Abstract
In the cytoplasm, DNA is sensed as a universal danger signal by the innate immune system. Cyclic GMP–AMP synthase (cGAS) is a cytosolic DNA sensor/enzyme that catalyzes formation of 2′–5′-cGAMP, an atypical cyclic di-nucleotide second messenger that binds and activates the Stimulator of Interferon Genes (STING), resulting in recruitment of Tank Binding Kinase 1 (TBK1), activation of the transcription factor Interferon Regulatory Factor 3 (IRF3), and trans-activation of innate immune response genes, including type I Interferon cytokines (IFN-I). Activation of the pro-inflammatory cGAS–STING–IRF3 response is triggered by direct recognition of the DNA genomes of bacteria and viruses, but also during RNA virus infection, neoplastic transformation, tumor immunotherapy and systemic auto-inflammatory diseases. In these circumstances, the source of immuno-stimulatory DNA has often represented a fundamental yet poorly understood aspect of the response. This review focuses on recent findings related to cGAS activation by an array of self-derived DNA substrates, including endogenous retroviral elements, mitochondrial DNA (mtDNA) and micronuclei generated as a result of genotoxic stress and DNA damage. These findings emphasize the role of the cGAS axis as a cell-intrinsic innate immune response to a wide variety of genomic insults.
Introduction
Cell-intrinsic innate immunity represents the early, essential and ubiquitous defensive response of susceptible cells to pathogenic threats. At sites of infection or exposure, innate immunity is initiated by sensing of common pathogen-associated molecular signatures by germline-encoded innate immune receptors, commonly termed pattern-recognition receptors (PRR) [1]. Immuno-stimulatory DNA encoded by invading and/or replicating intracellular pathogens is a potent innate immune ligand that elicits high levels of IFN-I and many other pro-inflammatory mediators in immune and stromal cells. Although Toll-Like Receptor 9 (TLR9) is a PRR for immuno-stimulatory DNA localized in endosomes, distinct receptors exist for sensing of cytoplasmic DNA [2•]; of these, the sensor/enzyme cGAS has emerged as a widely expressed, essential molecule for cytoplasmic DNA recognition [2•,3•]. cGAS dimers recognize B-form DNA, independently of its sequence context [4••], by binding to the sugar-phosphate backbone without contacting individual bases [5–8]. DNA binding induces a conformational change in the active site of the enzyme, which relieves auto-inhibition and allows for an atypical phosphodiester bond to be formed between the 2′ hydroxyl group of GMP and the 5′ phosphate of AMP, which is followed by cyclization to form an additional canonical 3′–5′ linkage. Short DNA fragments of 15–20 base pairs bind efficiently to cGAS and induce its activity in vitro [8]; however, longer DNA elements are required to fully activate cGAS, particularly in cells [9]. A cooperative mechanism by which longer DNA elements are recognized by a network of cGAS dimers has been recently described [10]. Assembly of a single dimer appears to nucleate formation of additional structures by inducing U-turn bends in flanking DNA, which favors recognition and binding by additional cGAS dimers that assemble ladder-like along the strand of DNA. The formation of U-bend structures is also enhanced by endogenous DNA-associated proteins like HMGB1, TFAM and Hu proteins [10]. In this way, cGAS recognizes a wide variety of DNA substrates, including microbial genomes, endogenous retroviral elements, and more recently mitochondrial DNA (mtDNA) and chromatinized nuclear DNA. This review focuses on recent studies demonstrating cGAS activation by different DNA species in wide-ranging scenarios of infection, auto-inflammatory disease, cancer and DNA damage.
DNA sensing during infections
cGAS is essential for early innate sensing and control of a diverse and ever-increasing number of DNA-encoding viruses and intracellular bacterial pathogens [2•]. The human retroviruses HIV1 and HIV2 are also detected by cGAS, upon cytosolic accumulation of reverse transcribed cDNA originating from damaged capsids or generated during active replication. Viral replication is highly sensitive to the effects of IFN-I and IFN-stimulated genes [11,12], and early innate sensing through the cGAS axis is essential for controlling infections. Mice deficient in cGAS fail to mount an IFN-I response to DNA-encoding herpes simplex virus, vaccinia virus and murine γ-herpesvirus 68 [13••,14••], rendering knockout mice highly susceptible to lethal infection. For other DNA-encoding pathogens the scenario is more complex, with the cGAS–STING pathway driving initial detection as a part of a greater bi-phasic response to sub-lethal infection. Human and mouse cytomegalovirus potently activate cGAS–STING signaling, and STING and cGAS knock-out mice initially exhibit impaired levels of IFN-I and high viral titers in organs [15]; as infection progresses and cGAS–STING independent, TLR9-dependent IFN-I responses develop in myeloid cells, the second wave of IFN-I is actually higher in the knock-out mice, presumably triggered by higher viral loads. Despite a strong secondary response, it is likely that failure to control initial infection in pathogen-susceptible cells promotes increased cytomegalovirus latency, a key factor driving viral persistence and pathogenesis long-term.
RNA viruses also trigger cGAS and STING signaling. One mechanism involves cytosolic sensing of viral membrane fusion, as observed during influenza A infection, which results in activation of STING in a cGAS-independent manner [16]. However, in vitro studies indicate that cGAS exerts broad anti-viral effects against numerous RNA-encoding viruses, and cGAS knock-out mice display impaired in vivo responses to infection with west nile virus [14••]. Although A-form RNA can occupy the active site of cGAS [9], it is not capable of relieving auto-inhibition of the enzyme, suggesting a distinct mechanism of action for RNA pathogens. RNA:DNA hybrid molecules are capable of binding cGAS and inducing its activity [17], although it is not known if these structures can accumulate in the cytoplasm during RNA virus infection. Activation of cGAS by cytosolic mtDNA has been demonstrated in several contexts [18•,19•,20•,21•]. Deficiency of pro-apoptotic caspases results in permeabilization of the mitochondrial outer membrane and causes mtDNA leakage into the cytosol, potently activating cGAS–STING responses [18•,19•]. Haplo insufficiency of TFAM, a DNA-binding protein of the inner mitochondrial membrane, causes aberrant mtDNA packaging and cytosolic leakage [20•]; interestingly, TFAM and other DNA binding proteins actually promote cGAS recognition of long DNA strands by inducing U-bend structures that nucleate dimer binding [10]. The vaccine adjuvant chitosan also produces cGAS and STING-dependent IFN-I responses through mitochondrial stress and cytosolic mtDNA accumulation [21•]. Notably, mtDNA leakage occurs during natural herpes simplex virus infection [20•], and recent studies also point to mtDNA as an innate sensor for the RNA-encoding virus Dengue [22,23]. As a strategy for evading innate immune detection by the host, the viral protease cofactor NS2B degrades cGAS [22], which is activated during the course of Dengue infection by mitochondrial damage and mtDNA leakage [22,23]. cGAMP molecules can traffic to adjacent cells via gap junctions [24••], and the mtDNA-dependent innate immune response to Dengue is important for limiting spread of the infection to neighboring cells [23]. It is highly likely that other DNA and RNA-encoding pathogens also induce and evade mitochondrial stress and mtDNA-related responses during the course of infection, providing an extra layer of cell-intrinsic protection against those pathogens that successfully mask or sequester their genomes.
DNA sensing during inflammation
Sensing of self-DNA by cGAS signaling is significant in that it implicates the pathway in settings of inflammation other than infectious disease. Indeed, induction of IFN-I responses in the absence of discernible microbial infection, a process called ‘sterile inflammation,’ was initially described in the context of rare human monogenic diseases of auto-inflammation [25•]. Regarding cytosolic DNA sensing, loss-of-function mutations in the cytoplasmic DNA endonuclease TREX1 [26,27••,28,29], or ribonuclease RNase H2 [30–33,34••] directly drive cGAS–STING pathway activation by self-DNA, precipitating a multi-organ inflammatory disease called Aicardi–Goutieres Syndrome (AGS). In TREX1-deficient cells, retroviral DNA elements from the cellular genome spontaneously accumulate in the cytoplasm [27••], whereas deficiency of RNase H2 precipitates genomic instability [32,33,34••]. In addition to rare monogenic syndromes, inappropriate activation of IFN-I responses through cGAS and STING is also implicated in common, chronic inflammatory diseases. Bai et al. have shown that aberrant activation of the cGAS–STING–IFN-I pathway exacerbates obesity-induced inflammation and insulin resistance [35]. Genetic ablation of Dsba-L, a protein chaperone localized to the mitochondrial matrix, enhanced mtDNA leakage and cGAS–STING activation in adipocytes, while over-expression protected against inflammatory responses and liver damage in obese mice [35,36]. Myocardial infarction also drives a classical IFN-I response downstream of cGAS–STING activation [37]. The mechanism involves extrinsic sensing of cellular debris and self-DNA by cardiac macrophages, and it is not yet known if cell-intrinsic DNA sensing may also play a role in non-myeloid cell types during MI.
Cancer, cancer therapy and genotoxic stress
The protective role of IFN-I as part of the host immune response to cancer is well documented. Mice deficient in IFN-I signaling display enhanced susceptibility to tumor formation [38•], and IFN-I signaling is critical for the development of anti-tumor activity of NK cells [39•] as well as antigen cross-priming of tumor-specific CD8+ T cells by DC [40•,41•]. Among an array innate immune signaling molecules examined for an essential role in tumor growth and immunogenicity, STING and cGAS appear to be uniquely indispensable for natural or spontaneous anti-tumor immunity [42••,43••], as well as the immunogenic effects of irradiation [44••], CD47 blockade [45], and immune checkpoint therapy [46]. Accordingly, cGAMP is a potent anti-tumor agent against many solid and suspension tumor types [47–50]. In addition to modulation of tumor immunogenicity, STING agonists have been demonstrated to directly affect tumor cells by promoting induction of apoptosis in breast tumor cells [51], transformed B cells [50] and transformed T cells [52]. Notably, the cell-intrinsic response of tumor cells to STING agonists is variable, depending on cancer cell type and whether the signaling pathway is intact.
Similar to scenarios of infection and inflammation, activation of cGAS–STING in tumors occurs via both extrinsic and intrinsic sensing of self-DNA. Antigen presenting DC internalize tumor-derived DNA via phagocytosis of tumor cell debris [42••,43••], creating an IFN-I-driven autocrine loop that drives CD8+ T cell priming and activation [42••,43••,44••,53]. Although extrinsic sensing of tumor cell debris is confined to cells with phagocytic capability, it appears that ubiquitous cell-intrinsic mechanisms exist for sensing genotoxic stress and DNA damage, which has long been appreciated as a robust inducer of the IFN-I response [54,55]. As part of a cell-intrinsic replicative stress response, prostate cancer cells continuously shed genomic DNA into the cytosol due to cleavage of DNA at stalled replication forks by the DNA repair endonuclease MUS81, which results in activation of STING-dependent cytosolic DNA sensing and contributes to tumor immunogenicity and rejection [56]. Extra-chromosomal telomere repeat DNA (ECTR) fragments, generated by trimming of telomeres elongated through the pro-oncogenic alternative lengthening of telomeres (ATL) pathway, accumulate in ~10–15% of cancer cells and can also trigger cGAS–STING–IFN-I signaling [57]. Notably, in ALT cancer cells cytosolic DNA sensing is defective, suggesting that loss of cell-intrinsic self-DNA sensing is requirement for transformation and maintenance of ALT cells. Mammary tumor cells subjected to increasing doses of radiation therapy also accumulate DNA in the cytoplasm, which correlates with cGAS-mediated expression of IFN-I and IFN-I stimulated genes [58]. TREX1 is an important regulator of the intrinsic process, as it is up-regulated at high doses of radiation and drives attenuation of the immunogenic signal. Recently, five independent studies have shed new insight into the role of the cGAS–STING axis in maintenance of genomic integrity by intrinsic sensing of genotoxic stress [34••,59••,60••,61••,62••]. In cells deficient in ribonuclease RNase H2, a causative factor in inflammatory AGS that is involved in DNA synthesis and repair [30–33,34••], cGAS specifically localizes to micronuclei, which are formed from mis-segregation of chromosomal DNA, where it initiates an inflammatory response [34••]. Similarly, continuous passaging of cells [59••,60••], hyperoxic conditions [60••] or ionizing radiation [61••,62••] also activate cGAS-mediated sensing of damaged genomic DNA fragments or micronuclei, with the resulting inflammatory response driving cellular senescence. Interestingly, recognition of chromatinized DNA substrates contained within micronuclei by cGAS requires the breakdown of the micronuclear membrane, which can only occur during mitotic progression [34••,62••]. The ability of the cGAS pathway to impair cellular immortalization by sensing genotoxic stress in a cell-intrinsic manner underscores a key role in preventing tumorigenesis, one that is distinct from immuno-modulation.
Conclusions
IFN-I are profoundly immuno-modulatory cytokines that exert pleiotropic effects on cells and tissues. As a nearly-universal defensive response to pathogens and cell or tissue damage, IFN-I are secreted by many different stromal and immune cell types during the course of infection, tissue damage, neoplastic transformation and tumor therapy. Accordingly, there exist many different pathways to IFN-I production that engage distinct but overlapping innate immune signal transduction cascades. The localization, timing and origin of the IFN-I response is crucial to its effectiveness, and early responses are particularly important for limiting damage and developing effective immunity. The cGAS–STING–IRF3 is a ubiquitous cellular mechanism for triggering robust IFN-I responses at directly at the site of infection, exposure or damage. Initially established for its essential role as a defensive response to intracellular bacteria and viruses encoding DNA genomes, it has become clear that innate immune sensing of a wide variety of mis-localized or aberrant DNA structures and fragments is a strategy for detecting pathogenic threats. With physiological relevance established as an intrinsic and extrinsic determinant of immunity to microbial pathogens, genotoxic stress, malignant transformation, and tumor growth, boosting or restoring cytosolic DNA sensing is of obvious clinical importance in the context of infection and cancer. However, lessons from inflammatory disease indicate that non-specific or excessive activation of this nearly-ubiquitous response can have dire consequences in terms of immune-related adverse events and systemic inflammation, the so-called ‘Achilles heel’ of immune-related therapy.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1.Medzhitov R. Pattern recognition theory and the launch of modern innate immunity. J Immunol. 2013;191:4473–4474. doi: 10.4049/jimmunol.1302427. [DOI] [PubMed] [Google Scholar]
- 2•.Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS– STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–1149. doi: 10.1038/ni.3558. See annotation to Ref. [3•] [DOI] [PubMed] [Google Scholar]
- 3•.Cai X, Chiu YH, Chen ZJ. The cGAS–cGAMP–STING pathway of cytosolic DNA sensing and signaling. Mol Cell. 2014;54:289–296. doi: 10.1016/j.molcel.2014.03.040. Refs. [2•,3•] are exceptional reviews that comprehesively summarize significant advancements and insights in the rapidly evolving field of cytosolic DNA sensing and cGAS signaling. [DOI] [PubMed] [Google Scholar]
- 4••.Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP–AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339:786–791. doi: 10.1126/science.1232458. Study that identified cGAS at the mammalian cytosolic DNA sensor/enzyme that produces 2′5′-cGAMP using classical biochemical methods. This study demonstrated that cGAS binds to DNA independently of its sequence context. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Civril F, Deimling T, de Oliveira Mann CC, Ablasser A, Moldt M, Witte G, Hornung V, Hopfner KP. Structural mechanism of cytosolic DNA sensing by cGAS. Nature. 2013;498:332–337. doi: 10.1038/nature12305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gao P, Ascano M, Wu Y, Barchet W, Gaffney BL, Zillinger T, Serganov AA, Liu Y, Jones RA, Hartmann G, et al. Cyclic [G(2′,5′) pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP–AMP synthase. Cell. 2013;153:1094–1107. doi: 10.1016/j.cell.2013.04.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Li X, Shu C, Yi G, Chaton CT, Shelton CL, Diao J, Zuo X, Kao CC, Herr AB, Li P. Cyclic GMP–AMP synthase is activated by double-stranded DNA-induced oligomerization. Immunity. 2013;39:1019–1031. doi: 10.1016/j.immuni.2013.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhang X, Wu J, Du F, Xu H, Sun L, Chen Z, Brautigam CA, Zhang X, Chen ZJ. The cytosolic DNA sensor cGAS forms an oligomeric complex with DNA and undergoes switch-like conformational changes in the activation loop. Cell Rep. 2014;6:421–430. doi: 10.1016/j.celrep.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Herzner AM, Hagmann CA, Goldeck M, Wolter S, Kubler K, Wittmann S, Gramberg T, Andreeva L, Hopfner KP, Mertens C, et al. Sequence-specific activation of the DNA sensor cGAS by Y-form DNA structures as found in primary HIV-1 cDNA. Nat Immunol. 2015;16:1025–1033. doi: 10.1038/ni.3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Andreeva L, Hiller B, Kostrewa D, Lassig C, de Oliveira Mann CC, Jan Drexler D, Maiser A, Gaidt M, Leonhardt H, Hornung V, et al. cGAS senses long and HMGB/TFAM-bound U-turn DNA by forming protein-DNA ladders. Nature. 2017;549:394–398. doi: 10.1038/nature23890. This study describes the use of cell-based assays, protein biochemistry and structure analysis to elucidate a model by which cGAS dimers cooperatively form ladder-like structures along long strands of DNA. Binding of the pioneering dimers or of structural DNA binding proteins such as HMGB1, Hu or TFAM promote formation of U-bend DNA structures that favour recognition and binding of additional cGAS dimers. [DOI] [PubMed] [Google Scholar]
- 11.Stark GR. How cells respond to interferons revisited: from early history to current complexity. Cytokine Growth Factor Rev. 2007;18:419–423. doi: 10.1016/j.cytogfr.2007.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu Rev Biochem. 1998;67:227–264. doi: 10.1146/annurev.biochem.67.1.227. [DOI] [PubMed] [Google Scholar]
- 13••.Li XD, Wu J, Gao D, Wang H, Sun L, Chen ZJ. Pivotal roles of cGAS–cGAMP signaling in antiviral defense and immune adjuvant effects. Science. 2013;341:1390–1394. doi: 10.1126/science.1244040. Comprehensive characterization of cytoslic DNA sensing in cGAS-deficiency in immune cells, stromal cells and animals, which fail to mount IRF3-dependent IFN-I responses to immuno-stimulatory DNA and DNA virus infections. It was also shown that cGAMP supplementation is effective as an immune adjuvant for stimulating T and B cell responses in vivo. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14••.Schoggins JW, MacDuff DA, Imanaka N, Gainey MD, Shrestha B, Eitson JL, Mar KB, Richardson RB, Ratushny AV, Litvak V, et al. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature. 2014;505:691–695. doi: 10.1038/nature12862. A comprehensive functional study of Interferon Stimulated genes (ISG) in mediating anti-viral immunity to viral pathogens. This study identified the cGAS gene product as a factor that elicits broad-spectrum immunity to DNA viruses and positive strand RNA-encoding viruses such as west nile virus. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lio CW, McDonald B, Takahashi M, Dhanwani R, Sharma N, Huang J, Pham E, Benedict CA, Sharma S. cGAS–STING signaling regulates initial innate control of cytomegalovirus infection. J Virol. 2016;90:7789–7797. doi: 10.1128/JVI.01040-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Holm CK, Rahbek SH, Gad HH, Bak RO, Jakobsen MR, Jiang Z, Hansen AL, Jensen SK, Sun C, Thomsen MK, et al. Influenza A virus targets a cGAS-independent STING pathway that controls enveloped RNA viruses. Nat Commun. 2016;7:10680. doi: 10.1038/ncomms10680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mankan AK, Schmidt T, Chauhan D, Goldeck M, Honing K, Gaidt M, Kubarenko AV, Andreeva L, Hopfner KP, Hornung V. Cytosolic RNA:DNA hybrids activate the cGAS–STING axis. EMBO J. 2014;33:2937–2946. doi: 10.15252/embj.201488726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18•.Rongvaux A, Jackson R, Harman CC, Li T, West AP, de Zoete MR, Wu Y, Yordy B, Lakhani SA, Kuan CY, et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell. 2014;159:1563–1577. doi: 10.1016/j.cell.2014.11.037. See annotation to Ref. [21•]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19•.White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC, Herold MJ, van Delft MF, Bedoui S, Lessene G, Ritchie ME, et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell. 2014;159:1549–1562. doi: 10.1016/j.cell.2014.11.036. See annotation to Ref. [21•] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20•.West AP, Khoury-Hanold W, Staron M, Tal MC, Pineda CM, Lang SM, Bestwick M, Duguay BA, Raimundo N, MacDuff DA, et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature. 2015;520:553–557. doi: 10.1038/nature14156. See annotation to Ref. [21•]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21•.Carroll EC, Jin L, Mori A, Munoz-Wolf N, Oleszycka E, Moran HBT, Mansouri S, McEntee CP, Lambe E, Agger EM, et al. The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS–STING-dependent induction of type I interferons. Immunity. 2016;44:597–608. doi: 10.1016/j.immuni.2016.02.004. Refs. [18•,19•,20•] show that cytosolic leakage of mtDNA due to mitochodrial stress and damage results in activation of the cGAS–STING pathway. Refs. [18•,19•] demonstrate this phenomenon in the context of apoptotic caspase deficiency and permeabililization of the mitochondrial outer membrane, and Ref. [20•] shows that loss of the mtDNA-binding protein TFAM results in aberrant mtDNA packaging. Ref. [20•] also demonstrates that mtDNA leakage occurs in cells infected with herpes simplex virus. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Aguirre S, Luthra P, Sanchez-Aparicio MT, Maestre AM, Patel J, Lamothe F, Fredericks AC, Tripathi S, Zhu T, Pintado-Silva J, et al. Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection. Nat Microbiol. 2017;2:17037. doi: 10.1038/nmicrobiol.2017.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sun B, Sundstrom KB, Chew JJ, Bist P, Gan ES, Tan HC, Goh KC, Chawla T, Tang CK, Ooi EE. Dengue virus activates cGAS through the release of mitochondrial DNA. Sci Rep. 2017;7:3594. doi: 10.1038/s41598-017-03932-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24••.Ablasser A, Schmid-Burgk JL, Hemmerling I, Horvath GL, Schmidt T, Latz E, Hornung V. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature. 2013;503:530–534. doi: 10.1038/nature12640. Report describing the inter-cellular transfer of cGAMP molecules from cells that produce it to neighboring cells via gap junctions. Lateral transfer of cGAMP molecules is a key step in the signaling cascade that primes and protects pathogen-susceptible cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25•.Crow YJ. Type I interferonopathies: mendelian type I interferon up-regulation. Curr Opin Immunol. 2015;32:7–12. doi: 10.1016/j.coi.2014.10.005. An exceptional review article describing rare human monogenic diseases of systemic inflammation that are characterized by up-regulation of IFN-I. Provides in-depth genetic and clinical discussion of AGS sub-types as well as other similar diseases. [DOI] [PubMed] [Google Scholar]
- 26.Crow YJ, Hayward BE, Parmar R, Robins P, Leitch A, Ali M, Black DN, van Bokhoven H, Brunner HG, Hamel BC, et al. Mutations in the gene encoding the 3′–5′ DNA exonuclease TREX1 cause Aicardi–Goutieres syndrome at the AGS1 locus. Nat Genet. 2006;38:917–920. doi: 10.1038/ng1845. [DOI] [PubMed] [Google Scholar]
- 27••.Stetson DB, Ko JS, Heidmann T, Medzhitov R. Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell. 2008;134:587–598. doi: 10.1016/j.cell.2008.06.032. Identification of the endonuclease TREX1, previously identified as an AGS susceptibility gene, as a negative regulator of cytosolic DNA sensing, and description of the mechanism by which single stranded retroviral elements spontaneously accumulate in the cytosol of TREX1-deficient cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gao D, Li T, Li XD, Chen X, Li QZ, Wight-Carter M, Chen ZJ. Activation of cyclic GMP–AMP synthase by self-DNA causes autoimmune diseases. Proc Natl Acad Sci U S A. 2015;112:E5699–E5705. doi: 10.1073/pnas.1516465112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gray EE, Treuting PM, Woodward JJ, Stetson DB. Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi–Goutieres syndrome. J Immunol. 2015;195:1939–1943. doi: 10.4049/jimmunol.1500969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Crow YJ, Leitch A, Hayward BE, Garner A, Parmar R, Griffith E, Ali M, Semple C, Aicardi J, Babul-Hirji R, et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi–Goutieres syndrome and mimic congenital viral brain infection. Nat Genet. 2006;38:910–916. doi: 10.1038/ng1842. [DOI] [PubMed] [Google Scholar]
- 31.Sanchis A, Cervero L, Bataller A, Tortajada JL, Huguet J, Crow YJ, Ali M, Higuet LJ, Martinez-Frias ML. Genetic syndromes mimic congenital infections. J Pediatr. 2005;146:701–705. doi: 10.1016/j.jpeds.2005.01.033. [DOI] [PubMed] [Google Scholar]
- 32.Mackenzie KJ, Carroll P, Lettice L, Tarnauskaite Z, Reddy K, Dix F, Revuelta A, Abbondati E, Rigby RE, Rabe B, et al. Ribonuclease H2 mutations induce a cGAS/STING-dependent innate immune response. EMBO J. 2016;35:831–844. doi: 10.15252/embj.201593339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pokatayev V, Hasin N, Chon H, Cerritelli SM, Sakhuja K, Ward JM, Morris HD, Yan N, Crouch RJ. RNase H2 catalytic core Aicardi–Goutieres syndrome-related mutant invokes cGAS–STING innate immune-sensing pathway in mice. J Exp Med. 2016;213:329–336. doi: 10.1084/jem.20151464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34••.Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, Olova N, Sutcliffe H, Rainger JK, Leitch A, et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. 2017;548:461–465. doi: 10.1038/nature23449. This manuscript demonstrates that deficiency of RNase H2 drives genomic instability and accumulation of mis-segregated chromosomal DNA within micronuclei, which are recognized by cGAS. Study demonstrates cGAS sensing of chromatinized DNA, and a dependency on progression through the cell cycle as a requirement for recognition of DNA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bai J, Cervantes C, Liu J, He S, Zhou H, Zhang B, Cai H, Yin D, Hu D, Li Z, et al. DsbA-L prevents obesity-induced inflammation and insulin resistance by suppressing the mtDNA release-activated cGAS–cGAMP–STING pathway. Proc Natl Acad Sci U S A. 2017;114:12196–12201. doi: 10.1073/pnas.1708744114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chen H, Bai J, Dong F, Fang H, Zhang Y, Meng W, Liu B, Luo Y, Liu M, Bai Y, et al. Hepatic DsbA-L protects mice from diet-induced hepatosteatosis and insulin resistance. FASEB J. 2017;31:2314–2326. doi: 10.1096/fj.201600985R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.King KR, Aguirre AD, Ye YX, Sun Y, Roh JD, Ng RP, Jr, Kohler RH, Arlauckas SP, Iwamoto Y, Savol A, et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. 2017;12:1481–1487. doi: 10.1038/nm.4428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38•.Dunn GP, Bruce AT, Sheehan KC, Shankaran V, Uppaluri R, Bui JD, Diamond MS, Koebel CM, Arthur C, White JM, et al. A critical function for type I interferons in cancer immunoediting. Nat Immunol. 2005;6:722–729. doi: 10.1038/ni1213. See annotation to Ref. [41•] [DOI] [PubMed] [Google Scholar]
- 39•.Swann JB, Hayakawa Y, Zerafa N, Sheehan KC, Scott B, Schreiber RD, Hertzog P, Smyth MJ. Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J Immunol. 2007;178:7540–7549. doi: 10.4049/jimmunol.178.12.7540. See annotation to Ref. [41•] [DOI] [PubMed] [Google Scholar]
- 40•.Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, Lee H, Arthur CD, White JM, Kalinke U, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208:1989–2003. doi: 10.1084/jem.20101158. See annotation to Ref. [41•] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41•.Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, Gajewski TF. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J Exp Med. 2011;208:2005–2016. doi: 10.1084/jem.20101159. Refs. [38•,39•,40•,41•] are among the first studies to link IFN-I and IFN-I signaling to the development of effective anti-tumor immune responses, through activation of NK cells (Ref. [39•]), or by promoting the cross-priming activity of DC for CD8+ T cells (Refs. [40•,41•]) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42••.Klarquist J, Hennies CM, Lehn MA, Reboulet RA, Feau S, Janssen EM. STING-mediated DNA sensing promotes antitumor and autoimmune responses to dying cells. J Immunol. 2014;193:6124–6134. doi: 10.4049/jimmunol.1401869. See annotation to Ref. [44••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43••.Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MY, Duggan R, Wang Y, Barber GN, Fitzgerald KA, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–842. doi: 10.1016/j.immuni.2014.10.017. See annotation to Ref. [44••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44••.Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, Li XD, Mauceri H, Beckett M, Darga T, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41:843–852. doi: 10.1016/j.immuni.2014.10.019. Refs. [42••,43••,44••] link cytosolic DNA sensing through STING as an essential factor in controlling tumor growth and immunogenicity of natural or spontaneously immunogenic tumors (Refs. [42••,43••]) or irradiated tumors [44••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Liu X, Pu Y, Cron K, Deng L, Kline J, Frazier WA, Xu H, Peng H, Fu YX, Xu MM. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat Med. 2015;21:1209–1215. doi: 10.1038/nm.3931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang H, Hu S, Chen X, Shi H, Chen C, Sun L, Chen ZJ. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc Natl Acad Sci U S A. 2017;114:1637–1642. doi: 10.1073/pnas.1621363114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Demaria O, De Gassart A, Coso S, Gestermann N, Di Domizio J, Flatz L, Gaide O, Michielin O, Hwu P, Petrova TV, et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci U S A. 2015;112:15408–15413. doi: 10.1073/pnas.1512832112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, Woo SR, Lemmens E, Banda T, Leong JJ, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015;11:1018–1030. doi: 10.1016/j.celrep.2015.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fu J, Kanne DB, Leong M, Glickman LH, McWhirter SM, Lemmens E, Mechette K, Leong JJ, Lauer P, Liu W, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci Transl Med. 2015;7:283ra252. doi: 10.1126/scitranslmed.aaa4306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tang CH, Zundell JA, Ranatunga S, Lin C, Nefedova Y, Del Valle JR, Hu CC. Agonist-mediated activation of STING induces apoptosis in malignant B cells. Cancer Res. 2016;76:2137–2152. doi: 10.1158/0008-5472.CAN-15-1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chandra D, Quispe-Tintaya W, Jahangir A, Asafu-Adjei D, Ramos I, Sintim HO, Zhou J, Hayakawa Y, Karaolis DK, Gravekamp C. STING ligand c-di-GMP improves cancer vaccination against metastatic breast cancer. Cancer Immunol Res. 2014;2:901–910. doi: 10.1158/2326-6066.CIR-13-0123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gulen MF, Koch U, Haag SM, Schuler F, Apetoh L, Villunger A, Radtke F, Ablasser A. Signalling strength determines proapoptotic functions of STING. Nat Commun. 2017;8:427. doi: 10.1038/s41467-017-00573-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Andzinski L, Spanier J, Kasnitz N, Kroger A, Jin L, Brinkmann MM, Kalinke U, Weiss S, Jablonska J, Lienenklaus S. Growing tumors induce a local STING dependent type I IFN response in dendritic cells. Int J Cancer. 2016;139:1350–1357. doi: 10.1002/ijc.30159. [DOI] [PubMed] [Google Scholar]
- 54.Brzostek-Racine S, Gordon C, Van Scoy S, Reich NC. The DNA damage response induces IFN. J Immunol. 2011;187:5336–5345. doi: 10.4049/jimmunol.1100040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yu Q, Katlinskaya YV, Carbone CJ, Zhao B, Katlinski KV, Zheng H, Guha M, Li N, Chen Q, Yang T, et al. DNA-damage-induced type I interferon promotes senescence and inhibits stem cell function. Cell Rep. 2015;11:785–797. doi: 10.1016/j.celrep.2015.03.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ho SS, Zhang WY, Tan NY, Khatoo M, Suter MA, Tripathi S, Cheung FS, Lim WK, Tan PH, Ngeow J, et al. The DNA structure-specific endonuclease MUS81 mediates DNA sensor STING-dependent host rejection of prostate cancer cells. Immunity. 2016;44:1177–1189. doi: 10.1016/j.immuni.2016.04.010. [DOI] [PubMed] [Google Scholar]
- 57.Chen YA, Shen YL, Hsia HY, Tiang YP, Sung TL, Chen LY. Extrachromosomal telomere repeat DNA is linked to ALT development via cGAS–STING DNA sensing pathway. Nat Struct Mol Biol. 2017;12:1124–1131. doi: 10.1038/nsmb.3498. [DOI] [PubMed] [Google Scholar]
- 58.Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, Inghirami G, Coleman CN, Formenti SC, Demaria S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. 2017;8:15618. doi: 10.1038/ncomms15618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59••.Yang H, Wang H, Ren J, Chen Q, Chen ZJ. cGAS is essential for cellular senescence. Proc Natl Acad Sci U S A. 2017;114:E4612–E4620. doi: 10.1073/pnas.1705499114. See annotation to Ref. [62••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60••.Gluck S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L, Ablasser A. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat Cell Biol. 2017;19:1061–1070. doi: 10.1038/ncb3586. See annotation to Ref. [62••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61••.Dou Z, Ghosh K, Vizioli MG, Zhu J, Sen P, Wangensteen KJ, Simithy J, Lan Y, Lin Y, Zhou Z, et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature. 2017;550:402–406. doi: 10.1038/nature24050. See annotation to Ref. [62••] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62••.Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature. 2017;548:466–470. doi: 10.1038/nature23470. Along with Ref. [34••], these four papers demonstrate that cGAS is an endogenous innate immune sensor for genotoxic stress and DNA damage due to continuous passaging or spontanoeus immortilization (Refs. [59••,60••]) or irradation (Refs. [61••,62••]), resulting in activation of an inflammatory program that drives cellular senescence. [DOI] [PMC free article] [PubMed] [Google Scholar]