadian Institutes for Health Research (CIHR, 201711MFE-395641-294537) and Fonds de recherche du Québec—Santé (FRQS, 259908). M.H. was awarded a DOC fellowship by the Austrian Academy of Sciences. S.U. was supported by the Hightech Agenda Bavaria.

Author information

Authors and Affiliations

Authors

Contributions

J.-P.A., M.Z., M.F. and U. Stifel designed the study, performed experiments, interpreted results and wrote the manuscript. B.K., R.V.T., C.G., G.E., C. Stoll, O.B.B., M.P.M., C. Scholtysek, M.B., K.P.-Z., M.S.J.M., M.D., M.K. and M.H. performed experiments, collected data and interpreted results. S.U., D.M., U. Sonnewald, D.S., A.K., S.F. and J.H. provided expertise, essential material and input and wrote the manuscript. D.C. and F.H. performed bioinformatics analysis and interpreted the data. S.B., G. Schabbauer, A.G., E.L. and G. Schett wrote the manuscript and provided valuable input. J.T. and G.K. designed the study and experiments and wrote the manuscript. All of the authors read and commented on the manuscript.

Corresponding author

Correspondence to Gerhard Krönke.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Luke O’Neill, David Ray and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Glucocorticoid-mediated control of transcription and metabolism.

(a) Heatmap illustrating the predicted activity of indicated transcription factors (TF) from the bulk mRNA sequencing data of bone marrow-derived macrophages (BMDMs) treated with vehicle (Ctrl), dexamethasone (GC, 100 nM) and LPS (100 ng/ml) for 4 or 24 h. (b) Pathway analysis derived from the bulk mRNA sequencing data illustrating pathways enriched in BMDMs treated with a combination of GC and LPS in comparison to BMDMs treated with LPS only. (c) Quantification of indicated parameters derived from a mito stress test (n = 4) and a glycolysis stress test (n = 8) of BMDMs treated with Ctrl, GC and LPS for 24 h. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured. (d) Gating strategy of the flow cytometry analysis of BMDMs treated with Ctrl, GC and LPS for 6 or 24 h and stained with MitoTracker Green and Mitotracker Red (corresponding to data in Fig. 1d and Fig. 2b). Data are presented as mean + SEM. One-sided Hypergeometric test with Bonferroni’s correction (b); one-way ANOVA with Tukey’s multiple comparison test (c).

Source Data

Extended Data Fig. 2 Regulation of TCA cycle metabolism by glucocorticoids.

(a) Mass spectrometry-based analysis of U-13C glucose carbon tracing in bone marrow-derived macrophages (BMDMs) treated with vehicle (Ctrl), dexamethasone (GC, 100 nM) and LPS (100 ng/ml). Relative U-13C exchange rate (RelER) for the indicated metabolites and the number of exchanged carbon atoms (M0 to M6). (b) Extracellular acidification rate (ECAR) of a glycolysis stress test, with quantified glycolysis and glycolytic capacity of fetal liver-derived macrophages from Nls+/+ and Nlsmut mice treated with vehicle, GC and LPS for 24 h (n = 8). (c) Immunofluorescence imaging of BMDMs treated with Ctrl, GC and LPS for 24 h and stained with MitoTracker Red and an antibody directed against the glucocorticoid receptor. (d) Immunofluorescence microscopy of PDH-X protein in BMDMs upon stimulation with Ctrl, GC and LPS for 6 h. Fluorescence signal rendering for PDH-X and MitoTracker is represented in the lower right panel, with quantification in Fig. 2g. (e) Western blot analysis of pyruvate dehydrogenase (PDH) protein levels in BMDMs treated with Ctrl, GC, 100 nM and LPS for 6, 12 and 24 h. Data are presented as mean + SEM. One-way ANOVA with Tukey’s multiple comparison test (b).

Source Data

Extended Data Fig. 3 Parallel regulation of metabolism and inflammation by glucocorticoids.

(a) ELISA-based quantification of the indicated cytokines in the supernatants human monocytes-derived macrophages from healthy donors under normoxia and hypoxia (1% O2) and treated as indicated with vehicle (Ctrl), dexamethasone (GC, 100 nM) and LPS and IFN-γ (100 ng/ml and 20 ng/ml, respectively) (n = 12). (b) Quantification of indicated parameters derived from a mito stress test and a glycolysis stress test of bone marrow-derived macrophages (BMDMs) treated with Ctrl, GC and LPS in the presence of a vehicle or roxadustat (RXD) (10 µM) for 24 h (n = 8). Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were measured. (c) ELISA-based quantification of the indicated cytokines in the supernatants of BMDMs treated with Ctrl, GC and LPS in the presence of a vehicle or UK5099 (10 µM) as indicated (n = 6). Data are presented as mean + SEM. One-way ANOVA with Tukey’s multiple comparison test (a-c).

Source Data

Extended Data Fig. 4 Itaconate as mediator of the action of glucocorticoids.

(a,b) ELISA-based quantification of the indicated cytokines in the supernatants of bone marrow-derived macrophages (BMDMs) from Acod1+/+ and Acod1−/− mice treated as indicated with vehicle (Ctrl), dexamethasone (GC, 100 nM), LPS (100 ng/ml) and (a) dimethyl itaconate (DMI; 62.5 µM) or (b) 4-octyl itaconate (4OI; 62.5 µM) (n = 7 for a, n = 6 for b). (c) Heatmap illustrating the predicted activity of indicated transcription factors (TF) derived from bulk mRNA sequencing data of BMDMs from Acod1+/+ and Acod1−/− mice treated with Ctrl, GC and LPS for 24 h. (d) ELISA-based quantification of the indicated cytokines in the supernatants of human monocytes-derived macrophages from healthy donors treated as indicated with Ctrl, GC and LPS and IFN-γ (20 ng/ml) in the presence of a vehicle or the ACOD1 inhibitor ERG344 (500 µM) (n = 12). (e) ELISA-based quantification of the indicated cytokines in the supernatants of human THP-1 monocyte-derived sgRen and sgAcod1 macrophages treated as indicated with Ctrl, GC and LPS and IFN-γ (n = 6). Similar results were observed with two other sgRNAs targeting Acod1. (f) Western blot analysis of NRF2 protein level in Nfe2l2+/+ and Nfe2l2−/− BMDMs. Six hours prior to collection, cells were treated with 2 µM MG-132 to promote NRF2 stabilization. (g) Western blot analysis of NRF2 protein level in BMDMs treated with Ctrl, GC and LPS for 24 h. Six hours prior to collection, cells were treated with 2 µM MG-132 to promote NRF2 stabilization. (h) ELISA-based quantification of the indicated cytokines in the supernatants of BMDMs from Nfe2l2+/+ and Nfe2l2−/− mice treated as indicated with Ctrl, GC and LPS (n = 6). Data are presented as mean + SEM. One-way ANOVA with Tukey’s multiple comparison test (a-b, d-e and h).

Source Data

Extended Data Fig. 5 Murine models of immune-mediated diseases.

(a) Timeline of the treatment protocol used for the mouse model of LPS-induced lung injury. (b-c) Gating strategy and representative plots of the flow cytometry analysis of cells isolated from the bronchoalveolar lavage fluid (BALF) of Acod1+/+ and Acod1−/− mice during LPS-induced lung injury (corresponding to data in Fig. 5a). (d) Representative H&E stainings of lung sections of Acod1+/+ and Acod1−/− mice during LPS-induced lung injury (scale bars = 100 µm). (e) Timeline of the used treatment protocol for the mouse model of K/BxN serum transfer arthritis. (f) Timeline of the treatment protocol used for the mouse model of ovalbumin (OVA)-induced allergic airway inflammation. (g) Gating strategy of the flow cytometry analysis of cells derived from the BALF of mice during ovalbumin-induced allergic airway inflammation (corresponding to data in Fig. 5i).

Extended Data Fig. 6 Metabolic rewiring promotes anti-inflammatory effects of glucocorticoids.

(a) Graphical summary of the results presented in the manuscript.

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Auger, JP., Zimmermann, M., Faas, M. et al. Metabolic rewiring promotes anti-inflammatory effects of glucocorticoids. Nature (2024). https://doi.org/10.1038/s41586-024-07282-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07282-7

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research