Itaconate drives mtRNA-mediated type I interferon production through inhibition of succinate dehydrogenase | Nature Metabolism

Blog

HomeHome / Blog / Itaconate drives mtRNA-mediated type I interferon production through inhibition of succinate dehydrogenase | Nature Metabolism

Oct 16, 2024

Itaconate drives mtRNA-mediated type I interferon production through inhibition of succinate dehydrogenase | Nature Metabolism

Nature Metabolism (2024)Cite this article Metrics details Itaconate is one of the most highly upregulated metabolites in inflammatory macrophages and has been shown to have immunomodulatory

Nature Metabolism (2024)Cite this article

Metrics details

Itaconate is one of the most highly upregulated metabolites in inflammatory macrophages and has been shown to have immunomodulatory properties. Here, we show that itaconate promotes type I interferon production through inhibition of succinate dehydrogenase (SDH). Using pharmacological and genetic approaches, we show that SDH inhibition by endogenous or exogenous itaconate leads to double-stranded mitochondrial RNA (mtRNA) release, which is dependent on the mitochondrial pore formed by VDAC1. In addition, the double-stranded RNA sensors MDA5 and RIG-I are required for IFNβ production in response to SDH inhibition by itaconate. Collectively, our data indicate that inhibition of SDH by itaconate links TCA cycle modulation to type I interferon production through mtRNA release.

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

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Subscribe to this journal

Receive 12 digital issues and online access to articles

$119.00 per year

only $9.92 per issue

Buy this article

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

The RNA-seq data in this manuscript have been deposited at Dryad (https://doi.org/10.5061/dryad.vhhmgqp36). The IREA data in this manuscript can be found in Supplementary Table 1. Source data are provided with this paper.

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

Article CAS PubMed PubMed Central Google Scholar

Michelucci, A. et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc. Natl Acad. Sci. USA 110, 7820–7825 (2013).

Article CAS PubMed PubMed Central Google Scholar

Peace, C. G. & O’Neill, L. A. The role of itaconate in host defense and inflammation. J. Clin. Invest. 132, e148548 (2022).

Article CAS PubMed PubMed Central Google Scholar

Cordes, T. et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J. Biol. Chem. 291, 14274–14284 (2016).

Article CAS PubMed PubMed Central Google Scholar

Swain, A. et al. Comparative evaluation of itaconate and its derivatives reveals divergent inflammasome and type I interferon regulation in macrophages. Nat. Metab. 2, 594–602 (2020).

Article CAS PubMed PubMed Central Google Scholar

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

He, W. et al. Mesaconate is synthesized from itaconate and exerts immunomodulatory effects in macrophages. Nat. Metab. 4, 524–533 (2022).

Article CAS PubMed PubMed Central Google Scholar

Hooftman, A. et al. The immunomodulatory metabolite itaconate modifies NLRP3 and inhibits inflammasome activation. Cell Metab. 32, 468–478 (2020).

Article CAS PubMed PubMed Central Google Scholar

Liao, S. T. et al. 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to exert anti-inflammatory effects. Nat. Commun. 10, 5091 (2019).

Article PubMed PubMed Central Google Scholar

Qin, W. et al. S-glycosylation-based cysteine profiling reveals regulation of glycolysis by itaconate. Nat. Chem. Biol. 15, 983–991 (2019).

Article CAS PubMed Google Scholar

Runtsch, M. C. et al. Itaconate and itaconate derivatives target JAK1 to suppress alternative activation of macrophages. Cell Metab. 34, 487–501 (2022).

Article CAS PubMed Google Scholar

Zhang, Z. et al. Itaconate is a lysosomal inducer that promotes antibacterial innate immunity. Mol. Cell 82, 2844–2857 (2022).

Article CAS PubMed Google Scholar

Chen, C. et al. ABCG2 is an itaconate exporter that limits antibacterial innate immunity by alleviating TFEB-dependent lysosomal biogenesis. Cell Metab. https://doi.org/10.1016/j.cmet.2023.12.015 (2024).

Article PubMed PubMed Central Google Scholar

Chouchani, E. T. et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature 515, 431–435 (2014).

Article CAS PubMed PubMed Central Google Scholar

Tannahill, G. M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

Article CAS PubMed PubMed Central Google Scholar

Cui, A. et al. Dictionary of immune responses to cytokines at single-cell resolution. Nature 625, 377–384 (2023).

Article PubMed PubMed Central Google Scholar

Hooftman, A. et al. Macrophage fumarate hydratase restrains mtRNA-mediated interferon production. Nature 615, 490–498 (2023).

Article CAS PubMed PubMed Central Google Scholar

Peace, C. G., O’Carroll, S. M. & O’Neill, L. A. J. Fumarate hydratase as a metabolic regulator of immunity. Trends Cell Biol. 34, 442–450 (2023).

Article PubMed Google Scholar

Miyadera, H. et al. Atpenins, potent and specific inhibitors of mitochondrial complex II (succinate–ubiquinone oxidoreductase). Proc. Natl Acad. Sci. USA 100, 473–477 (2003).

Article CAS PubMed PubMed Central Google Scholar

Sun, F. et al. Crystal structure of mitochondrial respiratory membrane protein complex II. Cell 121, 1043–1057 (2005).

Article CAS PubMed Google Scholar

Tigano, M., Vargas, D. C., Tremblay-Belzile, S., Fu, Y. & Sfeir, A. Nuclear sensing of breaks in mitochondrial DNA enhances immune surveillance. Nature 591, 477–481 (2021).

Article CAS PubMed Google Scholar

Dhir, A. et al. Mitochondrial double-stranded RNA trigger antiviral signalling in humans. Nature 560, 238–242 (2018).

Article CAS PubMed PubMed Central Google Scholar

Zhong, Z. et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560, 198–203 (2018).

Article CAS PubMed PubMed Central Google Scholar

Kim, J. et al. VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease. Science 366, 1531–1536 (2019).

Article CAS PubMed PubMed Central Google Scholar

O’Carroll, S. M., Henkel, F. D. R. & O’Neill, L. A. J. Metabolic regulation of type I interferon production. Immunol. Rev. 323, 276–287 (2024).

Article PubMed Google Scholar

Monzel, A. S., Enríquez, J. A. & Picard, M. Multifaceted mitochondria: moving mitochondrial science beyond function and dysfunction. Nat. Metab. 5, 546–562 (2023).

Article PubMed PubMed Central Google Scholar

Murphy, M. P. & O’Neill, L. A. J. A break in mitochondrial endosymbiosis as a basis for inflammatory diseases. Nature 626, 271–279 (2024).

Article CAS PubMed Google Scholar

Yoon, J., Kim, S., Lee, M. & Kim, Y. Mitochondrial nucleic acids in innate immunity and beyond. Exp. Mol. Med. 55, 2508–2518 (2023).

Article CAS PubMed PubMed Central Google Scholar

Peace, C. G., Hooftman, A. & Ryan, D. G. Inflammatory mitochondrial nucleic acids as drivers of pathophysiology. Clin. Transl. Med. 13, e1403 (2023).

Article CAS PubMed PubMed Central Google Scholar

Zecchini, V. et al. Fumarate induces vesicular release of mtDNA to drive innate immunity. Nature 615, 499–506 (2023).

Article CAS PubMed PubMed Central Google Scholar

Ryan, T. A. J. et al. Dimethyl fumarate and 4-octyl itaconate are anticoagulants that suppress tissue factor in macrophages via inhibition of type I interferon. Nat. Commun. 14, 3513 (2023).

Article CAS PubMed PubMed Central Google Scholar

Ryan, D. G. et al. Nrf2 activation reprograms macrophage intermediary metabolism and suppresses the type I interferon response. iScience 25, 103827 (2022).

Article CAS PubMed PubMed Central Google Scholar

Ackermann, W. W. & Potter, V. R. Enzyme inhibition in relation to chemotherapy. Proc. Soc. Exp. Biol. Med. 72, 1–9 (1949).

Article CAS PubMed Google Scholar

Daniels, B. P. et al. The nucleotide sensor ZBP1 and kinase RIPK3 induce the enzyme IRG1 to promote an antiviral metabolic state in neurons. Immunity 50, 64–76 (2019).

Article CAS PubMed PubMed Central Google Scholar

Cho, H. et al. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat. Med. 19, 458–464 (2013).

Article CAS PubMed PubMed Central Google Scholar

O’Carroll, S. M. & O’Neill, L. A. J. Targeting immunometabolism to treat COVID-19. Immunother. Adv. 1, ltab013 (2021).

Article PubMed PubMed Central Google Scholar

Gobelli, D. et al. The mitochondrial succinate dehydrogenase complex controls the STAT3–IL-10 pathway in inflammatory macrophages. iScience 26, 107473 (2023).

Article CAS PubMed PubMed Central Google Scholar

Salabei, J. K., Gibb, A. A. & Hill, B. G. Comprehensive measurement of respiratory activity in permeabilized cells using extracellular flux analysis. Nat. Protoc. 9, 421–438 (2014).

Article CAS PubMed PubMed Central Google Scholar

Download references

We acknowledge the Irish Blood Transfusion Service for supporting our research by approving us to use anonymized untransfused blood components in our research. L.A.J.O. was funded by the European Research Council (Metabinnate 834370) and the Science Foundation Ireland (20/SPP/3685). The Simarro lab is funded by (1) Agencia Estatal de Investigación, Grant Number PID2020-118517RB-I00, and (2) Programa Estratégico Instituto de Biomedicina y Genética Molecular (IBGM), Junta de Castilla y León, Award Number CCVC8485. We thank all members of the O’Neill laboratory for helpful discussions.

These authors contributed equally: Shane M. O’Carroll, Christian G. Peace.

School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland

Shane M. O’Carroll, Christian G. Peace, Juliana E. Toller-Kawahisa, Yukun Min, Alexander Hooftman, Sara Charki, Aline Zoller, Anne F. Mcgettrick, Emily A. Day & Luke A. J. O’Neill

Histology Lab, Children’s Health Ireland at Crumlin, Dublin, Ireland

Louise Kehoe & Maureen J. O’Sullivan

Departments of Histopathology and Paediatrics, Trinity College Dublin, Dublin, Ireland

Maureen J. O’Sullivan

Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain

Maria Simarro

Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain

Maria Simarro

Department of Medicine, Division of Endocrinology, Stanford School of Medicine, Stanford, CA, USA

Neali Armstrong & Justin P. Annes

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

S.M.O., C.G.P. and L.A.J.O. conceptualized the project. S.M.O. and C.G.P. were experimentalists, provided intellectual input, designed all experiments, analyzed and visualized the data and co-wrote the paper with input from all authors. J.E.T.-K., Y.M., A.H., S.C., A.Z., A.F.M., E.A.D. and L.K. performed in vitro experiments. M.J.O., M.S., N.A. and J.P.A. assisted with genetic models and provided intellectual input. L.A.J.O. obtained funding, provided intellectual input and oversaw the research program.

Correspondence to Luke A. J. O’Neill.

L.A.J.O. is a paid consultant for Sitryx Therapeutics, Sail Biomedicines and Montai Health. The other authors declare no competing interests.

Nature Metabolism thanks Ricardo Gazzinelli, Karsten Hiller and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Christoph Schmitt, in collaboration with the Nature Metabolism editorial team.

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

a, PCA plot of RNA-Seq analysis. b, Differential expression gene clustering heatmap with Z-score. c, MsigDB Hallmark pathway enrichment analysis of differentially expressed genes from itaconate pretreated BMDMs followed by 3 h LPS treatment. d, NRF2 pathway enrichment plot of itaconate-treated BMDMs. e) Western blot of IRG1 in Irg1+/+ and Irg1−/− BMDMs treated with LPS for 24 h and 48 h. f, qPCR of IRG1 (p = 1.017e-11) in human PBMCs that were harvested in the presence of Irg1 siRNA (n=3 donors; LPS 24 h). Data are presented as mean ± SEM. P values were calculated using two-way ANOVA for multiple comparisons.

Source data

a, IFNβ release and b, Ifnb1 expression in BMDMs pretreated with DMSO or 4-OI a (n=6 mice; LPS 4 h) b, (n=9; LPS 4 h). c, Ifnb1 expression of BMDMs pretreated with DMSO or DEM (n=3 mice; LPS 4 h) from 3 independent experiments. d, Normalised counts from RNA-seq data in BMDMs pretreated with DMSO or DMF (n=3 mice; LPS 4 h) from 3 independent experiments Data are presented as mean ± SEM. P values were calculated using one-way ANOVA for multiple comparisons or two-tailed student’s t-test for pairwise comparisons.

Source data

a, Sdhb and b, Il1b expression in Sdhbfl/fl and Sdhb−/− BMDMs (n=2 mice; LPS 4 h) from 2 independent experiments. c, IFNB expression in IRG1+/+ and IRG1−/− THP-1 cell lines pretreated with DMSO or TTFA (n=4 replicates; LPS/IFN𝛄 4 h) from 2 independent experiments d, IFNB expression in THP-1 derived macrophages pretreated with PBS or Mesaconate (n=4 replicates; LPS/IFNγ 4 h) from 2 independent experiments. Data are presented as mean ± SEM. P values were calculated using two-way ANOVA for multiple comparisons or two-tailed student’s t-test for pairwise comparisons.

Source data

a-d, Expression of a, Ddx58, b, Ifih1, c, Mb21d1, and d, Tlr9 in BMDMs in the presence of a,c, Mb21d1, Ddx58, b,d, Tlr9 and Ifih1 siRNA pretreated with DMSO or TTFA (n=3 mice; LPS 4 h) from 3 independent experiments. e-g, Expression of e, Mb21d1 f, Ifih1, and g, Ddx58 in the presence of Mb21d1, Ifih1, and Ddx58 siRNA in BMDMs pretreated with PBS or ITA (n=2-3 mice; LPS 4 h) from 2 or 3 independent experiments. h-j IFNβ release in BMDMs transfected with h, G3-YSD, i, Poly(I:C), or j, 5′PPP-dsRNA in the presence of h, Mb21d1, i, Ifih1, or j, Ddx58 siRNA (n=3 mice; 4 h) from 3 independent experiments. k, Ifnb1 and l, Tmem173 expression in BMDMs pretreated with DMSO or TTFA in the presence of Tmem173 siRNA (n=3 mice; LPS 4 h). Data are presented as mean ± SEM. P values were calculated using two-way ANOVA for multiple comparisons.

Source data

a, qPCR of mitochondrial DNA-encoded genes in BMDMs after mtDNA depletion with EtBr (6 days) followed by TTFA pretreatment (0.5 mM, 3 h) and LPS stimulation (n = 3 mice, 4 h) from 3 independent experiments. b, qPCR of mitochondrial DNA-encoded gene in BMDMs after POLRMT inhibition with IMT1 followed by itaconate pretreatment (3 h) and LPS stimulation (n = 3 mice, 4 h) from 3 independent experiments. c, Western blot of organellar and cytosolic fractions of BMDMs after subcellular digitonin fractionation (n=2 mice); Images representative of two independent experiments. d, qPCR of Ifnb1, Bak1, and Bax, after Bak1 or Bax siRNA knockdown followed by TTFA pretreatment (3 h) and LPS stimulation (n = 3 mice, 4 h) from 3 independent experiments. qPCR data of e, Vdac1 or f, Snx9 in BMDMs after Vdac1 or Snx9 siRNA knockdown followed by pretreated with TTFA (0.5 mM, 3 h) followed by LPS stimulation (n = 5 mice, 4 h) from 3 independent experiments. g, Immunofluorescence images of BMDMs pretreated with DMSO or TTFA (0.5 mM, 3 h) followed by LPS (n = 2 mice, 4 h). Images representative of 2 independent experiments. Data are presented as mean ± SEM. P values were calculated using two-way ANOVA for multiple comparisons.

Source data

Sequence of primers IREA raw data.

Source data for Fig. 1.

Source data for Fig. 2.

Source data for Fig. 3.

Source data for Fig. 4.

Source data for Extended Data Fig. 1.

Source data for Extended Data Fig. 2.

Source data for Extended Data Fig. 3.

Source data for Extended Data Fig. 4.

Source data for Extended Data Fig. 5.

Uncropped blots.

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

O’Carroll, S.M., Peace, C.G., Toller-Kawahisa, J.E. et al. Itaconate drives mtRNA-mediated type I interferon production through inhibition of succinate dehydrogenase. Nat Metab (2024). https://doi.org/10.1038/s42255-024-01145-1

Download citation

Received: 11 April 2024

Accepted: 19 September 2024

Published: 15 October 2024

DOI: https://doi.org/10.1038/s42255-024-01145-1

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative