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:

New mitochondrial DNA synthesis enables NLRP3 inflammasome activation

Abstract

Dysregulated NLRP3 inflammasome activity results in uncontrolled inflammation, which underlies many chronic diseases. Although mitochondrial damage is needed for the assembly and activation of the NLRP3 inflammasome, it is unclear how macrophages are able to respond to structurally diverse inflammasome-activating stimuli. Here we show that the synthesis of mitochondrial DNA (mtDNA), induced after the engagement of Toll-like receptors, is crucial for NLRP3 signalling. Toll-like receptors signal via the MyD88 and TRIF adaptors to trigger IRF1-dependent transcription of CMPK2, a rate-limiting enzyme that supplies deoxyribonucleotides for mtDNA synthesis. CMPK2-dependent mtDNA synthesis is necessary for the production of oxidized mtDNA fragments after exposure to NLRP3 activators. Cytosolic oxidized mtDNA associates with the NLRP3 inflammasome complex and is required for its activation. The dependence on CMPK2 catalytic activity provides opportunities for more effective control of NLRP3 inflammasome-associated diseases.

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: Newly synthesized mtDNA is required for NLRP3 inflammasome activation.
Fig. 2: IRF1-dependent mtDNA synthesis, ox-mtDNA generation and NLRP3 inflammasome activation.
Fig. 3: CMPK2 controls mtDNA synthesis, ox-mtDNA generation and NLRP3 inflammasome activation.
Fig. 4: CMPK2 catalytic activity is required for NLRP3 inflammasome activation.
Fig. 5: Newly synthesized mtDNA associates with the NLRP3 inflammasome complex.

Similar content being viewed by others

References

  1. Gross, O., Thomas, C. J., Guarda, G. & Tschopp, J. The inflammasome: an integrated view. Immunol. Rev. 243, 136–151 (2011).

    Article  PubMed  CAS  Google Scholar 

  2. Kotas, M. E. & Medzhitov, R. Homeostasis, inflammation, and disease susceptibility. Cell 160, 816–827 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Karin, M. & Clevers, H. Reparative inflammation takes charge of tissue regeneration. Nature 529, 307–315 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  4. Zhong, Z., Sanchez-Lopez, E. & Karin, M. Autophagy, inflammation, and immunity: a troika governing cancer and its treatment. Cell 166, 288–298 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Lu, A. et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell 156, 1193–1206 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Heneka, M. T., Kummer, M. P. & Latz, E. Innate immune activation in neurodegenerative disease. Nat. Rev. Immunol. 14, 463–477 (2014).

    Article  PubMed  CAS  Google Scholar 

  7. Lamkanfi, M. & Dixit, V. M. Inflammasomes and their roles in health and disease. Annu. Rev. Cell Dev. Biol. 28, 137–161 (2012).

    Article  PubMed  CAS  Google Scholar 

  8. Schroder, K. & Tschopp, J. The inflammasomes. Cell 140, 821–832 (2010).

    Article  PubMed  CAS  Google Scholar 

  9. Latz, E., Xiao, T. S. & Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 13, 397–411 (2013).

    Article  PubMed  CAS  Google Scholar 

  10. Nakahira, K. et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 12, 222–230 (2011).

    Article  PubMed  CAS  Google Scholar 

  11. Zhong, Z. et al. NF-κB restricts inflammasome activation via elimination of damaged mitochondria. Cell 164, 896–910 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011).

    Article  ADS  PubMed  CAS  Google Scholar 

  13. Shimada, K. et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36, 401–414 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Jiang, X. & Wang, X. Cytochrome c-mediated apoptosis. Annu. Rev. Biochem. 73, 87–106 (2004).

    Article  PubMed  CAS  Google Scholar 

  15. Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kang, D., Kim, S. H. & Hamasaki, N. Mitochondrial transcription factor A (TFAM): roles in maintenance of mtDNA and cellular functions. Mitochondrion 7, 39–44 (2007).

    Article  PubMed  CAS  Google Scholar 

  17. He, Y., Zeng, M. Y., Yang, D., Motro, B. & Núñez, G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature 530, 354–357 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  18. Shi, H. et al. NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component. Nat. Immunol. 17, 250–258 (2016).

    Article  PubMed  CAS  Google Scholar 

  19. Schmid-Burgk, J. L. et al. A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) screen identifies NEK7 as an essential component of NLRP3 inflammasome activation. J. Biol. Chem. 291, 103–109 (2016).

    Article  PubMed  CAS  Google Scholar 

  20. Jehl, S. P., Nogueira, C. V., Zhang, X. & Starnbach, M. N. IFNγ inhibits the cytosolic replication of Shigella flexneri via the cytoplasmic RNA sensor RIG-I. PLoS Pathog. 8, e1002809 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Xu, Y., Johansson, M. & Karlsson, A. Human UMP-CMP kinase 2, a novel nucleoside monophosphate kinase localized in mitochondria. J. Biol. Chem. 283, 1563–1571 (2008).

    Article  PubMed  CAS  Google Scholar 

  22. Milon, L. et al. The human nm23-H4 gene product is a mitochondrial nucleoside diphosphate kinase. J. Biol. Chem. 275, 14264–14272 (2000).

    Article  PubMed  CAS  Google Scholar 

  23. Chen, Y. L., Lin, D. W. & Chang, Z. F. Identification of a putative human mitochondrial thymidine monophosphate kinase associated with monocytic/macrophage terminal differentiation. Genes Cells 13, 679–689 (2008).

    Article  PubMed  CAS  Google Scholar 

  24. Martinon, F., Mayor, A. & Tschopp, J. The inflammasomes: guardians of the body. Annu. Rev. Immunol. 27, 229–265 (2009).

    Article  PubMed  CAS  Google Scholar 

  25. Elliott, E. I. & Sutterwala, F. S. Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol. Rev. 265, 35–52 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. West, A. P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–480 (2011).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  27. Hudson, G. & Chinnery, P. F. Mitochondrial DNA polymerase-gamma and human disease. Hum. Mol. Genet. 15, R244–R252 (2006).

    Article  PubMed  CAS  Google Scholar 

  28. Kawai, T. & Akira, S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat. Immunol. 11, 373–384 (2010).

    Article  PubMed  CAS  Google Scholar 

  29. Ballana, E. & Esté, J. A. SAMHD1: at the crossroads of cell proliferation, immune responses, and virus restriction. Trends Microbiol. 23, 680–692 (2015).

    Article  PubMed  CAS  Google Scholar 

  30. Yamamoto, M. et al. Role of adaptor TRIF in the MyD88-independent Toll-like receptor signaling pathway. Science 301, 640–643 (2003).

    Article  ADS  PubMed  CAS  Google Scholar 

  31. Zhong, Z. et al. TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nat. Commun. 4, 1611 (2013).

    Article  PubMed  CAS  Google Scholar 

  32. Hornung, V. et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 9, 847–856 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Malik, A. N., Czajka, A. & Cunningham, P. Accurate quantification of mouse mitochondrial DNA without co-amplification of nuclear mitochondrial insertion sequences. Mitochondrion 29, 59–64 (2016).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank eBioscience, Cell Signaling Technologies, Santa Cruz Technologies, and Thermo Fisher for gifts of reagents, and N. Yan and J. Rehwinkel for SAMHD1-deficient murine bone marrow. Z.Z. was supported by Cancer Research Institute (CRI) Irvington Fellowship, Prevent Cancer Foundation Board of Directors Research Fund, and American Association for the Study of Liver Diseases Pinnacle Research Award; F.H. was supported by Eli Lilly LIFA program; S.S. was supported by fellowships from CRI-Irvington and Prostate Cancer Foundation; J.W. was supported by a Canadian Institutes of Health Research fellowship (MFE-135425). Research was supported by NIH grants AI043477 and CA163798 to M.K., AA020172 and DK085252 to E.S., ES010337 to M.K. and E.S., DK109724 and P30DK063491 to A.L.H., DK099205, DK101737 and DK111866 to T.K., AA022614 to T.K. and M.K., Leukemia and Lymphoma Society SCOR grant 20132569 to T. Kipps and M.K., and the Alliance for Lupus Research grant 257214 and CART Foundation to M.K., who is an American Cancer Research Society Professor and holds the Ben and Wanda Hildyard Chair for Mitochondrial and Metabolic Diseases.

Reviewer information

Nature thanks M. Arditi, M. Murphy and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Z.Z., S.L. and M.K. conceived the project. Z.Z. and S.L. designed and performed most of the experiments. E.S.-L., F.H., S.S., X-J.L., J.W. and S.D. provided technical assistance. E.S., B.S., A.L.H., T.K. and H.G. provided reagents and research tools. Z.Z., S.L. and M.K. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Michael Karin.

Ethics declarations

Competing interests

The University of California San Diego is in the process of applying for a patent application (US Provisional Application Serial no. 62/690,175) covering the use of IRF1 and/or CMPK2 genetic/chemical inhibitors to treat NLRP3 inflammasome-associated diseases that lists Z.Z. and M.K. as inventors.

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 TFAM is required for ox-mtDNA generation and NLRP3 inflammasome activation.

a, Inflammasome activator-induced changes in mitochondrial membrane potential (Ψm) in non- or LPS-primed wild-type BMDMs were measured by TMRM fluorescence. Data are mean ± s.d. (n = 3 biological replicates). b, Relative mtROS amounts were measured by MitoSOX fluorescence in non- or LPS-primed wild-type BMDMs after stimulation with different inflammasome activators. Data are mean ± s.d. (n = 3 biological replicates). c, Amounts of 8-OH-dG in mtDNA isolated from the mitochondrial fraction of non- or LPS-primed wild-type BMDMs that were treated with different inflammasome activators. Data are mean ± s.d. (n = 3 biological replicates). d, Cytosolic release of mtDNA, determined by qPCR with primers specific for mtDNA (non-NUMT) and nDNA (B2m), in non- or LPS-primed wild-type BMDMs after treatment with different inflammasome activators. Data are mean ± s.d. (n = 4 biological replicates). e, Immunoblot analysis of TFAM in Tfamf/f and TfamΔMye BMDMs. Results are typical of three independent experiments. f, Relative total mtDNA amounts in Tfamf/f and TfamΔΜye BMDMs determined by qPCR with primers specific for mtDNA (D-loop, non-NUMT) and nDNA (Tert, B2m). Data are mean ± s.d. (n = 3 biological replicates). g, Relative mtROS amounts were measured by MitoSOX fluorescence in LPS-primed Tfamf/f and TfamΔMye BMDMs after stimulation with indicated NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). h, Amounts of 8-OH-dG in mtDNA isolated from the mitochondrial fraction of LPS-primed Tfamf/f and TfamΔMye BMDMs that were stimulated with various NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). i, Immunoblot analysis of Casp1 p20 and mature IL-1β (p17) in culture supernatants of Tfamf/f (W) and TfamΔMye (K) BMDMs that were stimulated with LPS plus different inflammasome activators. Results are typical of three separate experiments. j, Immunoblot analysis of pro-IL-1β, NLRP3, ASC and pro-Casp1 in the lysates of Tfamf/f and TfamΔMye BMDMs before and after LPS priming. Results are typical of three separate experiments. k, l, Amounts of IL-1β (k) and TNF (l) in culture supernatants of LPS-primed Tfamf/f and TfamΔMye BMDMs that were stimulated with various NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). m, Amounts of IL-1β in culture supernatants of LPS-primed Tfamf/f and TfamΔMye BMDMs that were stimulated with H2O2 in the absence and presence of nigericin. Data are mean ± s.d. (n = 3 biological replicates). **P < 0.01; ***P < 0.001; two-sided unpaired t-test.

Source Data

Extended Data Fig. 2 ox-mtDNA activates the NLRP3 inflammasome.

a, Immunoblot analysis of NLRP3, AIM2 and Polγ in shCtrl- or specific shRNA-transduced BMDMs. Data are typical of three separate experiments. b, Amounts of IL-1β in culture supernatants of LPS-primed TfamΔMye BMDMs that were transfected with mtDNA, methylated mtDNA, ox-mtDNA, and methylated ox-mtDNA. All mtDNAs were 90-bp long and of an identical sequence. Data are mean ± s.d. (n = 3 biological replicates). c, Representative fluorescent microscopy images of wild-type BMDMs that were co-stained for 8-OH-dG, ASC and DAPI before and after stimulation with LPS plus the indicated inflammasome activators. Results are typical of three independent experiments. Scale bars, 5 μm.

Source Data

Extended Data Fig. 3 LPS induces mtDNA replication.

a, Time-course analysis of total mtDNA amounts in wild-type BMDMs after LPS (200 ng ml−1) stimulation. Data are mean ± s.d. (n = 3 biological replicates). Results are typical of three separate experiments. b, Representative fluorescent microscopy images of EdU-labelled shCtrl- or shPolg-transduced wild-type BMDMs that were stimulated with or without LPS (200 ng ml−1) for 6 h. Scale bars, 5 μm. Results are typical of three separate experiments. c, Percentages of cells with mtDNA replication as determined in b. Data are mean ± s.d. (n = 3 different microscopic fields per group; original magnification, ×40). d, Immunoblot analysis of mitochondrial resident proteins, TOM20 and VDAC, in wild-type BMDMs before and after LPS stimulation. Results are typical of three separate experiments. e, Representative fluorescent microscopy images of the mitochondrial resident protein ATP5B and DAPI staining in wild-type BMDMs before and after LPS stimulation. Results are typical of three independent experiments. Arrows indicate fragmented mitochondria. ***P < 0.001, two-sided unpaired t-test.

Source Data

Extended Data Fig. 4 MyD88 and TRIF mediate LPS-induced IRF1 and CMPK2 expression and NLRP3 inflammasome activation.

a, Amounts of 8-OH-dG in mtDNA isolated from mitochondrial fractions of wild-type, Myd88−/− and Trif−/− BMDMs that were primed with LPS for different durations followed by stimulation with ATP. Data are mean ± s.d. (n = 3 biological replicates per time point). b, c, Quantification of fluorescent microscopy images of ASC puncta (b) and ASC puncta positive for EdU (c) in wild-type, Myd88−/− and Trif−/− BMDMs that were primed with LPS for different durations, followed by ATP stimulation. Data are mean ± s.d. (n = 3 different microscopic fields per group; original magnification, ×40). df, Immunoblot analysis of NLRP3, pro-IL-1β, IRF1 and CMPK2 in wild-type, Myd88−/− and Trif−/− BMDMs before and after LPS stimulation. Data are typical of three separate experiments. *P < 0.05; **P < 0.01; ***P < 0.001; two-sided unpaired t-test.

Source Data

Extended Data Fig. 5 Effects of IRF1 on priming, mitochondrial damage and non-canonical inflammasome activation.

a, Immunoblot analysis of NLRP3, ASC, pro-caspase-1, pro-IL-1β, NEK7 and IRF1 in the lysates of wild-type and Irf1−/− BMDMs before and after LPS stimulation. Data are typical of three separate experiments. b, Amounts of TNF in culture supernatants of LPS-primed wild-type and Irf1−/− BMDMs that were stimulated with the indicated inflammasome activators. Data are mean ± s.d. (n = 3 biological replicates). c, Percentages of Ψm preservation were measured by TMRM fluorescence in LPS-primed wild-type and Irf1−/− BMDMs that were stimulated with indicated NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). d, Relative amounts of mtROS were measured by MitoSOX fluorescence in LPS-primed wild-type and Irf1−/− BMDMs that were stimulated with indicated NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). e, Immunoblot analysis of caspase-11 and IRF1 in lysates from wild-type and Irf1−/− BMDMs before and after LPS stimulation. Results are typical of three independent experiments. f, Amounts of IL-1β in culture supernatants of LPS-primed wild-type and Irf1−/− BMDMs that were further transfected with FuGENE-complexed LPS. Data are mean ± s.d. (n = 3 biological replicates).

Source Data

Extended Data Fig. 6 IRF1 mediates LPS-induced CMPK2 expression.

a, Relative amounts of Cmpk2 mRNA in wild-type and Irf1−/− BMDMs before and after LPS stimulation. Data are mean ± s.d. (n = 3 biological replicates per time point). b, Immunoblot analysis of CMPK2, VDAC and tubulin in mitochondrial and cytosolic fractions of wild-type BMDMs after LPS stimulation. Results are typical of three separate experiments. c, Chromatin immunoprecipitation analysis of IRF1 recruitment to the Cmpk2 promoter. Data are mean ± s.d. (n = 4 biological replicates for wild-type and Irf1−/− groups; n = 5 and 6 biological replicates for wild-type + LPS and Irf1−/− + LPS groups, respectively). Cxcl10, a known IRF1-target gene, was included as a positive control. d, Relative mRNA amounts of dGK (also known as Dguok), Tk2, Ak2, Nme4 and Polg in wild-type BMDMs before and after 6 h LPS stimulation. Data are mean ± s.d. (n = 3 biological replicates). e, Immunoblot analysis of the enzymes encoded by the genes in d in the lysates of wild-type BMDMs before and after LPS stimulation. Results are typical of three independent experiments. f, Immunoblot analysis of CMPK2 and NME4 in shCtrl- or specific shRNA-transduced BMDMs. Data are typical of three independent experiments. *P < 0.05; **P < 0.01; two-sided unpaired t-test.

Source Data

Extended Data Fig. 7 CMPK2 deficiency does not affect inflammasome subunit expression nor NLRP3 activator-induced mitochondrial damage.

a, Immunoblot analysis of pro-IL-1β, NLRP3, ASC, pro-Casp1 and NEK7 in the lysates of wild-type (shCtrl) and CMPK2-deficient (shCmpk2) BMDMs before and after LPS priming. Results are typical of three separate experiments. b, NLRP3 activator-induced changes in Ψm in LPS-primed shCtrl and shCmpk2 BMDMs were measured by TMRM fluorescence. Data are mean ± s.d. (n = 3 biological replicates). c, Relative amounts of mtROS measured by MitoSOX fluorescence in LPS-primed shCtrl and shCmpk2 BMDMs after stimulation with the indicated NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). d, Representative fluorescent microscopy images of EdU-labelled wild-type BMDMs transduced with either shCtrl- or shCmpk2-encoding lentiviruses that were stimulated with or without LPS (200 ng ml−1) for 6 h. Scale bars, 5 μm. e, Percentages of cells with mtDNA replication as determined in d. Data are mean ± s.d. (n = 3 different microscopic fields per group; original magnification, ×40). f, Relative amounts of total mtDNA in wild-type and Irf1−/− BMDMs before and after treatments with the indicated TLR agonists. Data are mean ± s.d. (n = 3 biological replicates). g, Relative amounts of total mtDNA in shCtrl- or shCmpk2-encoding lentivirus-transduced wild-type BMDMs before and after treatments with the indicated TLR agonists. Data are mean ± s.d. (n = 3 biological replicates). *P < 0.05; **P < 0.01; ***P < 0.001; two-sided unpaired t-test.

Source Data

Extended Data Fig. 8 dNTP availability controls LPS-induced mtDNA synthesis and NLRP3 inflammasome activation.

a, Relative total mtDNA amounts in shCtrl and shNme4 BMDMs before and after LPS priming. Data are mean ± s.d. (n = 3 biological replicates). b, Amounts of 8-OH-dG in mtDNA isolated from the mitochondrial fraction of LPS-primed shCtrl and shNme4 BMDMs that were stimulated with various NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). c, Amounts of 8-OH-dG in cytosolic mtDNA from LPS-primed shCtrl and shNme4 BMDMs that were stimulated with various NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates). d, e, Amounts of IL-1β (d) and TNF (e) in supernatants of LPS-primed shCtrl and shNme4 BMDMs that were stimulated with various inflammasome activators. Data are mean ± s.d. (n = 3 biological replicates). f, Immunoblot analysis of SAMHD1 and Polγ in wild-type and Trif−/− BMDMs that were stimulated with LPS for different durations as indicated. Results are typical of three independent experiments. g, Relative total mtDNA amounts in wild-type and Samhd1−/− BMDMs before and after LPS stimulation. Data are mean ± s.d. (n = 3 biological replicates). h, i, Amounts of IL-1β (h) and TNF (i) in the culture supernatants of LPS-primed wild-type and Samhd1−/− BMDMs that were stimulated with inflammasome activators as indicated. Data are mean ± s.d. (n = 3 biological replicates). j, Amounts of NLRP3 activator-induced IL-1β in culture supernatants of LPS-primed wild-type and Samhd1−/− BMDMs with or without Polg expression. Data are mean ± s.d. (n = 3 biological replicates). *P < 0.05; **P < 0.01; ***P < 0.001; two-sided unpaired t-test.

Source Data

Extended Data Fig. 9 CMPK2 expression restores NLRP3 inflammasome activation in IRF1-deficient macrophages.

a, Immunoblot analysis of IRF1, CMPK2, pro-IL-1β, NLRP3, ASC and pro-Casp1 in lysates of wild-type and Irf1−/− BMDMs before and after transduction with a wild-type CMPK2-encoding lentivirus. Results are typical of three independent experiments. b, NLRP3 activator-induced changes in Ψm in LPS-primed CMPK2-transduced wild-type and Irf1−/− BMDMs were measured by TMRM fluorescence. Data are mean ± s.d. (n = 3 biological replicates) and analysed by two-sided unpaired t-test (not significant). c, Relative amounts of mtROS measured by MitoSOX fluorescence in LPS-primed control (vector)- or CMPK2-transduced wild-type and Irf1−/− BMDMs before and after stimulation with NLRP3 activators. Data are mean ± s.d. (n = 3 biological replicates) and analysed by two-sided unpaired t-test (not significant). d, Immunoblot analysis of CMPK2 in Irf1−/− BMDMs that were transduced with wild-type or mutant (CMPK2(D330A)) CMPK2-encoding lentiviruses. Results are typical of three separate experiments.

Source Data

Extended Data Fig. 10 IRF1 is required for in vivo mtDNA replication and NLRP3 inflammasome activation.

a, b, 12-week-old wild-type or Irf1−/− mice were injected intraperitoneally with LPS (50 mg per kg of body weight) and their sera were collected 3 h later and analysed by ELISA for IL-1β (a) and TNF (b). Results are mean ± s.d. (n = 6 and 7 for WT and Irf1−/− mice, respectively). c, Survival of wild-type or Irf1−/− mice that were injected intraperitoneally with LPS (50 mg per kg body weight; n = 10 and 11 for WT and Irf1−/− mice, respectively). d, Relative amounts of total mtDNA in peritoneal infiltrates of wild-type or Irf1−/− mice before and after LPS (50 mg per kg body weight) injection. Data are mean ± s.d. (n = 6 in PBS-treated groups; n = 12 in LPS-treated groups). e, Peritoneal IL-1β in wild-type or Irf1−/− mice 4 h after intraperitoneal injection of alum (1 mg) or PBS. Data are mean ± s.d. (n = 5 in PBS-treated groups; n = 6 in alum-treated groups). f, g, Alum-induced peritoneal infiltration of neutrophils (CD11b+Ly6G+F4/80) (f) and monocytes (CD11b+Ly6C+Ly6G) (g) in wild-type and Irf1−/− mice 12 h after alum (1 mg) or PBS injection. Data are mean ± s.e.m. (n = 3 for PBS-treated groups and n = 6 for alum-treated groups). ***P < 0.001; two-sided unpaired t-test (a, b, dg) and log-rank test (c). h, A working model to illustrate how TLR-mediated priming controls mtDNA replication and NLRP3 inflammasome activation. Whereas IRF1 acts positively to induce the transcription of CMPK2, which supplies rate-limiting dCDP for mtDNA synthesis, TRIF-dependent signalling also acts negatively to limit dNTP supply through the induction of SAMHD1.

Source Data

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1 and 2.

Reporting Summary

Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhong, Z., Liang, S., Sanchez-Lopez, E. et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560, 198–203 (2018). https://doi.org/10.1038/s41586-018-0372-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0372-z

This article is cited by

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

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