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.

  • Review Article
  • Published:

Regulation of cGAS- and RLR-mediated immunity to nucleic acids

Abstract

Pathogen-derived nucleic acids are crucial signals for innate immunity. Despite the structural similarity between those and host nucleic acids, mammalian cells have been able to evolve powerful innate immune signaling pathways that originate from the detection of cytosolic nucleic acid species, one of the most prominent being the cGAS–STING pathway for DNA and the RLR–MAVS pathway for RNA, respectively. Recent advances have revealed a plethora of regulatory mechanisms that are crucial for balancing the activity of nucleic acid sensors for the maintenance of overall cellular homeostasis. Elucidation of the various mechanisms that enable cells to maintain control over the activity of cytosolic nucleic acid sensors has provided new insight into the pathology of human diseases and, at the same time, offers a rich and largely unexplored source for new therapeutic targets. This Review addresses the emerging literature on regulation of the sensing of cytosolic DNA and RNA via cGAS and RLRs.

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: Sensing of dsDNA by the cGAS–STING pathway.
Fig. 2: The RLR signal-transduction pathway.
Fig. 3: Regulatory mechanism that acts on DNA-mediated activation of cGAS.

Similar content being viewed by others

References

  1. Barbalat, R., Ewald, S. E., Mouchess, M. L. & Barton, G. M. Nucleic acid recognition by the innate immune system. Annu. Rev. Immunol. 29, 185–214 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Paludan, S. R. & Bowie, A. G. Immune sensing of DNA. Immunity 38, 870–880 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ablasser, A. & Chen, Z. J. cGAS in action: expanding roles in immunity and inflammation. Science 363, eaat8657 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Hur, S. Double-stranded RNA sensors and modulators in innate immunity. Annu. Rev. Immunol. 37, 349–375 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Roers, A., Hiller, B. & Hornung, V. Recognition of endogenous nucleic acids by the innate immune system. Immunity 44, 739–754 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Woo, S. R. et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ishizuka, J. J. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 565, 43–48 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Glück, S. & Ablasser, A. Innate immunosensing of DNA in cellular senescence. Curr. Opin. Immunol. 56, 31–36 (2019).

    Article  PubMed  CAS  Google Scholar 

  9. Crowl, J. T., Gray, E. E., Pestal, K., Volkman, H. E. & Stetson, D. B. Intracellular nucleic acid detection in autoimmunity. Annu. Rev. Immunol. 35, 313–336 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Ablasser, A. et al. cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Diner, E. J. et al. The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell Rep. 3, 1355–1361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gao, P. et al. Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153, 1094–1107 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang, X. et al. Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol. Cell 51, 226–235 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Kato, K., Omura, H., Ishitani, R. & Nureki, O. Cyclic GMP-AMP as an endogenous second messenger in innate immune signaling by cytosolic DNA. Annu. Rev. Biochem. 86, 541–566 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Li, X. et al. Cyclic GMP-AMP synthase is activated by double-stranded DNA-induced oligomerization. Immunity 39, 1019–1031 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Zhang, X. et al. The cytosolic DNA sensor cGAS forms an oligomeric complex with DNA and undergoes switch-like conformational changes in the activation loop. Cell Rep. 6, 421–430 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Andreeva, L. et al. cGAS senses long and HMGB/TFAM-bound U-turn DNA by forming protein-DNA ladders. Nature 549, 394–398 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Hooy, R. M. & Sohn, J. The allosteric activation of cGAS underpins its dynamic signaling landscape. eLife 7, e39984 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Du, M. & Chen, Z. J. DNA-induced liquid phase condensation of cGAS activates innate immune signaling. Science 361, 704–709 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Tao, J. et al. Nonspecific DNA Binding of cGAS N terminus promotes cGAS activation. J. Immunol. 198, 3627–3636 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Xie, W. et al. Human cGAS catalytic domain has an additional DNA-binding interface that enhances enzymatic activity and liquid-phase condensation. Proc. Natl Acad. Sci. USA 116, 11946–11955 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ablasser, A. et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bridgeman, A. et al. Viruses transfer the antiviral second messenger cGAMP between cells. Science 349, 1228–1232 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gentili, M. et al. Transmission of innate immune signaling by packaging of cGAMP in viral particles. Science 349, 1232–1236 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Ritchie, C., Cordova, A. F., Hess, G. T., Bassik, M. C. & Li, L. SLC19A1 is an importer of the immunotransmitter cGAMP. Mol. Cell 75, 372–381.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Carozza, J.A. et al. 2′3′-cGAMP is an immunotransmitter produced by cancer cells and regulated by ENPP1. Preprint at https://doi.org/10.1101/539312 (2019).

  28. Ergun, S. L., Fernandez, D., Weiss, T. M. & Li, L. STING polymer structure reveals mechanisms for activation, hyperactivation, and inhibition. Cell 178, 290–301.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Shang, G., Zhang, C., Chen, Z. J., Bai, X. C. & Zhang, X. Cryo-EM structures of STING reveal its mechanism of activation by cyclic GMP-AMP. Nature 567, 389–393 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gui, X. et al. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 567, 262–266 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Dobbs, N. et al. STING activation by translocation from the ER is associated with infection and autoinflammatory disease. Cell Host Microbe 18, 157–168 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Stempel, M., Chan, B. & Brinkmann, M. M. Coevolution pays off: herpesviruses have the license to escape the DNA sensing pathway. Med. Microbiol. Immunol. 208, 495–512 (2019).

    Article  PubMed  CAS  Google Scholar 

  33. Zhang, C. et al. Structural basis of STING binding with and phosphorylation by TBK1. Nature 567, 394–398 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, B. et al. A conserved PLPLRT/SD motif of STING mediates the recruitment and activation of TBK1. Nature 569, 718–722 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu, S. et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 347, aaa2630 (2015).

    Article  PubMed  CAS  Google Scholar 

  36. Saitoh, T. et al. Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response. Proc. Natl Acad. Sci. USA 106, 20842–20846 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gulen, M. F. et al. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 8, 427 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Larkin, B. et al. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J. Immunol. 199, 397–402 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Fang, R. et al. MAVS activates TBK1 and IKKε through TRAFs in NEMO dependent and independent manner. PLoS Pathog. 13, e1006720 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Lei, Y. et al. MAVS-mediated apoptosis and its inhibition by viral proteins. PLoS One 4, e5466 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Guan, K. et al. MAVS regulates apoptotic cell death by decreasing K48-linked ubiquitination of voltage-dependent anion channel 1. Mol. Cell. Biol. 33, 3137–3149 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Huang, Y. et al. MAVS-MKK7-JNK2 defines a novel apoptotic signaling pathway during viral infection. PLoS Pathog. 10, e1004020 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Yoneyama, M. et al. Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J. Immunol. 175, 2851–2858 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Rothenfusser, S. et al. The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I. J. Immunol. 175, 5260–5268 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Saito, T. et al. Regulation of innate antiviral defenses through a shared repressor domain in RIG-I and LGP2. Proc. Natl Acad. Sci. USA 104, 582–587 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Venkataraman, T. et al. Loss of DExD/H box RNA helicase LGP2 manifests disparate antiviral responses. J. Immunol. 178, 6444–6455 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Satoh, T. et al. LGP2 is a positive regulator of RIG-I- and MDA5-mediated antiviral responses. Proc. Natl Acad. Sci. USA 107, 1512–1517 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. van der Veen, A. G. et al. The RIG-I-like receptor LGP2 inhibits Dicer-dependent processing of long double-stranded RNA and blocks RNA interference in mammalian cells. EMBO J. 37, e97479 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Bruns, A. M., Leser, G. P., Lamb, R. A. & Horvath, C. M. The innate immune sensor LGP2 activates antiviral signaling by regulating MDA5-RNA interaction and filament assembly. Mol. Cell 55, 771–781 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yoneyama, M. et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 5, 730–737 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Kowalinski, E. et al. Structural basis for the activation of innate immune pattern-recognition receptor RIG-I by viral RNA. Cell 147, 423–435 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Jiang, X. et al. Ubiquitin-induced oligomerization of the RNA sensors RIG-I and MDA5 activates antiviral innate immune response. Immunity 36, 959–973 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wu, B. et al. Structural basis for dsRNA recognition, filament formation, and antiviral signal activation by MDA5. Cell 152, 276–289 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Peisley, A., Wu, B., Xu, H., Chen, Z. J. & Hur, S. Structural basis for ubiquitin-mediated antiviral signal activation by RIG-I. Nature 509, 110–114 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Peisley, A., Wu, B., Yao, H., Walz, T. & Hur, S. RIG-I forms signaling-competent filaments in an ATP-dependent, ubiquitin-independent manner. Mol. Cell 51, 573–583 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Cadena, C. et al. Ubiquitin-dependent and -independent roles of E3 Ligase RIPLET in innate immunity. Cell 177, 1187–1200.e16 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wu, B. et al. Molecular imprinting as a signal-activation mechanism of the viral RNA sensor RIG-I. Mol. Cell 55, 511–523 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hou, F. et al. MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell 146, 448–461 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Luecke, S. et al. cGAS is activated by DNA in a length-dependent manner. EMBO Rep. 18, 1707–1715 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gehrke, N. et al. Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity 39, 482–495 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Steinhagen, F. et al. Suppressive oligodeoxynucleotides containing TTAGGG motifs inhibit cGAS activation in human monocytes. Eur. J. Immunol. 48, 605–611 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Lahaye, X. et al. NONO detects the nuclear HIV capsid to promote cGAS-mediated innate immune activation. Cell 175, 488–501.e22 (2018).

    Article  CAS  PubMed  Google Scholar 

  63. Zierhut, C. et al. The cytoplasmic DNA sensor cGAS promotes mitotic cell death. Cell 178, 302–315.e23 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Stetson, D. B., Ko, J. S., Heidmann, T. & Medzhitov, R. Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell 134, 587–598 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Crow, Y. J. & Manel, N. Aicardi-Goutières syndrome and the type I interferonopathies. Nat. Rev. Immunol. 15, 429–440 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Uggenti, C., Lepelley, A. & Crow, Y. J. Self-awareness: nucleic acid-driven inflammation and the type I interferonopathies. Annu. Rev. Immunol. 37, 247–267 (2019).

    Article  CAS  PubMed  Google Scholar 

  67. Crow, Y. J. et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 cause Aicardi-Goutières syndrome at the AGS1 locus. Nat. Genet. 38, 917–920 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Rice, G. I. et al. Mutations involved in Aicardi-Goutières syndrome implicate SAMHD1 as regulator of the innate immune response. Nat. Genet. 41, 829–832 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Crow, Y. J. et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutières syndrome and mimic congenital viral brain infection. Nat. Genet. 38, 910–916 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Lindahl, T., Gally, J. A. & Edelman, G. M. Properties of deoxyribonuclease 3 from mammalian tissues. J. Biol. Chem. 244, 5014–5019 (1969).

    Article  CAS  PubMed  Google Scholar 

  71. Goldstone, D. C. et al. HIV-1 restriction factor SAMHD1 is a deoxynucleoside triphosphate triphosphohydrolase. Nature 480, 379–382 (2011).

    Article  CAS  PubMed  Google Scholar 

  72. Reijns, M. A. et al. Enzymatic removal of ribonucleotides from DNA is essential for mammalian genome integrity and development. Cell 149, 1008–1022 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Gall, A. et al. Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity 36, 120–131 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ablasser, A. et al. TREX1 deficiency triggers cell-autonomous immunity in a cGAS-dependent manner. J. Immunol. 192, 5993–5997 (2014).

    Article  CAS  PubMed  Google Scholar 

  75. Gao, D. et al. Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. Proc. Natl Acad. Sci. USA 112, E5699–E5705 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Gray, E. E., Treuting, P. M., Woodward, J. J. & Stetson, D. B. Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi-Goutières syndrome. J. Immunol. 195, 1939–1943 (2015).

    Article  CAS  PubMed  Google Scholar 

  77. Mackenzie, K. J. et al. Ribonuclease H2 mutations induce a cGAS/STING-dependent innate immune response. EMBO J. 35, 831–844 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Maelfait, J., Bridgeman, A., Benlahrech, A., Cursi, C. & Rehwinkel, J. Restriction by SAMHD1 limits cGAS/STING-dependent innate and adaptive immune responses to HIV-1. Cell Rep. 16, 1492–1501 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Pokatayev, V. et al. RNase H2 catalytic core Aicardi-Goutières syndrome-related mutant invokes cGAS-STING innate immune-sensing pathway in mice. J. Exp. Med. 213, 329–336 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Volkman, H. E. & Stetson, D. B. The enemy within: endogenous retroelements and autoimmune disease. Nat. Immunol. 15, 415–422 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Rice, G. I. et al. Reverse-transcriptase inhibitors in the Aicardi–Goutières syndrome. N. Engl. J. Med. 379, 2275–2277 (2018).

    Article  PubMed  Google Scholar 

  82. Yang, Y. G., Lindahl, T. & Barnes, D. E. Trex1 exonuclease degrades ssDNA to prevent chronic checkpoint activation and autoimmune disease. Cell 131, 873–886 (2007).

    Article  CAS  PubMed  Google Scholar 

  83. Mackenzie, K. J. et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Coquel, F. et al. SAMHD1 acts at stalled replication forks to prevent interferon induction. Nature 557, 57–61 (2018).

    Article  CAS  PubMed  Google Scholar 

  85. Gratia, M. et al. Bloom syndrome protein restrains innate immune sensing of micronuclei by cGAS. J. Exp. Med. 216, 1199–1213 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kawane, K. et al. Chronic polyarthritis caused by mammalian DNA that escapes from degradation in macrophages. Nature 443, 998–1002 (2006).

    Article  CAS  PubMed  Google Scholar 

  87. Ahn, J., Gutman, D., Saijo, S. & Barber, G. N. STING manifests self DNA-dependent inflammatory disease. Proc. Natl Acad. Sci. USA 109, 19386–19391 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Rodero, M. P. et al. Type I interferon-mediated autoinflammation due to DNase II deficiency. Nat. Commun. 8, 2176 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Lan, Y. Y., Londoño, D., Bouley, R., Rooney, M. S. & Hacohen, N. Dnase2a deficiency uncovers lysosomal clearance of damaged nuclear DNA via autophagy. Cell Rep. 9, 180–192 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Sisquella, X. et al. Malaria parasite DNA-harbouring vesicles activate cytosolic immune sensors. Nat. Commun. 8, 1985 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Nandakumar, R. et al. Intracellular bacteria engage a STING-TBK1-MVB12b pathway to enable paracrine cGAS-STING signalling. Nat. Microbiol. 4, 701–713 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. West, A. P. et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520, 553–557 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Aguirre, S. et al. Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection. Nat. Microbiol. 2, 17037 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Sliter, D. A. et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. McArthur, K. et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science 359, eaao6047 (2018).

    Article  PubMed  CAS  Google Scholar 

  96. Rongvaux, A. et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell 159, 1563–1577 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. White, M. J. et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wiens, K. E. & Ernst, J. D. The mechanism for type I interferon induction by Mycobacterium tuberculosis is bacterial strain-dependent. PLoS Pathog. 12, e1005809 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Hatch, E. M., Fischer, A. H., Deerinck, T. J. & Hetzer, M. W. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 154, 47–60 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Liu, S. et al. Nuclear envelope assembly defects link mitotic errors to chromothripsis. Nature 561, 551–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Harding, S. M. et al. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548, 466–470 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ivanov, A. et al. Lysosome-mediated processing of chromatin in senescence. J. Cell Biol. 202, 129–143 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Dou, Z. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Glück, S. et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 19, 1061–1070 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Yang, H., Wang, H., Ren, J., Chen, Q. & Chen, Z. J. cGAS is essential for cellular senescence. Proc. Natl Acad. Sci. USA 114, E4612–E4620 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Härtlova, A. et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 42, 332–343 (2015).

    Article  PubMed  CAS  Google Scholar 

  107. Wolf, C. et al. RPA and Rad51 constitute a cell intrinsic mechanism to protect the cytosol from self DNA. Nat. Commun. 7, 11752 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Hornung, V. et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 314, 994–997 (2006).

    Article  PubMed  Google Scholar 

  109. Pichlmair, A. et al. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5′-phosphates. Science 314, 997–1001 (2006).

    Article  CAS  PubMed  Google Scholar 

  110. Schlee, M. et al. Recognition of 5′ triphosphate by RIG-I helicase requires short blunt double-stranded RNA as contained in panhandle of negative-strand virus. Immunity 31, 25–34 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Kato, H. et al. Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5. J. Exp. Med. 205, 1601–1610 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ahmad, S. et al. Breaching self-tolerance to Alu duplex RNA underlies MDA5-mediated inflammation. Cell 172, 797–810.e13 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Fitzgerald, M. E., Rawling, D. C., Vela, A. & Pyle, A. M. An evolving arsenal: viral RNA detection by RIG-I-like receptors. Curr. Opin. Microbiol. 20, 76–81 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Sohn, J. & Hur, S. Filament assemblies in foreign nucleic acid sensors. Curr. Opin. Struct. Biol. 37, 134–144 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Schuberth-Wagner, C. et al. A conserved histidine in the RNA sensor RIG-I controls immune tolerance to N1–2’O-methylated self RNA. Immunity 43, 41–51 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Devarkar, S. C. et al. Structural basis for m7G recognition and 2′-O-methyl discrimination in capped RNAs by the innate immune receptor RIG-I. Proc. Natl Acad. Sci. USA 113, 596–601 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Uzri, D. & Gehrke, L. Nucleotide sequences and modifications that determine RIG-I/RNA binding and signaling activities. J. Virol. 83, 4174–4184 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Chen, Y. G. et al. Sensing self and foreign circular RNAs by intron identity. Mol. Cell 67, 228–238.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Chen, Y. G. et al. N6-methyladenosine modification controls circular RNA immunity. Mol. Cell 76, 96–109.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mu, X., Greenwald, E., Ahmad, S. & Hur, S. An origin of the immunogenicity of in vitro transcribed RNA. Nucleic Acids Res. 46, 5239–5249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Mannion, N. M. et al. The RNA-editing enzyme ADAR1 controls innate immune responses to RNA. Cell Rep. 9, 1482–1494 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Liddicoat, B. J. et al. RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself. Science 349, 1115–1120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Pestal, K. et al. Isoforms of RNA-editing enzyme ADAR1 independently control nucleic acid sensor MDA5-driven autoimmunity and multi-organ development. Immunity 43, 933–944 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Rice, G. I. et al. Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a type I interferon signature. Nat. Genet. 44, 1243–1248 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Züst, R. et al. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 12, 137–143 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Dong, H. et al. Flavivirus RNA methylation. J. Gen. Virol. 95, 763–778 (2014).

    Article  CAS  PubMed  Google Scholar 

  127. Nabet, B. Y. et al. Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer. Cell 170, 352–366.e13 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Ranoa, D. R. et al. Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs. Oncotarget 7, 26496–26515 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Chiang, J. J. et al. Viral unmasking of cellular 5S rRNA pseudogene transcripts induces RIG-I-mediated immunity. Nat. Immunol. 19, 53–62 (2018).

    Article  CAS  PubMed  Google Scholar 

  130. Eckard, S. C. et al. The SKIV2L RNA exosome limits activation of the RIG-I-like receptors. Nat. Immunol. 15, 839–845 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Chiappinelli, K. B. et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 162, 974–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Roulois, D. et al. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 162, 961–973 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Cuellar, T. L. et al. Silencing of retrotransposons by SETDB1 inhibits the interferon response in acute myeloid leukemia. J. Cell Biol. 216, 3535–3549 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Sheng, W. et al. LSD1 ablation stimulates anti-tumor immunity and enables checkpoint blockade. Cell 174, 549–563.e19 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Hayman, T. J. et al. RIPLET, and not TRIM25, is required for endogenous RIG-I-dependent antiviral responses. Immunol. Cell Biol. 97, 840–852 (2019).

    Article  CAS  PubMed  Google Scholar 

  137. Zhou, W. et al. Structure of the Human cGAS-DNA complex reveals enhanced control of immune surveillance. Cell 174, 300–311.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Barnett, K. C. et al. Phosphoinositide interactions position cGAS at the plasma membrane to ensure efficient distinction between self- and viral DNA. Cell 176, 1432–1446.e11 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Gentili, M. et al. The N-terminal domain of cGAS determines preferential association with centromeric DNA and innate immune activation in the nucleus. Cell Rep. 26, 3798 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Volkman, H.E., Cambier, S., Gray, E.E. & Stetson, D.B. cGAS is predominantly a nuclear protein. Preprint at https://doi.org/10.1101/486118 (2019).

  141. Jiang, H. et al. Chromatin-bound cGAS is an inhibitor of DNA repair and hence accelerates genome destabilization and cell death. EMBO J. 38, e102718 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wies, E. et al. Dephosphorylation of the RNA sensors RIG-I and MDA5 by the phosphatase PP1 is essential for innate immune signaling. Immunity 38, 437–449 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Binder, M. et al. Molecular mechanism of signal perception and integration by the innate immune sensor retinoic acid-inducible gene-I (RIG-I). J. Biol. Chem. 286, 27278–27287 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Patel, J. R. et al. ATPase-driven oligomerization of RIG-I on RNA allows optimal activation of type-I interferon. EMBO Rep. 14, 780–787 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Devarkar, S. C., Schweibenz, B., Wang, C., Marcotrigiano, J. & Patel, S. S. RIG-I uses an ATPase-powered translocation-throttling mechanism for kinetic proofreading of RNAs and oligomerization. Mol. Cell 72, 355–368.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Peisley, A. et al. Cooperative assembly and dynamic disassembly of MDA5 filaments for viral dsRNA recognition. Proc. Natl Acad. Sci. USA 108, 21010–21015 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Peisley, A. et al. Kinetic mechanism for viral dsRNA length discrimination by MDA5 filaments. Proc. Natl Acad. Sci. USA 109, E3340–E3349 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Wang, W. et al. RNF122 suppresses antiviral type I interferon production by targeting RIG-I CARDs to mediate RIG-I degradation. Proc. Natl Acad. Sci. USA 113, 9581–9586 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Arimoto, K. et al. Negative regulation of the RIG-I signaling by the ubiquitin ligase RNF125. Proc. Natl Acad. Sci. USA 104, 7500–7505 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Hao, Q. et al. A non-canonical role of the p97 complex in RIG-I antiviral signaling. EMBO J. 34, 2903–2920 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Wang, S. et al. RNF123 has an E3 ligase-independent function in RIG-I-like receptor-mediated antiviral signaling. EMBO Rep. 17, 1155–1168 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Narayan, K. et al. TRIM13 is a negative regulator of MDA5-mediated type I interferon production. J. Virol. 88, 10748–10757 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Zhao, C. et al. The E3 ubiquitin ligase TRIM40 attenuates antiviral immune responses by targeting MDA5 and RIG-I. Cell Rep. 21, 1613–1623 (2017).

    Article  CAS  PubMed  Google Scholar 

  154. Zhao, K. et al. Cytoplasmic STAT4 promotes antiviral type I IFN production by blocking CHIP-mediated degradation of RIG-I. J. Immunol. 196, 1209–1217 (2016).

    Article  CAS  PubMed  Google Scholar 

  155. Inn, K. S. et al. Linear ubiquitin assembly complex negatively regulates RIG-I- and TRIM25-mediated type I interferon induction. Mol. Cell 41, 354–365 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Chen, W. et al. Induction of Siglec-G by RNA viruses inhibits the innate immune response by promoting RIG-I degradation. Cell 152, 467–478 (2013).

    Article  CAS  PubMed  Google Scholar 

  157. Takashima, K., Oshiumi, H., Matsumoto, M. & Seya, T. DNAJB1/HSP40 suppresses melanoma differentiation-associated gene 5-mitochondrial antiviral signaling protein function in conjunction with HSP70. J. Innate Immun. 10, 44–55 (2018).

    Article  CAS  PubMed  Google Scholar 

  158. Eaglesham, J. B., Pan, Y., Kupper, T. S. & Kranzusch, P. J. Viral and metazoan poxins are cGAMP-specific nucleases that restrict cGAS-STING signalling. Nature 566, 259–263 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Li, L. et al. Hydrolysis of 2‘3’-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat. Chem. Biol. 10, 1043–1048 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Luteijn, R. D. et al. SLC19A1 transports immunoreactive cyclic dinucleotides. Nature 573, 434–438 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Mukai, K. et al. Activation of STING requires palmitoylation at the Golgi. Nat. Commun. 7, 11932 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Haag, S. M. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018).

    Article  CAS  PubMed  Google Scholar 

  163. Jeremiah, N. et al. Inherited STING-activating mutation underlies a familial inflammatory syndrome with lupus-like manifestations. J. Clin. Invest. 124, 5516–5520 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Liu, Y. et al. Activated STING in a vascular and pulmonary syndrome. N. Engl. J. Med. 371, 507–518 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Srikanth, S. et al. The Ca2+ sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum. Nat. Immunol. 20, 152–162 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wang, Q. et al. The E3 ubiquitin ligase AMFR and INSIG1 bridge the activation of TBK1 kinase by modifying the adaptor STING. Immunity 41, 919–933 (2014).

    Article  CAS  PubMed  Google Scholar 

  167. Chen, W. et al. ER adaptor SCAP translocates and recruits IRF3 to perinuclear microsome induced by cytosolic microbial DNAs. PLoS Pathog. 12, e1005462 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  168. Luo, W. W. et al. iRhom2 is essential for innate immunity to DNA viruses by mediating trafficking and stability of the adaptor STING. Nat. Immunol. 17, 1057–1066 (2016).

    Article  CAS  PubMed  Google Scholar 

  169. Vece, T. J. et al. Copa syndrome: a novel autosomal dominant immune dysregulatory disease. J. Clin. Immunol. 36, 377–387 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Gonugunta, V. K. et al. Trafficking-mediated STING degradation requires sorting to acidified endolysosomes and can be targeted to enhance anti-tumor response. Cell Rep. 21, 3234–3242 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Shen, Y. Y. et al. Adaptive evolution of energy metabolism genes and the origin of flight in bats. Proc. Natl Acad. Sci. USA 107, 8666–8671 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Seluanov, A., Gladyshev, V. N., Vijg, J. & Gorbunova, V. Mechanisms of cancer resistance in long-lived mammals. Nat. Rev. Cancer 18, 433–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Xie, J. et al. Dampened STING-dependent interferon activation in bats. Cell Host Microbe 23, 297–301.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Brubaker, S. W., Gauthier, A. E., Mills, E. W., Ingolia, N. T. & Kagan, J. C. A bicistronic MAVS transcript highlights a class of truncated variants in antiviral immunity. Cell 156, 800–811 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Qi, N. et al. Multiple truncated isoforms of MAVS prevent its spontaneous aggregation in antiviral innate immune signalling. Nat. Commun. 8, 15676 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Shi, Y. et al. An autoinhibitory mechanism modulates MAVS activity in antiviral innate immune response. Nat. Commun. 6, 7811 (2015).

    Article  CAS  PubMed  Google Scholar 

  177. Gu, W. et al. Distinct argonaute-mediated 22G-RNA pathways direct genome surveillance in the C. elegans germline. Mol. Cell 36, 231–244 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. You, F. et al. PCBP2 mediates degradation of the adaptor MAVS via the HECT ubiquitin ligase AIP4. Nat. Immunol. 10, 1300–1308 (2009).

    Article  CAS  PubMed  Google Scholar 

  179. Zhong, B. et al. The E3 ubiquitin ligase RNF5 targets virus-induced signaling adaptor for ubiquitination and degradation. J. Immunol. 184, 6249–6255 (2010).

    Article  CAS  PubMed  Google Scholar 

  180. Castanier, C. et al. MAVS ubiquitination by the E3 ligase TRIM25 and degradation by the proteasome is involved in type I interferon production after activation of the antiviral RIG-I-like receptors. BMC Biol. 10, 44 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Pan, Y. et al. Smurf2 negatively modulates RIG-I-dependent antiviral response by targeting VISA/MAVS for ubiquitination and degradation. J. Immunol. 192, 4758–4764 (2014).

    Article  CAS  PubMed  Google Scholar 

  182. Du, J. et al. pVHL negatively regulates antiviral signaling by targeting MAVS for proteasomal degradation. J. Immunol. 195, 1782–1790 (2015).

    Article  CAS  PubMed  Google Scholar 

  183. Xia, P. et al. IRTKS negatively regulates antiviral immunity through PCBP2 sumoylation-mediated MAVS degradation. Nat. Commun. 6, 8132 (2015).

    Article  PubMed  Google Scholar 

  184. Yoo, Y. S. et al. The mitochondrial ubiquitin ligase MARCH5 resolves MAVS aggregates during antiviral signalling. Nat. Commun. 6, 7910 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Jin, S. et al. Tetherin suppresses type I interferon signaling by targeting MAVS for NDP52-mediated selective autophagic degradation in human cells. Mol. Cell 68, 308–322.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  186. Qin, Y. et al. NLRX1 mediates MAVS degradation to attenuate the hepatitis C virus-induced innate immune response through PCBP2. J. Virol. 91, e01264–e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. He, X. et al. RNF34 functions in immunity and selective mitophagy by targeting MAVS for autophagic degradation. EMBO J. 38, e100978 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Vitour, D. et al. Polo-like kinase 1 (PLK1) regulates interferon (IFN) induction by MAVS. J. Biol. Chem. 284, 21797–21809 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Song, T. et al. c-Abl tyrosine kinase interacts with MAVS and regulates innate immune response. FEBS Lett. 584, 33–38 (2010).

    Article  CAS  PubMed  Google Scholar 

  190. Xiang, W. et al. PPM1A silences cytosolic RNA sensing and antiviral defense through direct dephosphorylation of MAVS and TBK1. Sci. Adv. 2, e1501889 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  191. Castanier, C., Garcin, D., Vazquez, A. & Arnoult, D. Mitochondrial dynamics regulate the RIG-I-like receptor antiviral pathway. EMBO Rep. 11, 133–138 (2010).

    Article  CAS  PubMed  Google Scholar 

  192. Yasukawa, K. et al. Mitofusin 2 inhibits mitochondrial antiviral signaling. Sci. Signal. 2, ra47 (2009).

    Article  PubMed  Google Scholar 

  193. Koshiba, T., Yasukawa, K., Yanagi, Y. & Kawabata, S. Mitochondrial membrane potential is required for MAVS-mediated antiviral signaling. Sci. Signal. 4, ra7 (2011).

    Article  PubMed  CAS  Google Scholar 

  194. Horner, S. M., Liu, H. M., Park, H. S., Briley, J. & Gale, M. Jr. Mitochondrial-associated endoplasmic reticulum membranes (MAM) form innate immune synapses and are targeted by hepatitis C virus. Proc. Natl Acad. Sci. USA 108, 14590–14595 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Dixit, E. et al. Peroxisomes are signaling platforms for antiviral innate immunity. Cell 141, 668–681 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. de Brito, O. M. & Scorrano, L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456, 605–610 (2008).

    Article  PubMed  CAS  Google Scholar 

  197. Sugiura, A. et al. MITOL regulates endoplasmic reticulum-mitochondria contacts via Mitofusin2. Mol. Cell 51, 20–34 (2013).

    Article  CAS  PubMed  Google Scholar 

  198. Schneider, W. M., Chevillotte, M. D. & Rice, C. M. Interferon-stimulated genes: a complex web of host defenses. Annu. Rev. Immunol. 32, 513–545 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Vanpouille-Box, C., Hoffmann, J. A. & Galluzzi, L. Pharmacological modulation of nucleic acid sensors ― therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 18, 845–867 (2019). (2019).

    Article  CAS  PubMed  Google Scholar 

  200. Demaria, O. et al. Harnessing innate immunity in cancer therapy. Nature 574, 45–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  201. Vanpouille-Box, C. et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 8, 15618 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  202. Vincent, J. et al. Small molecule inhibition of cGAS reduces interferon expression in primary macrophages from autoimmune mice. Nat. Commun. 8, 750 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Lama, L. et al. Development of human cGAS-specific small-molecule inhibitors for repression of dsDNA-triggered interferon expression. Nat. Commun. 10, 2261 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Andrea Ablasser or Sun Hur.

Ethics declarations

Competing interests

A.A. is a shareholder in a company that is developing STING- and cGAS-directed therapeutics.

Additional information

Peer review information Jamie D.K. Wilson was the primary editor on this review article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ablasser, A., Hur, S. Regulation of cGAS- and RLR-mediated immunity to nucleic acids. Nat Immunol 21, 17–29 (2020). https://doi.org/10.1038/s41590-019-0556-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0556-1

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