Abstract
The innate immune systems detect pathogens via pattern-recognition receptors including nucleic acid sensors and non-nucleic acid sensors. Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS, also known as MB21D1) is a cytosolic DNA sensor that recognizes double-stranded DNA (dsDNA) and catalyzes the synthesis of 2′,3′-cGAMP. Subsequently, 2′,3′-cGAMP binds to the adaptor protein mediator of IRF3 activation (MITA, also known as STING, MPYS, ERIS, and TMEM173) to activate downstream signaling cascades. The cGAS-MITA/STING signaling critically mediates immune responses against DNA viruses, retroviruses, bacteria, and protozoan parasites. In addition, recent discoveries have extended our understanding of the roles of the cGAS-MITA/STING pathway in autoimmune diseases and cancers. Here, we summarize the identification and activation of cGAS and MITA/STING, present the updated functions and regulatory mechanisms of cGAS-MITA/STING signaling and provide a comprehensive understanding of the cGAS-MITA/STING axis in autoimmune diseases and cancers.
Keywords: cGAS, MITA/STING, Innate immunity, Autoimmune diseases, Cancer
Introduction
The innate immunity plays a vital role in defense against the invasion of pathogenic micro-organisms and the maintenance of immune homeostasis. Innate immune cells recognize the structurally conserved components of pathogenic microorganisms called pathogen-associated molecular patterns (PAMPs) by pattern-recognition receptors (PRRs). Subsequently, PRRs activate downstream signaling pathways and elicit anti-microbial immune responses or pro-inflammatory responses. For example, pathogen-derived RNA is detected by the cytosolic RNA sensors retinoic acid-inducible gene (RIG-I)-like receptors (RLRs) (Hu & Shu, 2018). Upon binding to dsRNA or 5′ triphosphorylated ssRNA, RIG-I undergoes conformational change to recruit and elicit the oligomerization and activation of the mitochondrial antiviral signaling protein (MAVS, also known as VISA, IPS-1, and Cardif) (Kawai et al., 2005; Meylan et al., 2005; Seth et al., 2005; Xu et al., 2005). Activated MAVS further induces the expression of type I IFNs and pro-inflammatory cytokines via the activation of transcription factors IRF3 and NF-κB. To date, multiple cytosolic DNA sensors detecting pathogen DNA have been identified (Chiu et al., 2009; Li, Shu, et al., 2013; Sun et al., 2013; Takaoka et al., 2007; Unterholzner et al., 2010; Zhang et al., 2011). Specifically, the cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) detects dsDNA and catalyzes the synthesis of 2′,3′-cGAMP which binds to the adaptor protein mediator of IRF3 activation (MITA, also known as STING, MPYS, ERIS, and TMEM173) and activates signaling cascades leading to the induction of type I IFNs and pro-inflammatory cytokines (Li, Wu, et al., 2013; Sun et al., 2013). Accordingly, knockout of cGAS or MITA significantly inhibits the production of type I IFNs after HSV-1 infection, and mice deficient in cGAS or MITA are hypersensitive to HSV-1 infection (Ishikawa et al., 2009; Li, Wu, et al., 2013). In addition to the antimicrobial functions of cGAS and MITA, emerging studies have broadened their roles in autoimmune diseases and cancers. Here, we review the recent advances in activation and regulation of the cGAS-MITA signaling pathway and summarize the updated roles of the cGAS-MITA signaling axis in autoimmune diseases and cancers.
An overview of the cGAS-MITA/STING signaling pathway
Structure, localization, and activation of MITA/STING
Because the MAVS-deficient MEFs respond normally to cytoplasmic DNA challenge (Sun et al., 2006), it is believed that there exist other adaptor proteins mediating intracellular DNA- and DNA virus-triggered signaling. In 2008–2009, four groups have independently identified a key adaptor protein that mediates DNA virus-triggered and cytoplasmic DNA-induced signal transduction and has been named as MITA (Zhong et al., 2008), STING (Ishikawa & Barber, 2008), MPYS (Jin et al., 2008), and ERIS (Sun et al., 2009). The phosphorylation of IRF3 and the production of type I IFNs are almost completely inhibited after DNA virus infection or DNA transfection in MITA-knockout cells and the MITA-deficient mice exhibit hypersensitivity to HSV-1 infection compared to the wild-type controls, indicating that MITA plays an indispensable role in cytosolic DNA-induced immune responses (Ishikawa et al., 2009).
The human and the murine MITA consist of 379 or 378 amino acid residues respectively and share with 81% homology. MITA consists of 4 transmembrane (TM) domains at the N-terminus responsible for its membrane localization, a cytoplasmic ligand-binding domain (LBD), and a flexible C-terminal domain (CTD) that is responsible for downstream TBK1 interaction (Burdette & Vance, 2013; Yin et al., 2012). The LBD domain exists as a dimer that displays the butterfly shape and is capable of binding cyclic di-nucleotide (CDN) such as s c-di-GMP, c-di-AMP, and 2′,3′-cGAMP (Burdette et al., 2011; Huang et al., 2012; Whiteley et al., 2019; Yin et al., 2012; Zhang, Bai, & Chen, 2020). The binding of CDN in the LBD domain induces the conformational changes of the LBD domain and a 180° rotation in relation to the TM domain for the ordering of the CTD of MITA and the oligomerization of MITA, which promotes the recruitment of TBK1 to activate IRF3 and NF-κB (de Oliveira Mann et al., 2019; Morehouse et al., 2020; Shang et al., 2019; Wu et al., 2014; Zhang et al., 2019, 2020c; Zhao et al., 2019).
MITA is strictly regulated in uninfected cells. On one hand, the Ca2+ sensor stromal interaction molecule 1 (STIM1) and TOLLIP interact with MITA to retain and stabilize it in the ER membrane, respectively (Pokatayev et al., 2020; Srikanth et al., 2019). On the other hand, the unfolded protein response (UPR) effector IRE1α promotes MITA trafficking from ER to lysosomes for degradation, which is partially mediated by the SREBP2-SCAP complex and the lysosomal membrane protein Niemann–Pick type C1 (NPC1) (Chu et al., 2021; Pokatayev et al., 2020). The binding of CDNs to MITA disrupts the STIM1-MITA interactions and initiates the translocation of MITA from ER to the Golgi apparatus via the ER-Golgi intermediate compartment (ERGIC). In such a process, iRhom2 interacts with MITA and recruits the Sec5/TRAPβ/Sec61β complexes to facilitate the translocation to ERGIC (Luo et al., 2016) and STEEP promotes ER membrane curvature to reinforce SEC24C-mediated MITA ER exit via COPII vesicles (Gui et al., 2019; Zhang, Nandakumar, et al., 2020). Meanwhile, the Surface 4 integral membrane protein (SURF4) interacts with sectional MITA at the Golgi apparatus to promote the encapsulation of MITA into COPI vesicles, which retrieves MITA from the Golgi apparatus back to the ER to counteract the activation process of MITA (Deng et al., 2020). At the Golgi apparatus, MITA undergoes palmitoylation, which promotes the recruitment of TANK binding kinase 1 (TBK1) (Kwon & Bakhoum, 2020). Fang and colleagues have recently reported a mechanistic understanding of MITA activation at the Golgi apparatus (Fang et al., 2021). Sulfated Glycosaminoglycans (sGAGs), a kind of linear acidic polysaccharides, can bind to MITA at the Golgi apparatus, leading to the polymerization of MITA and the recruitment of TBK1. Afterward, TBK1 phosphorylates the C-terminal domains of MITA to recruit IRF3 for activation. Phosphorylated IRF3 binds to the nuclear shuttle protein Karyopherin alpha 2 (KPNA2) and is translocated to the nucleus to induce transcription of an array of downstream genes (Cai et al., 2020). In parallel, MITA also activates the IKK complex to mediate the activation of NF-κB and the induction of NF-κB-driven inflammatory genes (Chen, Sun, & Chen, 2016). In addition, the active MITA-containing ERGIC serves as a membrane source for LC3 lipidation, which is a key step for autophagosome biogenesis dependent on WIPI2 and ATG5 but independent of the ULK and VPS34-Beclin kinase complexes (Gui et al., 2019). MITA-mediated activation of autophagy is important for the clearance of DNA and viruses in the cytosol and such a process is independent of the recruitment of TBK1. Afterward, MITA is transported to perinuclear compartments and undergoes lysosome-mediated degradation to terminate hyperimmune responses (Kwon & Bakhoum, 2020). Alternatively, the overactivated MITA by excessive CDNs forms a puzzle-shaped membranous structure (known as MITA-TBK1-CDN sponge) to constrain MITA and TBK1 and prevent innate immunity from overactivation in a manner dependent on microtubules (Yu et al., 2021). Consistently, MITAE336G/E337G loses the condensation ability and thereby exhibits enhanced antiviral activity in cells. Therefore, the location of MITA is tightly controlled temporally and spatially which balances the quality and quantity of innate immune signaling in the absence and presence of viral infections.
Structure and activation of cGAS
Multiple cytosolic DNA sensors have been identified including DAI, RNA polymerase III, IFI16, and DDX41 (Chiu et al., 2009; Takaoka et al., 2007; Unterholzner et al., 2010; Zhang et al., 2011). However, it is still elusive how cytosolic DNA induces the expression of type I IFNs through distinct but functionally overlapped sensors. In 2013, by biochemical fractionation from cytosolic extracts of the murine fibrosarcoma cell line L929 cells and quantitative mass spectrometry assays, Dr. Zhijian Chen's group has identified a cytosolic DNA sensor cGAS (Sun et al., 2013) which directly binds to DNA and catalyzes the synthesis of cGAMP to induce type I IFNs in a manner dependent on MITA.
As a nucleotidyltransferase (NTase), human cGAS consists of 507 amino acids and is composed of a poorly conserved N-terminal domain (amino acid residues 1–160), a highly conserved C-terminal NTase (160–330) domain, and a Mab21 (213–513) domain (Li, Zhang, et al., 2013; Wu et al., 2014). cGAS contains three DNA-binding sites (site A, B and C) at its C-terminus that are important for its activation (Li, Shu, et al., 2013; Xie et al., 2019; Zhang, Wu, et al., 2014). The positively charged patches (such as K407 and K411) in these sites of cGAS mediate the cGAS-DNA binding and mutations of the key residues in any of the three DNA-binding sites of cGAS dampen the enzymatic activity of cGAS and the induction of type I IFNs in cells (Li, Shu, et al., 2013; Xie et al., 2019; Zhang, Wu, et al., 2014). Structural analysis of human cGAS demonstrates that the K187 and L195 of hcGAS in site A interfere with the binding of DNA to cGAS (Zhou et al., 2018). Interestingly, murine cGAS has much higher enzymatic activity than hcGAS and mcGAS contains N172 and R180 that correspond to K187 and L195 of hcGAS (Zhou et al., 2018). Furthermore, mutation of these two sites (hcGASK187N/L195R) significantly increases the enzymatic activity of hcGAS to a comparable level of the activity of mcGAS. These biochemical and structural studies have elegantly revealed the mechanism of DNA binding to cGAS.
The binding of DNA to cGAS results in the formation of a 2 cGAS:2 DNA complex, which efficiently catalyzes ATP and GTP into 2′,3′-cGAMP (Fig. 1). The length of DNA for sufficient cGAS activation is ∼45 bp and longer DNA would recruit cGAS dimers to form ladder-like networks with DNA and efficiently promote the synthesis of 2′,3′-cGAMP by cGAS (Andreeva et al., 2017; Kranzusch et al., 2013). Consistent with this notion, short DNA (20 bp) barely activates cGAS in vivo and in vitro, though it binds to cGAS and induces the formation of cGAS dimers (Andreeva et al., 2017). The binding of DNA induces liquid phase separation of cGAS and thereby the cGAS-DNA complex is encapsulated in liquid droplets (Fig. 1), in which the activated cGAS catalyzes the synthesis of 2′,3′-cGAMP in a manner dependently on Zn2+ and the concentration of cGAS and DNA (Du & Chen, 2018). Recently, Mn2+ is reported to be indispensable for the activation of cGAS (Wang et al., 2018). Structural evidence shows that Mn2+ activates monomeric cGAS in the absence of DNA and also synergizes with dsDNA for the activation of cGAS (Hooy et al., 2020; Zhao, Ma, et al., 2020). Collectively, these available data have clearly demonstrated the step-wise and ion-mediated activation of cGAS.
Fig. 1.
The cGAS-MITA/STING pathway. The accumulation of cytosolic DNA is a mark of pathogen invasion. Cytosolic DNA sourced from DNA virus, damaged mitochondria, or reverse transcription of retroelements is detected by cGAS, leading to the formation of cGAS-DNA liquid droplets, in which cGAS, ATP, and GTP are concentrated to initiate the synthesis of cGAMP. MITA subsequently binds to cGAMP and undergoes dimerization and polymerization. Polymerized MITA encapsulated in SEC24C- mediated COPII vesicles translocates from the ER to Golgi via ERGIC, where MITA triggers autophagosome formation via LC3 lipidation, which leads to the clearance of cytosolic DNA and pathogens. At the Golgi apparatus, MITA undergoes palmitoylation and binds to sGAGs, which contributes to the recruitment of TBK1 and autophosphorylation of TBK1. TBK1 phosphorylates the C-terminal domains of MITA to recruit IRF3, after that IRF3 also is phosphorylated by TBK1 and translocates to the nucleus in a KPNA2-dependent manner, mediating the expression of type I IFNs. Moreover, MITA also activates NF-κB to mediate the expression of inflammatory genes. After the translocation process, MITA would be targeted to the lysosome for degradation and retrieve from Golgi back to the ER to avoid hyperactivation. cGAMP, 2′,3′-cyclic GMP-AMP; sGAG, sulfated glycosaminoglycans; IFN, interferon.
Localization of cGAS
It has been proposed that cGAS is a cytoplasmic protein to sense cytosolic DNA by biochemical and confocal immunofluorescent assays (Sun et al., 2013). However, recent studies have suggested that cGAS also localizes in the plasma membrane and the nucleus. cGAS interacts with phosphoinositide through its N terminal domain and thereby resides on the plasma membrane to distinct self- and viral DNA in resting murine and human phagocytes (Barnett et al., 2019). Upon binding to DNA, cGAS disassociates from phosphoinositide and translocates from the cell surface to cytoplasm, which facilitates liquid droplet formation as well as signaling transduction. cGAS has been observed in the nucleus when overexpressed in normal human fibroblasts and keratinocytes (Orzalli et al., 2015). Treatment of genotoxic agents, including etoposide, H2O2, and camptothecin, induces DNA damage, which leads to the translocation of cGAS from the cytosol to the nucleus (Liu, Zhang, Wu, et al., 2018). In the nucleus, cGAS is recruited to the double-stranded breaks and interacts with PARP1 to suppress homologous recombination, thereby promoting the accumulation of DNA damages and cell death (Liu, Zhang, Wu, et al., 2018; Ray Chaudhuri & Nussenzweig, 2017). During mitosis, cGAS is post-translationally regulated and rapidly translocates from the cytoplasm to chromosomes where cGAS is sequestered and inhibited (discussed below). These studies collectively demonstrate that cGAS is localized in the nucleus and plasma membrane under specific conditions. More quantitative studies of the cellular distribution of cGAS are required in future investigations.
cGAS-mediated sensing of pathogenic microorganisms
Because cGAS binds to DNA irrespective of its sequences, cGAS could be activated by all pathogenic microorganisms that produce dsDNA or ssDNA during infections and replication. Multiple studies showed that DNA viruses, including herpes simplex virus 1 (HSV-1) and HSV-2, adenoviruses, murine gammaherpesvirus 68, vaccinia virus, human papillomavirus (HPV), cytomegalovirus, and papillomavirus, are sensed by cytoplasmic cGAS to initiate immune responses (Fig. 1) (Lam et al., 2014; Schoggins et al., 2014; Sun et al., 2013; Tan et al., 2018). Consistently, cGAS knockout mice are unable to produce type I interferons and inflammatory cytokines and, thus, are susceptible to these viral pathogens. In addition, retroviruses including HIV-1 and HIV-2, murine leukemia virus (MLV), and simian immunodeficiency virus (SIV) can activate cGAS-mediated immune responses in a manner dependently on the reverse-transcribed viral cDNA (Gao et al., 2013; Lahaye et al., 2013; Sumner et al., 2020) (Fig. 1). Several RNA viruses also indirectly stimulate the cGAS-MITA pathway by inducing mitochondrial DNA release (Fig. 1) (Aguirre et al., 2017; Moriyama et al., 2019). Compared to the wild-type mice or cells, the cGAS knockout mice or cells exhibit impaired production of type I IFNs after infections of Plasmodium (Gallego-Marin et al., 2018; Hahn et al., 2018) and various intracellular bacteria, including Listeria monocytogenes (Nandakumar et al., 2019), Chlamydia trachomatis (Zhang, Yeruva, et al., 2014), and Francisella tularensis (F. novicida) (Man et al., 2015) (Fig. 1). These findings together suggest that cGAS senses a broad spectrum of invading microorganisms.
Regulation of thecGAS-MITA/STINGsignaling pathway
To avoid harmful pathology caused by aberrant cGAS-MITA-mediated innate inflammatory responses, the immune system has evolved comprehensive and multi-level mechanisms to tightly regulate the cGAS-MITA signaling pathway, including (1) homeostatic regulation of cGAS activation; (2) transcriptional regulation of cGAS and MITA; and (3) post-translational modifications (PTMs) of cGAS and MITA.
Homeostatic regulation of cGAS activation by controlling the availability of self-DNA
Under normal conditions, mitochondrial DNA (mtDNA) is encased within the outer and inner mitochondrial membrane and thus inaccessible to cGAS (Zierhut & Funabiki, 2020). Intrinsic apoptosis in a manner of activation of Bak and Bax leads to mitochondrial damage, ultimately resulting in the release of mtDNA. Yet, this cytoplasmic mtDNA does not lead to the activation of cGAS, as apoptosome complex blocks the cGAS-MITA pathway via proteolytic cleavage of cGAS (White et al., 2014). Alternatively, damaged mitochondria are immediately cleared in selective degradation of a process termed mitophagy that avoids the activation of cGAS (Sliter et al., 2018). Recently, one report demonstrates that apoptotic cells generate extrachromosomal circular DNA (eccDNA) that could trigger MITA-mediated immune responses (Wang et al., 2021). Whether this process involves cGAS-mediated cGAMP production is unknown.
The nuclear envelope (NE) provides a physical barrier that separates cytosolic cGAS from chromosomal DNA. However, the NE is disassembled during cell division, thereby making chromosomal DNA accessible to cGAS. Some key mechanisms to ensure the nonresponsiveness of cGAS to chromosomal DNA have been characterized. First, binding to the nucleosome of cGAS during cell mitosis locks cGAS into a monomeric state, and such a steric hindrance inhibits spurious activation of cGAS by chromosomal DNA (Boyer et al., 2020; Cao et al., 2020; Kujirai et al., 2020; Michalski et al., 2020; Pathare et al., 2020; Zhao, Xu, et al., 2020). Second, barrier-to-autointegration factor 1 (BAF) outcompetes cGAS for DNA binding during mitosis, which prohibits the formation of cGAS-DNA complexes (Guey et al., 2020). Third, circular RNA antagonist for cGAS (cia-cGAS) harbors a stronger binding affinity to nuclear cGAS than chromosomal-DNA, consequently restraining cGAS-mediated production of type I IFNs in long-term hematopoietic stem cells (LT-HSCs) (Xia et al., 2018). A fourth mechanism has been proposed by two recent reports (Li et al., 2021; Zhong et al., 2020). During mitosis, Aurora kinase B and cyclin-dependent kinase 1 (CDK1) selectively phosphorylate cGAS, which significantly prohibits its activity. Together, these findings suggest that cGAS activation by chromosomal or mitochondrial self-DNA is limited under unperturbed conditions.
The endogenous retroelements-derived DNA also activates cGAS-mediated immune responses (Zierhut & Funabiki, 2020). Human endogenous retroviruses (HERVs) sourcing from exogenous retroviruses that successfully integrate themselves into the host genome are inherited by successive generations (Ishak et al., 2018). Multiple reverse transcription events of HERVs make reverse-transcribed cDNA accessible to cGAS. DNase enzymes, including DNase I (Ahn & Barber, 2014), DNase II (Rodero et al., 2017), and exonuclease 1(Trex1) (Stetson et al., 2008), are responsible for clearing retroelements-derived DNA, thereby limiting the engagement between self-DNA and cGAS.
Moreover, the binding of cGAS to DNA involves extensive ionic interactions between the positively charged surface of cGAS and the negatively charged DNA (Du & Chen, 2018). Such interactions are susceptible to the cytosolic salt concentration, which may be responsible for limiting the spurious activation of cGAS by self-DNA below a certain threshold. Overall, it appears that vertebrates have evolved a series of precise regulatory elements to prevent self-DNA-mediated spontaneous activation of cGAS. Future studies are necessary to establish the mechanistic basis of homeostatic cGAS regulation by self-DNA and the signaling consequences.
Transcriptional regulation of cGAS and MITA/STING
It has been reported that cGAS is induced by type I IFNs through JAK-STAT-axis in both human and mouse macrophages (Ma et al., 2015). However, this positive feedback induction of cGAS by type I IFNs is contrary to the observations that MITA inhibits the activation of JAK1-STAT1 signaling cascades by inducing phosphorylation of SHP1 and SHP2 (Dong et al., 2015). Moreover, NCOA3 is an epigenetic factor maintaining the transcriptional levels of cGAS expression, and miR25/93 targets NCOA3 to repression of cGAS in hypoxic tumor cells (Wu et al., 2017). In several human colorectal cancer and melanoma cell lines, cGAS expression is also silenced via the epigenetic hypermethylation process, which could be overturned by treating with the demethylation agent (Xia et al., 2016b, 2016c).
Similar to cGAS expression, MITA expression at the transcriptional level is also inhibited in many cancer cell lines (Xia et al., 2016b, 2016c). In addition, a recent study shows that mutation or loss of LKB1, an omnipresently expressed master serine/threonine kinase, results in significant silencing of MITA expression, which is partially mediated by epigenetic silencing enzymes such as DNMT1 and EZH2 (Kitajima et al., 2019). Human and murine primary hepatocytes do not express MITA, which might be responsible for the impaired immune responses to Hepatitis B virus (HBV) infection (Thomsen et al., 2016). In addition, the P-body protein LSm14A was reported to regulate pre-mRNA processing of MITA (Liu et al., 2016). In Lsm14a−/- DCs, mRNA of MITA but not cGAS, TBK1, or IRF3 is remarkably reduced, suggesting that LSm14A is indispensable for maintaining the mRNA level of MITA. These studies have demonstrated epigenetic and post-transcriptional regulation of cGAS and MITA.
Post-translational modification of cGAS
PTMs of cGAS and MITA, including polyubiquitination, phosphorylation, acetylation, SUMOylation, glutamylation, and palmitoylation have been shown to extensively regulate the cGAS-MITA signaling pathway during the resting state and viral infection (Table 1).
Table 1.
Post-translational modifications of cGAS and MITA/STING.
| Protein | Occurring contexts | Enzyme | Modification | Residues | Refs |
|---|---|---|---|---|---|
| Signaling activation | |||||
| cGAS | Resting cells | TRIM38 | SUMOylation | K231 (mouse K217) | Hu et al. (2016) |
| USP29 | Removal of K48-linked polyubiquitination | Mouse K271 | Zhang et al. (2020) | ||
| Stimulated cells | TRIM56 | Monoubiquitination | K335 | Seo et al. (2018) | |
| RINCK | Monoubiquitination | N.D. | Seo et al. (2018) | ||
| TRIM38 | SUMOylation | K479 (mouse K464) | Hu et al. (2016) | ||
| RNF185 | K27-linked polyubiquitination | K173/K384 | Wang et al. (2017) | ||
| USP29 | Removal of K48-linked polyubiquitination | Mouse K271 | Zhang, Bai, and Chen (2020) | ||
| USP14 | Removal of K48-linked polyubiquitination | K414 | Chen, Meng, et al. (2016) | ||
| CCP5 | Removal of monoglutamylation | E302 | Xia, Ye, et al. (2016) | ||
| CCP6 | Removal of polyglutamylation | E272 | Xia, Ye, et al. (2016) | ||
| KAT5 | Acetylation | K47/K56/K62/K83 | Song et al. (2020) | ||
| HDAC3 | deacetylation | N.D. | Dai et al. (2019) | ||
| MITA | Resting cells | EIF3S5 | Removal of K48-linked polyubiquitination | N.D. | Luo et al. (2016) |
| Stimulated cells | RNF26 | K11-linked polyubiquitination | K150 | (Qin et al., 2014) | |
| AMFR | K27-linked polyubiquitination | K137/K150/K224/K236 | Wang et al. (2014) | ||
| MUL1 | K63-linked polyubiquitination | K224/K236/K289/K338 | Ni et al. (2017) | ||
| TRIM56 | K63-linked polyubiquitination | K150 | Tsuchida et al. (2010) | ||
| TRIM32 | K63-linked polyubiquitination | K20/K150/K224/K236 | Zhang et al. (2012) | ||
| RNF115 | K63-linked polyubiquitination | K20/K224/K289 | Zhong et al. (2020e) | ||
| TRIM38 | SUMOylation | K338 | Hu et al. (2016) | ||
| USP20 | Removal of K48-linked polyubiquitination | N.D. | Zhang et al. (2016) | ||
| CYLD | Removal of K48-linked polyubiquitination | N.D. | Zhang et al. (2018) | ||
| EIF3S5 | Removal of K48-linked polyubiquitination | N.D. | Luo et al. (2016) | ||
| USP44 | Removal of K48-linked polyubiquitination | K236 | Zhang, Tang, et al. (2020) | ||
| OTUD5 | Removal of K48-linked polyubiquitination | K347 | Guo et al. (2021) | ||
| CSK | Phosphorylation | Y240/Y245 | Gao et al. (2020) | ||
| EGFR | Phosphorylation | Y245 | Wang et al. (2020) | ||
| TBK1 | Phosphorylation | S366 | Zhang et al. (2019) | ||
| N.D. | Palmitoylation | C88/C91 | Mukai et al. (2016) | ||
| Signaling inhibition | |||||
| cGAS | Resting cells | CDK1 | Phosphorylation | S305 (mouse S291) | Zhong et al. (2020) |
| AurB | Phosphorylation | N.D. | Li et al. (2021) | ||
| TTLL4 | monoglutamylation | E302 | Xia, Ye, et al. (2016) | ||
| TTLL6 | polyglutamylation | E272 | Xia, Ye, et al. (2016) | ||
| PPP6C | Dephosphorylation | S435 (mouse S420) | Li and Shu (2020) | ||
| N.D | Acetylation | K384/K394/K414 | Dai et al. (2019) | ||
| Stimulated cells | AKT | Phosphorylation | S305 (mouse S291) | Seo et al. (2015) | |
| DNA-PK | Phosphorylation | T68/S213 | Sun et al. (2020) | ||
| MITA | Resting cells | USP13 | Removal of K27-linked polyubiquitination | N.D. | Sun et al. (2017) |
| Stimulated cells | USP13 | Removal of K27-linked polyubiquitination | N.D. | Sun et al. (2017) | |
| USP49 | Removal of K63-linked polyubiquitination | N.D. | Ye et al. (2019) | ||
| USP21 | Removal of K27/K63-linked polyubiquitination | N.D. | (Chen et al., 2017) | ||
| N.D. | Nitro-alkylation | C88/C91/H16 | Hansen et al. (2018) | ||
| PPM1A | Dephosphorylation | S358 | Li et al. (2015) | ||
| Degradation | |||||
| cGAS | Stimulated cells | SENP2 | DeSUMOylation | K479 (mouse K464) | Hu et al. (2016) |
| MITA | Stimulated cells | SENP2 | DeSUMOylation | K338 | Hu et al. (2016) |
| TRIM30α | K48-linked polyubiquitination | K275 | Wang et al. (2015) | ||
| RNF5 | K48-linked polyubiquitination | K150 | Zhong et al. (2009) | ||
| TRIM29 | K48-linked polyubiquitination | K370 | Xing et al. (2017) | ||
| ULK1 | Phosphorylation | S366 | Konno et al. (2013) | ||
| PTPN1/2 | Dephosphorylation | Y245 | Xia et al. (2019) | ||
In resting cells, tubulin tyrosine ligase-like 6 (TTLL6) and TTLL4 catalyze polyglutamylation and monoglutamylation of cGAS, resulting in inhibition of its DNA-binding ability and synthase activity, respectively (Xia, Ye, et al., 2016). cGAS also undergoes acetylation at K384, K394, or K414, which keeps cGAS inactive without DNA challenges, though the enzyme responsible for this process is unknown (Dai et al., 2019). During mitosis (as described above), cyclin-dependent kinase 1 (CDK1) and mitotic kinases, including Aurora kinase B (AurB), phosphorylates cGAS to block its ability of self-DNA detection, which helps to prevent autoimmunity (Li et al., 2021; Zhong et al., 2020). PPP6C constitutively dephosphorylates mcGAS in un-infected cells at S420, impairing its ability to bind to GTP and thereby leading to its inactivation (Li & Shu, 2020). Moreover, cGAS is constitutively ubiquitinated at K271 and undergoes degradation in resting cells. TRIM38 catalyzes SUMOylation of cGAS at K217, which inhibits K48-linked polyubiquitination of cGAS at K271 through the physical/spatial hindrance, and ultimately stabilizes cGAS (Hu et al., 2016). The deubiquitinase ubiquitin Specific Protease 29 (USP29) constitutively targets cGAS for deubiquitination at K271, thereby stabilizing cGAS in uninfected cells (Zhang, Tang, et al., 2020). However, the E3 ligases that regulate the ubiquitination of homeostatic cGAS remain to be identified.
Viral infection induces a vast number of post-translational modifications of cGAS (Table 1). Cytosolic carboxypeptidase 5 (CCP5) removes the monoglutamylation of cGAS, whereas CCP6 hydrolyzes the polyglutamylation of cGAS, both of which lead to activation of cGAS in response to DNA challenges (Xia, Ye, et al., 2016). The acetylation exerted by lysine acetyltransferase 5 (KAT5) at the N-terminal domain of cGAS is indispensable to initiate the immune responses in stimulated cells, whereas the acetylation of the C-terminal of cGAS blocks the activity of cGAS (Dai et al., 2019; Song et al., 2020), suggesting that the acetylation at different domains of cGAS may lead to a different function of cGAS. Besides, the E3 ubiquitin ligase TRIM14 recruits deubiquitinase USP14 to remove K48-linked ubiquitin chains and thereby inhibits degradation of cGAS by autophagic pathway in HSV-1 infected cells (Chen, Meng, et al., 2016). Interestingly, we have recently reported that HSV-1 infection induces the degradation of cGAS via the proteasomal pathway, which is counteracted by the deubiquitinase USP29 (Zhang, Tang, et al., 2020). It is thus likely that the stability of cGAS after viral infection is controlled by multiple mechanisms.
Two studies revealed that the E3 ubiquitin ligase TRIM56 and RINCK target cGAS for monoubiquitination, thereby positively regulating the synthesis of cGAMP (Liu, Zhang, Cai, et al., 2018; Seo et al., 2018). TRIM38 also specifically catalyzes the SUMOylation of cGAS to prevent the proteasomal degradation of cGAS at the early phase of viral infection. Subsequently, SENP2 de-SUMOylates cGAS at the late phase of infection to shut down the immune responses (Hu et al., 2016), demonstrating an elegant temporal step-wise control of cGAS activity and stability. The kinases AKT and DNA-PK phosphorylate cGAS during infection of DNA virus, which robustly suppresses its enzymatic activity and fine-tunes immune responses to DNA stimulation (Seo et al., 2015; Sun et al., 2020). Future investigations are required to elucidate whether and how these PTMs function cooperatively or redundantly to regulate the activity and stability of cGAS in a cell-type or stimuli-dependent manner.
Post-translational modification of MITA/STING
Resting-state MITA is regulated by ubiquitination and deubiquitination. The K27 linked-polyubiquitin chains are removed by the deubiquitinating enzyme (DUB) ubiquitin-specific protease 13 (USP13), which inhibits the basal activity of MITA in a manner dependent on blocking its recruitment of TBK1 (Sun et al., 2017). Moreover, inactive rhomboid protein 2 (iRhom2) recruits the deubiquitination enzyme EIF3S5 to constitutively deconjugate K48-linked polyubiquitin chains from MITA and prevent its degradation through the proteasomal pathways (Luo et al., 2016). Recently, one report has shown that death-associated protein kinase 3 (DAPK3) stabilizes MITA in resting cells via suppressing its K48-linked poly-ubiquitination and degradation of proteasome pathway (Takahashi et al., 2021). However, the E3 ligases that regulate the ubiquitination of homeostatic MITA have not been identified yet.
Though the binding of cGAMP leads to dimerization of MITA, the full activation of MITA requires various PTMs accompanied by budding off the ER to ERGIC. HSV-1-infection induces K27-linked polyubiquitin of MITA and such a PTM is mediated by AMFR, which provides an anchoring platform for recruiting TBK1 (Wang et al., 2014). It has been shown that TRIM56, TRIM32, and MUL1 catalyze K63-linked ubiquitination of MITA, which promotes the oligomerization and full activation of MITA (Ni et al., 2017; Tsuchida et al., 2010; Zhang et al., 2012). However, studies with TRIM56 and TRIM32 knockout mice suggest that deletion either of them has minimal effect on ubiquitination of MITA after HSV-1 infection (Seo et al., 2018; Yang, Liu, et al., 2017). Whether these two enzymes function redundantly for the ubiquitination of MITA is unknown. More recently, we have demonstrated that knockout of RNF115 impairs HSV-1-induced K63-linked ubiquitination of MITA and the oligomerization of MITA, suggesting that RNF115 is a bona fide E3 ligase for the K63-linked ubiquitination of MITA after HSV-1 infection (Zhang, Xiong, et al., 2020). In contrast, cGAMP- or HSV-1-induced dimerization of MITA is not affected by the knockout of RNF115, indicating that the oligomerization of MITA is followed by cGAMP-mediated dimerization and requires 63-mediated ubiquitination.
The MITA dimers or oligomers bud off the ER and are trafficking to the Golgi apparatus where it undergoes palmitoylation to activate downstream signaling cascades (Mukai et al., 2016). Such a PTM is suppressed by nitro-alkylation modified by endogenously formed nitro-fatty acids in the late phase of viral infection (Hansen et al., 2018; Mukai et al., 2016). Afterward, TBK1 is recruited to and phosphorylates MITA at S366, thereby promoting the recruitment and phosphorylation of IRF3 (Zhang et al., 2019). Consistently, the S366A mutation in MITA was found to be unable to interact with and activate IRF3 in infected cells. However, the MITAS365A (an ortholog of hMITAS366A) mice do not show hypersensitivity to HSV-1 infection compared to the wild-type mice (Yamashiro et al., 2020; Yum et al., 2021), indicating a TBK1-independent role of MITA for anti-viral immune responses. In this context, MITA also promotes autophagy to clear viral DNA which is believed as a primordial function of MITA (Gui et al., 2019). Other kinases including DAPK3 and CSK are also involved in the phosphorylation and activation of MITA (Gao et al., 2020; Takahashi et al., 2021; Wang et al., 2020). Collectively, these data indicate a sequential multi-step activation of MITA after viral infection, i.e., cGAMP-mediated dimerization, ubiquitination-mediated oligomerization, palmitoylation- and phosphorylation-mediated recruitment of TBK1 and IRF3.
To avoid overwhelming activation of MITA and harmful immune responses, suitable PTMs are essential to tune down the activation of MITA. The E3 ligases RNF5, TRIM30α, and TRIM29 catalyze K48-linked ubiquitination and proteasomal degradation of MITA at the late stage of viral infection, which is counteracted by USP20, USP44, CYLD, EIF3S5, and OTUD5 (Guo et al., 2021; Luo et al., 2016; Wang et al., 2015; Xing et al., 2017; Xu et al., 2020; Zhang et al., 2016, 2018; Zhong et al., 2009). USP49 removes K63-linked polyubiquitination of MITA, thereby negatively regulating the signaling pathway (Ye et al., 2019). Besides, serine/threonine UNC-51-like kinase (ULK1) phosphorylates MITA after the activation of IRF3 and NF-κB during the infection of HSV-1, which finally avoids sustained production of inflammatory cytokines (Konno et al., 2013). Phosphatase PPM1A and PTPN1/2 also act as negative regulators by dephosphorylation of MITA to avert hyperactivation of the MITA-mediated immune responses (Li et al., 2015; Xia et al., 2019). Together, although a mass of enzymes and their corresponding PTMs have been identified to covalently modify MITA (Table 1) during different phases of viral infection, further work in this field is needed to elucidate how different PTMs crosstalk and dynamically regulate the activity and stability of MITA.
cGAS-MITA/STING pathway in autoimmune disease
As described above, cGAS is inactive or kept away from self-DNA under homeostatic conditions. Accordingly, dysregulation of such a process would result in constitutively and spontaneously activation of cGAS, which promotes the upregulation of proinflammatory cytokines and induces autoimmune disorders. MITA is kept in ER as monomer under homeostatic conditions. Removal of the ER-retention factors or gain-of-function mutations of MITA would lead to constitutive activation of MITA and auto-inflammatory diseases.
DNase and RNase mutations
To identify endogenous DNA sensors, Stetson et al. have identified TREX1 as a sensor of cytosolic DNA (Stetson et al., 2008). However, TREX1 does not trigger activation of IRF3 and the induction of type I IFNs. Rather, the knockout of TREX1 leads to the accumulation of cytosolic DNA and sustained autoantibody production and inflammation. Trex1 is a 3′→5′ DNA exonuclease and consists of the N-terminal DNase domain necessary for exonuclease activity and the C-terminal transmembrane helix necessary for ER localization. Accumulative DNA in the cytoplasm from various sources, including occasional leakage of nuclear DNA, the release of mtDNA, and DNA from reverse-transcribed endogenous RNA, can be recognized and digested by TREX1 (Fig. 1). A number of clinical studies reveal that mutations in TREX1 have been identified in autoimmune diseases including Aicardi-Goutieres syndrome (AGS), systemic lupus erythematosus (SLE), and retinal vasculopathy with cerebral leukodystrophy (RVCL) (Crow & Manel, 2015; Grieves et al., 2015; Hasan et al., 2015; Hemphill et al., 2021; Stetson et al., 2008). These mutations in both N-terminal and C-terminal domain result in loss-of-function of TREX1 and the abnormal accumulation of cytosolic DNA and ultimately constant activation of the cGAS-MITA pathway. Similarly, Trex1-deficient or -mutated (Trex1D18N/D18N) mice exhibit the lethal autoimmune phenotype, which can be genetically rescued by depletion of cGAS or MITA (Gao et al., 2015; Morita et al., 2004; Simpson et al., 2020; Stetson et al., 2008; Xiao et al., 2019). These observations suggest that the activation of the cGAS-MITA pathway mediates the autoimmune phenotypes in Trex1–/– or Trex1D18N/D18N mice.
Deoxyribonuclease (DNase) II encoded by DNASE2, an endonuclease that digests ssDNA and dsDNA in the lysosome, is also related to autoimmune diseases in humans (Hong et al., 2020; Rodero et al., 2017). Biallelic mutations in DNASE2 result in deficiency of Dnase II endonuclease activity, consequently leading to overproduction of interferons and ISGs in the lymphocytes and monocytes. Similarly, deletion of cGas, Mita, or Ifnar1 rescues the lethal autoimmune phenotypes of DNaseII−/− mice (Ahn et al., 2012; Gao et al., 2015).
Several mutations in ribonuclease H2 (RNase H2) are also identified in patients with AGS syndrome (Gunther et al., 2015; Nishimura et al., 2019). RNase H2 removes RNA/DNA hetero-stranded nucleic acids, such as RNA embedded in double-stranded DNA during DNA replication and R loops formed during transcription structure. Rnaseh2aG37S and Rnaseh2aA174T are activity-attenuated mutants. The Rnaseh2aG37S and Rnaseh2aA174T knock-in mice exhibit increased expression of interferons and ISGs and autoimmunity, which can be rescued by deletion of MITA (Mackenzie et al., 2016; Pokatayev et al., 2016). A subset of AGS patients carry mutations in three other genes including SAMHD1, ADAR1, and IFIH1, and exhibits the phenotypes of upregulation of ISGs (Coggins et al., 2020; Oda et al., 2014; Rice et al., 2009, 2012). However, whether such symptom is dependent on cGAS-MITA is unknown. In addition, the gain-of-function mutations of cGAS have not been identified.
MITA/STINGgain-of-functionmutations
In 2014, a clinical study reported the discovery of MITA gain-of-function mutations (MITA N154S and V155M) in six patients exhibiting systemic inflammation, scaling lesions of extremities and cheeks, and interstitial lung disease, which is termed STING-associated vasculopathy with onset in infancy (SAVI) (Liu et al., 2014). Over the past 7 years, other gain-of-function mutations of MITA have been identified, including S102P/F279L, V147L, G166E, C206Y, R281Q, R284Q, R284G, and R284S (Chia et al., 2016; Konig et al., 2017; Konno et al., 2018; Melki et al., 2017; Motwani et al., 2019a, 2019b; Munoz et al., 2015; Saldanha et al., 2018; Seo et al., 2017). All of the mutant MITA spontaneously dimerize and bud off from the ER to the Golgi apparatus even in the absence of cGAMP. Continuous accumulation of mutant MITA in the Golgi apparatus leads to activation of TBK1 and IRF3, which results in the production of interferons and ISGs. Furthermore, homozygous knock-in mice carrying MITAN153S mutation (human MITA N154S ortholog) can not survive gestation, whereas heterozygous MITAN153S and MITAV154M (human MITA V155M ortholog) mice develop T cell cytopenia, myeloid cell expansion, spontaneous pulmonary and renal inflammation, similar to SAVI patients (Bennion et al., 2019; Bouis et al., 2019; Warner et al., 2017). MITAN153S mice also exhibit defects in lymph-node organogenesis and innate lymphoid cell development (Bennion et al., 2020). However, the overwhelming type I IFN gene expression is not observed in the MITAN153S mice, which has been reported in patients with SAVI. In contrast, activation of the functional NF-κB pathway plays a dominant role in the SAVI-like disease of MITAN153S mice. The different phenotypes between the SAVI patients and the gain-of-function MITA mutated mice indicate different regulatory mechanisms of human and mouse MITA. One recent study shows that MITAN153S mice develop intestinal inflammation in the colon in a manner dependent on the abnormal accumulation of MITA in myeloid cells, suggesting that MITAN153S could be the potential target for patients with inflammatory bowel disease (IBD) (Shmuel-Galia et al., 2021). Collectively, these studies indicate that abnormal activation of the MITA pathway contributes to various autoimmune diseases, suggesting that therapeutic targeting of these responses could be beneficial for the treatment of the related diseases.
cGAS-MITA/STING pathway in cancer
In recent years, emerging studies have implied the important regulatory function of the cGAS-MITA signaling pathway in tumors. Several small-molecule compounds as agonists for MITA have been developed for clinical trials to test their effects on cancer prevention.
Tumor suppressive role
Accumulating evidence has shown that the cGAS-MITA pathway plays a tumor-suppressive role (Fig. 2). One of the most important features of cancer cells is chromosomal instability (CIN) that leads to chromosomal missegregation during mitosis, which ultimately promotes the formation of micronuclei (Crasta et al., 2012). The micronuclear envelopes are subject to break up, which causes the accumulation of genomic DNA in the cytoplasm (Harding et al., 2017; Mackenzie et al., 2017). Alternatively, radiotherapies, and chemo-reagents such as cisplatin, and intrinsic DNA damage also lead to the production of cytoplasmic DNA (Ahn et al., 2014; Dou et al., 2017; Harding et al., 2017; Mackenzie et al., 2017). Besides, oxidative stress in cancer cells causes mitochondrial damage by permeabilization of the inner and outer mitochondrial membranes, consequently leading to the release of mtDNA in the cytoplasm (Sansone et al., 2017; Tan et al., 2015). When the DNA in the cytoplasm accumulates above the threshold, it would activate the cGAS-MITA pathway that upregulates the production of type I IFNs to promote the infiltration and activation of immune cells, such as T cells and nature killer (NK) cells to elicit host immune responses (Harlin et al., 2009; Marcus et al., 2018). Moreover, the cGAS-MITA signaling upregulates the senescence-associated secretory phenotype (SASP) genes including inflammatory cytokines, chemokines, growth factors, and proteases, thereby facilitating senescence of cancer cells and tumor clearance (Dou et al., 2017; Kwon & Bakhoum, 2020; Yang, Wang, et al., 2017). Alternatively, tumor-derived DNA or cGAMP that is engulfed by surrounding DCs activates cGAS-MITA signaling and promotes IFN-β production. Subsequently, IFN-β promotes the cross-presentation of CD8α+ DC subset for activation of tumor antigen-specific CD8+ T cells (Woo et al., 2014). Consistently, mice that lack IFNAR1 in DCs display significant defects in antigen cross-presentation to CD8+ T cells (Diamond et al., 2011). DC or tumor-derived IFN-β also activates the NK cells in a manner dependent on IFNAR signaling in NK cells (Marcus et al., 2018). Ultimately, activated T cells and NK cells in turn traffic to tumor sites and promote tumor suppression.
Fig. 2.
Regulation of cGAS-MITA/STINGpathway in tumor suppression. In early neoplastic cells, the cGAS-MITA pathway plays a tumor-suppressive role. DNA damage sourced from such as oxidative stress, radiation, CIN, or cisplatin leads to DNA accumulate in the cytoplasm, which activates the cGAS and promotes the synthesis of cGAMP. On one hand, cGAMP binds to MITA to upregulate the expression of type I IFNs and SASP genes in tumors, which in turn mediates tumor-suppressive effects. On the other hand, the cGAS-MITA pathway could mediate the crosstalk between the tumor cells and DCs nearby. Tumor DNA or tumor-derived cGAMP could activate the cGAS-MITA signaling in DCs, finally initiating tumor clearance mediated by immune cells such as CD8+ T cells or NK cells. CIN, chromosomal instability; cGAMP, 2′,3′-cyclic GMP-AMP; IFN, interferon; SASP, senescence-associated secretory phenotype; DC, dendritic cell; NK cells, natural killer cells. IFN-R, IFN receptor.
Under selective pressure, some surviving cancer cells are prone to harbor cGAS or MITA deficiency. MITA expression has been reported to be undetectable in several cancerous melanomas and colorectal adenocarcinoma cell lines (Xia et al., 2016b, 2016c). Moreover, KRAS-driven lung cancers markedly silence MITA expression owing to deficiency of LKB1 (Kitajima et al., 2019). The deficiency of MLH1 that is responsible for regulating exonuclease 1 nuclease activity results in DNA accumulating in the cytosol and activating the cGAS-MITA pathway (Guan et al., 2021). One study demonstrates that cGAS expression is deficient in human defective MMR gene Mlh1 (dMLH1) cancer lines (Lu et al., 2021), and loss of or impaired cGAS-MITA responses confers defective MMR (dMMR) tumors resistance to immune-checkpoint blockade (ICB) therapy, which suggests that the cGAS-MITA pathway is a potential biomarker for immunotherapy in patients with dMMR cancers.
Considering MITA suppresses tumorigenesis, multiple agonists of MITA have been developed to mimic the activation of the cGAS-MITA pathway for tumor suppression. The agonist of MITA, 5,6-dimethylxanthenone-4 acetic acid (DMXAA), promotes the activation of MITA and destroys the blood vessel walls in tumor tissues, which significantly facilitates the fade of solid tumors in mice (Gao et al., 2014; Zhao et al., 2002). However, clinical trials of treatment DMXAA (also known as ASA404) with non-small cell lung cancer patients are failed in phase III due to tiny therapeutic effects (Lara et al., 2011). It is now clear that DMXAA only binds and activates mouse MITA but not human MITA (Conlon et al., 2013). CDN 3′3′-cGAMP induces the expression of type I interferon in mouse melanoma, liver cancer, and lung cancer cells by activating MITA (Tang et al., 2016). Intraperitoneal injection of 3′3′-cGAMP into Eμ-TCL1 transgenic mice can significantly inhibit the process of chronic lymphocytic leukemia (CLL). In addition, 2′,3′-cGAMP also presents an anti-cancer effect on mouse colon cancer, reducing the size of mouse tumors and prolonging the survival of mice (Yang et al., 2019). Expect for endogenous CDNs, synthetic modified CDNs are well developed. ML RR-S2 CDN (also known as ADU-S100 or MIW815) exhibits significant antitumor efficiency in multiple mouse cancer models and binds human MITA (Corrales et al., 2015). Follow-up clinical trials of ADU-S100 or in combination with checkpoint (CTLA-4 or PD-1) inhibitors are now ongoing to phase I or phase II (Table 2). Other drugs including E7766, BMS-986301, IMSA101, GSK3745417, IMSA101, MK-1454, and SB 11285 are currently undergoing clinical trials and are present in Table 2. Recently, MSA-2 and SR-717, non-nucleotide MITA agonists, were reported to display anti-tumor activity (Chin et al., 2020; Pan et al., 2020). MSA-2 amenable to oral administration induces elevations of IFN-β and almost complete tumor regression in MC38 tumor-bearing mice. Intraperitoneal injection of SR-717 exhibits anti-tumor activity in a manner dependent on activating of CD8+ T cells, NK cells, dendritic cells, and the cross-priming of antigens. However, MSA-2 and SR-717 have not entered into clinical trials yet.
Table 2.
Summary of MITA/STING agonists in clinical trials.
| Agent | Cancer type | Phase | Target | NCT number |
|---|---|---|---|---|
| ASA404 | Adult Solid Tumor | I | MITA | NCT00003697 |
| ASA404 | Hormone Refractory Metastatic Prostate Cancer | II | MITA | NCT00111618 |
| ASA404 + Paclitaxel + Carboplatin | Non-small Cell Lung Cance | I | MITA + microtubule + DNA synthesis | NCT00674102 |
| ASA404 + Paclitaxel + Carboplatin | Locally Advanced and Metastatic NSCLC | I/II | MITA + microtubule + DNA synthesis | NCT00832494 |
| ASA404 | Refractory Tumors | I | MITA | NCT00856336 |
| ASA404 | Solid Tumors | I | MITA | NCT00863733 |
| ASA404 | Advanced or Recurrent Solid Tumors | I | MITA | NCT01285453 |
| ASA404 + Paclitaxel + Carboplatin | Small Cell Lung Cancer | II | MITA + microtubule + DNA synthesis | NCT01057342 |
| ADU-S100 | Head and neck cancer | II | MITA | NCT03937141 |
| ADU-S100 ± ipilimumab | Solid tumors/lymphomas | I | MITA ± CTLA-4 | NCT02675439 |
| ADU-S100 + PDR001 | Solid tumors/lymphomas | I | MITA + PD1 | NCT03172936 |
| E7766 | Urinary bladder neoplasms | I | MITA | NCT04109092 |
| E7766 | Lymphoma/advanced solid tumors | I | MITA | NCT04144140 |
| GSK3745417 | Neoplasms | I | MITA | NCT03843359 |
| MK-1454 | Solid tumors/lymphomas | I | MITA | NCT03010176 |
| MK-1454 + pembrolizumab | Head and neck squamous cell carcinoma | II | MITA + PD1 | NCT04220866 |
| BMS-986301 | Solid cancers | I | MITA | NCT03956680 |
| SB 11285 | Solid tumor | I | MITA | NCT04096638 |
Tumor promoting role
Though cGAS and MITA are lowly expressed in tumor cells, high expression of cGAS and MITA has already been reported to be positively correlated with poor prognosis in a small part of patients with colorectal cancer (An et al., 2019). Chronic inflammation induced by MITA-associated SASP may facilitate oncogene-driven senescence suppression (Fig. 3) (Dou et al., 2017; Toso et al., 2014). Moreover, activation of cGAS-MITA, triggered by DNA derived from CIN-generated micronuclei, induces noncanonical NF-κB signaling responses and metastasis in a tumor cell-autonomous manner (Bakhoum et al., 2018). Alternatively, Brain metastatic cancer cells can utilize gap-junctions to transfer cGAMP to astrocytes, activating MITA and downstream inflammatory genes such as IFN-α and TNF, which, in turn, elicits the STAT1 and NF-κB pathways in brain metastatic cancer cells, ultimately promoting tumor growth and chemoresistance (Chen, Boire, et al., 2016). In addition, activation of MITA enhances infiltration of regulatory T cells and enzyme indoleamine 2,3-dioxygenase (IOD), consequently mediating tolerance of immune response and inhibition of T cells proliferation (Liang et al., 2015; Munn & Mellor, 2016). MITA agonist is able to upregulate the expression of PD-L1, a protein that inhibits the immune response, which promotes tumor progression (Corrales et al., 2016; Fu et al., 2015). Autophagy mediated by MITA is another inducement of immune evasion and tumor promotion (Pommier et al., 2018; Terai et al., 2018). T cells expressing activated MITA exhibit cytopenia phenotype, indicating that constitutive activation of MITA in T cells leads to dysfunction or exhaustion of T cells (Cerboni et al., 2017; Wu et al., 2019). In this context, one recent report has revealed that MITA possesses IFN-independent activities in T cells, which is responsible for tumor immune evasion by inducing T cell death (Wu et al., 2020), though the mechanism by which MITA induces T cell death is unknown now. It is thus likely that the stage and types of tumors are associated with the tumor-suppressive or promoting roles of the cGAS-MITA pathway. Therefore, several agonists showed impressive potential in antitumor immunity due to the suppressive role of MITA in early tumorigenesis. However, prolonged activation of the cGAS-MITA signaling may negligently inhibit antitumor immunity and drive tumor metastasis, which makes the application of MITA agonists more challenging in the clinic. To conquer this challenge, further work is required to unveil the molecular requirements and regulations that function in tumor progression or suppression via identifying the downstream cascade of the cGAS-MITA pathway.
Fig. 3.
Regulation of cGAS-MITA/STING pathway in tumor promotion. The cGAS-MITA pathway exerts its function as a tumor promoter in metastatic tumor cells. Tumors carrying high chromosome instability could promote the formation of micronuclei, which ruptures and release DNA to the cytosol, triggering the activation of the cGAS-MITA signaling. Chronic activation of the pathway promotes the suppression of type I IFNs expression and initiates the upregulation of noncanonical NF-κB signaling, facilitating tumor metastasis. The IDO and pro-inflammatory cytokines together induce the formation of immunosuppressive TME. Moreover, MITA activation could lead to T cell exhaustion in a manner independently of IFN, which also contributes to the maintenance of immunosuppressive TME. MITA may also promote tumor metastasis via PD-L1 upregulation and autophagy process. In addition, tumors can directly transfer the cGAMP to neighboring cells such as astrocytes by gap junctions, ultimately accelerating the progression of tumor metastasis.
Concluding remarks
The discovery of cGAS and MITA has fundamentally advanced the understanding of immune responses mediated by immune-stimulatory DNA. The activation of the cGAS-MITA pathway ensures the sufficient expression of downstream anti-pathogenic genes, whereas excess immune responses lead to tissue damage and immune pathology (Crowl et al., 2017). Thus, it is reasonable that a series of regulatory elements sequentially functions to fine-tune the immune responses in different phases of immunostimulatory DNA stimulation or viral infection. Recent studies showed that cGAS and MITA undergo phase condensation during viral infection, which provides deeper insights into the regulation of their activities (Du & Chen, 2018; Minhas & Holehouse, 2021; Xu et al., 2021; Yu et al., 2021). In this context, studies are required to explore how the regulators crosstalk to each other to dynamically adjust the activities of the cGAS-MITA pathway.
The cGAS-MITA pathway plays a vital role in autoimmune diseases and cancers. TREX1 deficiency and MITA mutations, as we know, lead to autoimmune diseases, such as AGS, SLE, RVCL, and SAVI, suggesting that the cGAS-MITA axis can be targeted for therapeutic intervention of these diseases in the future. However, the activation of the cGAS-MITA pathway in cancers is a double-edged sword. Acute immune responses induced by the cGAS-MITA signaling may function as a barrier to early neoplastic progression, while chronic activation of the pathway may induce the formation of an immunosuppressive tumor environment (TME), eventually leading to tumor growth and metastasis. MITA agonists have been developed to enhance tumor immunogenicity. However, it should be cautious that hyperactivation of MITA signaling may inadvertently worsen the pathology of patients with cancers. Thus, further work is necessary to understand the different regulations behind two different outcomes induced by the cGAS-MITA signaling, to ensure careful selection of patients to proceed personalized therapeutic regimen.
Declaration of competing interest
The authors declare no conflict of interests.
Acknowledgements
Studies in our lab are supported by grants from the National Key Research and Development Program of China (2018TFE0204500 and 2018YFC1004601), Natural Science Foundation of China (32070900), Innovative Research Groups of Natural Science Foundation of Hubei Province (2020CFA015), Fundamental Research Funds for Central Universities (2042020kf0207), Medical Science Advancement Program (Basic Medical Sciences) of Wuhan University (TFJC2018004), and the Non-Profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2020-PT320-004).
References
- Aguirre S., Luthra P., Sanchez-Aparicio M.T., Maestre A.M., Patel J., Lamothe F., Fredericks A.C., Tripathi S., Zhu T., Pintado-Silva J., et al. Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection. Nature Microbiology. 2017;2 doi: 10.1038/nmicrobiol.2017.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahn J., Barber G.N. Self-DNA, STING-dependent signaling and the origins of autoinflammatory disease. Current Opinion in Immunology. 2014;31:121–126. doi: 10.1016/j.coi.2014.10.009. [DOI] [PubMed] [Google Scholar]
- Ahn J., Gutman D., Saijo S., Barber G.N. STING manifests self DNA-dependent inflammatory disease. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:19386–19391. doi: 10.1073/pnas.1215006109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ahn J., Xia T., Konno H., Konno K., Ruiz P., Barber G.N. Inflammation-driven carcinogenesis is mediated through STING. Nature Communications. 2014;5:5166. doi: 10.1038/ncomms6166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andreeva L., Hiller B., Kostrewa D., Lassig C., de Oliveira Mann C.C., 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. [DOI] [PubMed] [Google Scholar]
- An X., Zhu Y., Zheng T., Wang G., Zhang M., Li J., Ji H., Li S., Yang S., Xu D., et al. An analysis of the expression and association with immune cell infiltration of the cGAS/STING pathway in pan-cancer. Molecular Therapy - Nucleic Acids. 2019;14:80–89. doi: 10.1016/j.omtn.2018.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bakhoum S.F., Ngo B., Laughney A.M., Cavallo J.A., Murphy C.J., Ly P., Shah P., Sriram R.K., Watkins T.B.K., Taunk N.K., et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature. 2018;553:467–472. doi: 10.1038/nature25432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barnett K.C., Coronas-Serna J.M., Zhou W., Ernandes M.J., Cao A., Kranzusch P.J., Kagan J.C. Phosphoinositide interactions position cGAS at the plasma membrane to ensure efficient distinction between self- and viral DNA. Cell. 2019;176:1432–1446. doi: 10.1016/j.cell.2019.01.049. e1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennion B.G., Croft C.A., Ai T.L., Qian W., Menos A.M., Miner C.A., Fremond M.L., Doisne J.M., Andhey P.S., Platt D.J., et al. STING gain-of-function disrupts lymph node organogenesis and innate lymphoid cell development in mice. Cell Reports. 2020;31 doi: 10.1016/j.celrep.2020.107771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennion B.G., Ingle H., Ai T.L., Miner C.A., Platt D.J., Smith A.M., Baldridge M.T., Miner J.J. A human gain-of-function STING mutation causes immunodeficiency and gammaherpesvirus-induced pulmonary fibrosis in mice. Journal of Virology. 2019;93 doi: 10.1128/JVI.01806-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouis D., Kirstetter P., Arbogast F., Lamon D., Delgado V., Jung S., Ebel C., Jacobs H., Knapp A.M., Jeremiah N., et al. Severe combined immunodeficiency in stimulator of interferon genes (STING) V154M/wild-type mice. The Journal of Allergy and Clinical Immunology. 2019;143:712–725. doi: 10.1016/j.jaci.2018.04.034. e715. [DOI] [PubMed] [Google Scholar]
- Boyer J.A., Spangler C.J., Strauss J.D., Cesmat A.P., Liu P., McGinty R.K., Zhang Q. Structural basis of nucleosome-dependent cGAS inhibition. Science. 2020;370:450–454. doi: 10.1126/science.abd0609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burdette D.L., Monroe K.M., Sotelo-Troha K., Iwig J.S., Eckert B., Hyodo M., Hayakawa Y., Vance R.E. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478:515–518. doi: 10.1038/nature10429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burdette D.L., Vance R.E. STING and the innate immune response to nucleic acids in the cytosol. Nature Immunology. 2013;14:19–26. doi: 10.1038/ni.2491. [DOI] [PubMed] [Google Scholar]
- Cai Z., Zhang M.X., Tang Z., Zhang Q., Ye J., Xiong T.C., Zhang Z.D., Zhong B. USP22 promotes IRF3 nuclear translocation and antiviral responses by deubiquitinating the importin protein KPNA2. Journal of Experimental Medicine. 2020;217 doi: 10.1084/jem.20191174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao D., Han X., Fan X., Xu R.M., Zhang X. Structural basis for nucleosome-mediated inhibition of cGAS activity. Cell Research. 2020;30:1088–1097. doi: 10.1038/s41422-020-00422-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cerboni S., Jeremiah N., Gentili M., Gehrmann U., Conrad C., Stolzenberg M.C., Picard C., Neven B., Fischer A., Amigorena S., et al. Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes. Journal of Experimental Medicine. 2017;214:1769–1785. doi: 10.1084/jem.20161674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q., Boire A., Jin X., Valiente M., Er E.E., Lopez-Soto A., Jacob L., Patwa R., Shah H., Xu K., et al. Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533:493–498. doi: 10.1038/nature18268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen M., Meng Q., Qin Y., Liang P., Tan P., He L., Zhou Y., Chen Y., Huang J., Wang R.F., et al. TRIM14 inhibits cGAS degradation mediated by selective autophagy receptor p62 to promote innate immune responses. Molecular Cell. 2016;64:105–119. doi: 10.1016/j.molcel.2016.08.025. [DOI] [PubMed] [Google Scholar]
- Chen Q., Sun L., Chen Z.J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nature Immunology. 2016;17:1142–1149. doi: 10.1038/ni.3558. [DOI] [PubMed] [Google Scholar]
- Chia J., Eroglu F.K., Ozen S., Orhan D., Montealegre-Sanchez G., de Jesus A.A., Goldbach-Mansky R., Cowen E.W. Failure to thrive, interstitial lung disease, and progressive digital necrosis with onset in infancy. Journal of the American Academy of Dermatology. 2016;74:186–189. doi: 10.1016/j.jaad.2015.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chin E.N., Yu C., Vartabedian V.F., Jia Y., Kumar M., Gamo A.M., Vernier W., Ali S.H., Kissai M., Lazar D.C., et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science. 2020;369:993–999. doi: 10.1126/science.abb4255. [DOI] [PubMed] [Google Scholar]
- Chiu Y.H., Macmillan J.B., Chen Z.J. RNA polymerase III detects cytosolic DNA and induces type I interferons through the RIG-I pathway. Cell. 2009;138:576–591. doi: 10.1016/j.cell.2009.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu T.T., Tu X., Yang K., Wu J., Repa J.J., Yan N. Tonic prime-boost of STING signalling mediates Niemann-Pick disease type C. Nature. 2021;596:570–575. doi: 10.1038/s41586-021-03762-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coggins S.A., Mahboubi B., Schinazi R.F., Kim B. SAMHD1 functions and human diseases. Viruses. 2020;12 doi: 10.3390/v12040382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conlon J., Burdette D.L., Sharma S., Bhat N., Thompson M., Jiang Z., Rathinam V.A., Monks B., Jin T., Xiao T.S., et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. The Journal of Immunology. 2013;190:5216–5225. doi: 10.4049/jimmunol.1300097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corrales L., Glickman L.H., McWhirter S.M., Kanne D.B., Sivick K.E., Katibah G.E., Woo S.R., Lemmens E., Banda T., Leong J.J., et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Reports. 2015;11:1018–1030. doi: 10.1016/j.celrep.2015.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corrales L., McWhirter S.M., Dubensky T.W., Jr., Gajewski T.F. The host STING pathway at the interface of cancer and immunity. Journal of Clinical Investigation. 2016;126:2404–2411. doi: 10.1172/JCI86892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crasta K., Ganem N.J., Dagher R., Lantermann A.B., Ivanova E.V., Pan Y., Nezi L., Protopopov A., Chowdhury D., Pellman D. DNA breaks and chromosome pulverization from errors in mitosis. Nature. 2012;482:53–58. doi: 10.1038/nature10802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crowl J.T., Gray E.E., Pestal K., Volkman H.E., Stetson D.B. Intracellular nucleic acid detection in autoimmunity. Annual Review of Immunology. 2017;35:313–336. doi: 10.1146/annurev-immunol-051116-052331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crow Y.J., Manel N. Aicardi-Goutieres syndrome and the type I interferonopathies. Nature Reviews Immunology. 2015;15:429–440. doi: 10.1038/nri3850. [DOI] [PubMed] [Google Scholar]
- Dai J., Huang Y.J., He X., Zhao M., Wang X., Liu Z.S., Xue W., Cai H., Zhan X.Y., Huang S.Y., et al. Acetylation blocks cGAS activity and inhibits self-DNA-induced autoimmunity. Cell. 2019;176:1447–1460. doi: 10.1016/j.cell.2019.01.016. e1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng Z., Chong Z., Law C.S., Mukai K., Ho F.O., Martinu T., Backes B.J., Eckalbar W.L., Taguchi T., Shum A.K. A defect in COPI-mediated transport of STING causes immune dysregulation in COPA syndrome. Journal of Experimental Medicine. 2020;217 doi: 10.1084/jem.20201045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diamond M.S., Kinder M., Matsushita H., Mashayekhi M., Dunn G.P., Archambault J.M., Lee H., Arthur C.D., White J.M., Kalinke U., et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. Journal of Experimental Medicine. 2011;208:1989–2003. doi: 10.1084/jem.20101158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong G., You M., Ding L., Fan H., Liu F., Ren D., Hou Y. STING negatively regulates double-stranded DNA-activated JAK1-STAT1 signaling via SHP-1/2 in B cells. Molecular Cell. 2015;38:441–451. doi: 10.14348/molcells.2015.2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dou Z., Ghosh K., Vizioli M.G., Zhu J., Sen P., Wangensteen K.J., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du M., Chen Z.J. DNA-induced liquid phase condensation of cGAS activates innate immune signaling. Science. 2018;361:704–709. doi: 10.1126/science.aat1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang R., Jiang Q., Guan Y., Gao P., Zhang R., Zhao Z., Jiang Z. Golgi apparatus-synthesized sulfated glycosaminoglycans mediate polymerization and activation of the cGAMP sensor STING. Immunity. 2021;54:962–975. doi: 10.1016/j.immuni.2021.03.011. e968. [DOI] [PubMed] [Google Scholar]
- Fu J., Kanne D.B., Leong M., Glickman L.H., McWhirter S.M., Lemmens E., Mechette K., Leong J.J., Lauer P., Liu W., et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Science Translational Medicine. 2015;7 doi: 10.1126/scitranslmed.aaa4306. 283ra252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gallego-Marin C., Schrum J.E., Andrade W.A., Shaffer S.A., Giraldo L.F., Lasso A.M., Kurt-Jones E.A., Fitzgerald K.A., Golenbock D.T. Cyclic GMP-AMP synthase is the cytosolic sensor of Plasmodium falciparum genomic DNA and activates type I IFN in malaria. The Journal of Immunology. 2018;200:768–774. doi: 10.4049/jimmunol.1701048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao P., Hu M.M., Shu H.B. CSK promotes innate immune response to DNA virus by phosphorylating MITA. Biochemical and Biophysical Research Communications. 2020;526:199–205. doi: 10.1016/j.bbrc.2020.03.069. [DOI] [PubMed] [Google Scholar]
- Gao D., Li T., Li X.D., Chen X., Li Q.Z., Wight-Carter M., Chen Z.J. Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E5699–E5705. doi: 10.1073/pnas.1516465112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao D., Wu J., Wu Y.T., Du F., Aroh C., Yan N., Sun L., Chen Z.J. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science. 2013;341:903–906. doi: 10.1126/science.1240933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao P., Zillinger T., Wang W., Ascano M., Dai P., Hartmann G., Tuschl T., Deng L., Barchet W., Patel D.J. Binding-pocket and lid-region substitutions render human STING sensitive to the species-specific drug DMXAA. Cell Reports. 2014;8:1668–1676. doi: 10.1016/j.celrep.2014.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grieves J.L., Fye J.M., Harvey S., Grayson J.M., Hollis T., Perrino F.W. Exonuclease TREX1 degrades double-stranded DNA to prevent spontaneous lupus-like inflammatory disease. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:5117–5122. doi: 10.1073/pnas.1423804112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guan J., Lu C., Jin Q., Lu H., Chen X., Tian L., Zhang Y., Ortega J., Zhang J., Siteni S., et al. MLH1 deficiency-triggered DNA hyperexcision by exonuclease 1 activates the cGAS-STING pathway. Cancer Cell. 2021;39:109–121. doi: 10.1016/j.ccell.2020.11.004. e105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guey B., Wischnewski M., Decout A., Makasheva K., Kaynak M., Sakar M.S., Fierz B., Ablasser A. BAF restricts cGAS on nuclear DNA to prevent innate immune activation. Science. 2020;369:823–828. doi: 10.1126/science.aaw6421. [DOI] [PubMed] [Google Scholar]
- Gui X., Yang H., Li T., Tan X., Shi P., Li M., Du F., Chen Z.J. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature. 2019;567:262–266. doi: 10.1038/s41586-019-1006-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunther C., Kind B., Reijns M.A., Berndt N., Martinez-Bueno M., Wolf C., Tungler V., Chara O., Lee Y.A., Hubner N., et al. Defective removal of ribonucleotides from DNA promotes systemic autoimmunity. Journal of Clinical Investigation. 2015;125:413–424. doi: 10.1172/JCI78001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo Y., Jiang F., Kong L., Wu H., Zhang H., Chen X., Zhao J., Cai B., Li Y., Ma C., et al. OTUD5 promotes innate antiviral and antitumor immunity through deubiquitinating and stabilizing STING. Cellular and Molecular Immunology. 2021;18:1945–1955. doi: 10.1038/s41423-020-00531-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hahn W.O., Butler N.S., Lindner S.E., Akilesh H.M., Sather D.N., Kappe S.H., Hamerman J.A., Gale M., Jr., Liles W.C., Pepper M. cGAS-mediated control of blood-stage malaria promotes Plasmodium-specific germinal center responses. JCI Insight. 2018;3 doi: 10.1172/jci.insight.94142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen A.L., Buchan G.J., Ruhl M., Mukai K., Salvatore S.R., Ogawa E., Andersen S.D., Iversen M.B., Thielke A.L., Gunderstofte C., et al. Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling. Proceedings of the National Academy of Sciences of the United States of America. 2018;115:E7768–E7775. doi: 10.1073/pnas.1806239115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harding S.M., Benci J.L., Irianto J., Discher D.E., Minn A.J., Greenberg R.A. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature. 2017;548:466–470. doi: 10.1038/nature23470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harlin H., Meng Y., Peterson A.C., Zha Y., Tretiakova M., Slingluff C., McKee M., Gajewski T.F. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Research. 2009;69:3077–3085. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasan M., Fermaintt C.S., Gao N., Sakai T., Miyazaki T., Jiang S., Li Q.Z., Atkinson J.P., Morse H.C., 3rd, Lehrman M.A., et al. Cytosolic nuclease TREX1 regulates oligosaccharyltransferase activity independent of nuclease activity to suppress immune activation. Immunity. 2015;43:463–474. doi: 10.1016/j.immuni.2015.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hemphill W.O., Simpson S.R., Liu M., Salsbury F.R., Jr., Hollis T., Grayson J.M., Perrino F.W. TREX1 as a novel immunotherapeutic target. Frontiers in Immunology. 2021;12 doi: 10.3389/fimmu.2021.660184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hong Y., Capitani M., Murphy C., Pandey S., Cavounidis A., Takeshita H., Nanthapisal S., Yasuda T., Bader-Meunier B., McCreary D., et al. Janus kinase inhibition for autoinflammation in patients with DNASE2 deficiency. The Journal of Allergy and Clinical Immunology. 2020;145:701–705. doi: 10.1016/j.jaci.2019.11.020. e708. [DOI] [PubMed] [Google Scholar]
- Hooy R.M., Massaccesi G., Rousseau K.E., Chattergoon M.A., Sohn J. Allosteric coupling between Mn2+ and dsDNA controls the catalytic efficiency and fidelity of cGAS. Nucleic Acids Research. 2020;48:4435–4447. doi: 10.1093/nar/gkaa084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang Y.H., Liu X.Y., Du X.X., Jiang Z.F., Su X.D. The structural basis for the sensing and binding of cyclic di-GMP by STING. Nature Structural & Molecular Biology. 2012;19:728–730. doi: 10.1038/nsmb.2333. [DOI] [PubMed] [Google Scholar]
- Hu M.M., Shu H.B. Cytoplasmic mechanisms of recognition and defense of microbial nucleic acids. Annual Review of Cell and Developmental Biology. 2018;34:357–379. doi: 10.1146/annurev-cellbio-100617-062903. [DOI] [PubMed] [Google Scholar]
- Hu M.M., Yang Q., Xie X.Q., Liao C.Y., Lin H., Liu T.T., Yin L., Shu H.B. Sumoylation promotes the stability of the DNA sensor cGAS and the adaptor STING to regulate the kinetics of response to DNA virus. Immunity. 2016;45:555–569. doi: 10.1016/j.immuni.2016.08.014. [DOI] [PubMed] [Google Scholar]
- Ishak C.A., Classon M., De Carvalho D.D. Deregulation of retroelements as an emerging therapeutic opportunity in cancer. Trends Cancer. 2018;4:583–597. doi: 10.1016/j.trecan.2018.05.008. [DOI] [PubMed] [Google Scholar]
- Ishikawa H., Barber G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674–678. doi: 10.1038/nature07317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa H., Ma Z., Barber G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461:788–792. doi: 10.1038/nature08476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin L., Waterman P.M., Jonscher K.R., Short C.M., Reisdorph N.A., Cambier J.C. MPYS, a novel membrane tetraspanner, is associated with major histocompatibility complex class II and mediates transduction of apoptotic signals. Molecular and Cellular Biology. 2008;28:5014–5026. doi: 10.1128/MCB.00640-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawai T., Takahashi K., Sato S., Coban C., Kumar H., Kato H., Ishii K.J., Takeuchi O., Akira S. IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nature Immunology. 2005;6:981–988. doi: 10.1038/ni1243. [DOI] [PubMed] [Google Scholar]
- Kitajima S., Ivanova E., Guo S., Yoshida R., Campisi M., Sundararaman S.K., Tange S., Mitsuishi Y., Thai T.C., Masuda S., et al. Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer. Cancer Discovery. 2019;9:34–45. doi: 10.1158/2159-8290.CD-18-0689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Konig N., Fiehn C., Wolf C., Schuster M., Cura Costa E., Tungler V., Alvarez H.A., Chara O., Engel K., Goldbach-Mansky R., et al. Familial chilblain lupus due to a gain-of-function mutation in STING. Annals of the Rheumatic Diseases. 2017;76:468–472. doi: 10.1136/annrheumdis-2016-209841. [DOI] [PubMed] [Google Scholar]
- Konno H., Chinn I.K., Hong D., Orange J.S., Lupski J.R., Mendoza A., Pedroza L.A., Barber G.N. Pro-inflammation associated with a gain-of-function mutation (R284S) in the innate immune sensor STING. Cell Reports. 2018;23:1112–1123. doi: 10.1016/j.celrep.2018.03.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Konno H., Konno K., Barber G.N. Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. Cell. 2013;155:688–698. doi: 10.1016/j.cell.2013.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kranzusch P.J., Lee A.S., Berger J.M., Doudna J.A. Structure of human cGAS reveals a conserved family of second-messenger enzymes in innate immunity. Cell Reports. 2013;3:1362–1368. doi: 10.1016/j.celrep.2013.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kujirai T., Zierhut C., Takizawa Y., Kim R., Negishi L., Uruma N., Hirai S., Funabiki H., Kurumizaka H. Structural basis for the inhibition of cGAS by nucleosomes. Science. 2020;370:455–458. doi: 10.1126/science.abd0237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon J., Bakhoum S.F. The cytosolic DNA-sensing cGAS-STING pathway in cancer. Cancer Discovery. 2020;10:26–39. doi: 10.1158/2159-8290.CD-19-0761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lahaye X., Satoh T., Gentili M., Cerboni S., Conrad C., Hurbain I., El Marjou A., Lacabaratz C., Lelievre J.D., Manel N. The capsids of HIV-1 and HIV-2 determine immune detection of the viral cDNA by the innate sensor cGAS in dendritic cells. Immunity. 2013;39:1132–1142. doi: 10.1016/j.immuni.2013.11.002. [DOI] [PubMed] [Google Scholar]
- Lam E., Stein S., Falck-Pedersen E. Adenovirus detection by the cGAS/STING/TBK1 DNA sensing cascade. Journal of Virology. 2014;88:974–981. doi: 10.1128/JVI.02702-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lara P.N., Jr., Douillard J.Y., Nakagawa K., von Pawel J., McKeage M.J., Albert I., Losonczy G., Reck M., Heo D.S., Fan X., et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. Journal of Clinical Oncology. 2011;29:2965–2971. doi: 10.1200/JCO.2011.35.0660. [DOI] [PubMed] [Google Scholar]
- Liang D., Xiao-Feng H., Guan-Jun D., Er-Ling H., Sheng C., Ting-Ting W., Qin-Gang H., Yan-Hong N., Ya-Yi H. Activated STING enhances Tregs infiltration in the HPV-related carcinogenesis of tongue squamous cells via the c-jun/CCL22 signal. Biochimica et Biophysica Acta. 2015;1852:2494–2503. doi: 10.1016/j.bbadis.2015.08.011. [DOI] [PubMed] [Google Scholar]
- Li T., Huang T., Du M., Chen X., Du F., Ren J., Chen Z.J. Phosphorylation and chromatin tethering prevent cGAS activation during mitosis. Science. 2021;371 doi: 10.1126/science.abc5386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z., Liu G., Sun L., Teng Y., Guo X., Jia J., Sha J., Yang X., Chen D., Sun Q. PPM1A regulates antiviral signaling by antagonizing TBK1-mediated STING phosphorylation and aggregation. PLoS Pathogens. 2015;11 doi: 10.1371/journal.ppat.1004783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M., Shu H.B. Dephosphorylation of cGAS by PPP6C impairs its substrate binding activity and innate antiviral response. Protein Cell. 2020;11:584–599. doi: 10.1007/s13238-020-00729-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X., Shu C., Yi G., Chaton C.T., Shelton C.L., Diao J., Zuo X., Kao C.C., Herr A.B., 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]
- Liu Y., Jesus A.A., Marrero B., Yang D., Ramsey S.E., Sanchez G.A.M., Tenbrock K., Wittkowski H., Jones O.Y., Kuehn H.S., et al. Activated STING in a vascular and pulmonary syndrome. New England Journal of Medicine. 2014;371:507–518. doi: 10.1056/NEJMoa1312625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu T.T., Yang Q., Li M., Zhong B., Ran Y., Liu L.L., Yang Y., Wang Y.Y., Shu H.B. LSm14A plays a critical role in antiviral immune responses by regulating MITA level in a cell-specific manner. The Journal of Immunology. 2016;196:5101–5111. doi: 10.4049/jimmunol.1600212. [DOI] [PubMed] [Google Scholar]
- Liu Z.S., Zhang Z.Y., Cai H., Zhao M., Mao J., Dai J., Xia T., Zhang X.M., Li T. RINCK-mediated monoubiquitination of cGAS promotes antiviral innate immune responses. Cell & Bioscience. 2018;8:35. doi: 10.1186/s13578-018-0233-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H., Zhang H., Wu X., Ma D., Wu J., Wang L., Jiang Y., Fei Y., Zhu C., Tan R., et al. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature. 2018;563:131–136. doi: 10.1038/s41586-018-0629-6. [DOI] [PubMed] [Google Scholar]
- Li X.D., Wu J., Gao D., Wang H., Sun L., Chen Z.J. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science. 2013;341:1390–1394. doi: 10.1126/science.1244040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S., Zhang L., Yao Q., Li L., Dong N., Rong J., Gao W., Ding X., Sun L., Chen X., et al. Pathogen blocks host death receptor signalling by arginine GlcNAcylation of death domains. Nature. 2013;501:242–246. doi: 10.1038/nature12436. [DOI] [PubMed] [Google Scholar]
- Lu C., Guan J., Lu S., Jin Q., Rousseau B., Lu T., Stephens D., Zhang H., Zhu J., Yang M., et al. DNA sensing in mismatch repair-deficient tumor cells is essential for anti-tumor immunity. Cancer Cell. 2021;39:96–108. doi: 10.1016/j.ccell.2020.11.006. e106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo W.W., Li S., Li C., Lian H., Yang Q., Zhong B., Shu H.B. iRhom2 is essential for innate immunity to DNA viruses by mediating trafficking and stability of the adaptor STING. Nature Immunology. 2016;17:1057–1066. doi: 10.1038/ni.3510. [DOI] [PubMed] [Google Scholar]
- Mackenzie K.J., Carroll P., Lettice L., Tarnauskaite Z., Reddy K., Dix F., Revuelta A., Abbondati E., Rigby R.E., Rabe B., et al. Ribonuclease H2 mutations induce a cGAS/STING-dependent innate immune response. The EMBO Journal. 2016;35:831–844. doi: 10.15252/embj.201593339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mackenzie K.J., Carroll P., Martin C.A., Murina O., Fluteau A., Simpson D.J., Olova N., Sutcliffe H., Rainger J.K., Leitch A., et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature. 2017;548:461–465. doi: 10.1038/nature23449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma F., Li B., Liu S.Y., Iyer S.S., Yu Y., Wu A., Cheng G. Positive feedback regulation of type I IFN production by the IFN-inducible DNA sensor cGAS. The Journal of Immunology. 2015;194:1545–1554. doi: 10.4049/jimmunol.1402066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Man S.M., Karki R., Malireddi R.K., Neale G., Vogel P., Yamamoto M., Lamkanfi M., Kanneganti T.D. The transcription factor IRF1 and guanylate-binding proteins target activation of the AIM2 inflammasome by Francisella infection. Nature Immunology. 2015;16:467–475. doi: 10.1038/ni.3118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marcus A., Mao A.J., Lensink-Vasan M., Wang L., Vance R.E., Raulet D.H. Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response. Immunity. 2018;49:754–763. doi: 10.1016/j.immuni.2018.09.016. e754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melki I., Rose Y., Uggenti C., Van Eyck L., Fremond M.L., Kitabayashi N., Rice G.I., Jenkinson E.M., Boulai A., Jeremiah N., et al. Disease-associated mutations identify a novel region in human STING necessary for the control of type I interferon signaling. The Journal of Allergy and Clinical Immunology. 2017;140:543–552. doi: 10.1016/j.jaci.2016.10.031. e545. [DOI] [PubMed] [Google Scholar]
- Meylan E., Curran J., Hofmann K., Moradpour D., Binder M., Bartenschlager R., Tschopp J. Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature. 2005;437:1167–1172. doi: 10.1038/nature04193. [DOI] [PubMed] [Google Scholar]
- Michalski S., de Oliveira Mann C.C., Stafford C.A., Witte G., Bartho J., Lammens K., Hornung V., Hopfner K.P. Structural basis for sequestration and autoinhibition of cGAS by chromatin. Nature. 2020;587:678–682. doi: 10.1038/s41586-020-2748-0. [DOI] [PubMed] [Google Scholar]
- Minhas S., Holehouse A.S. Step on the cGAS! Viral inhibition of cGAS phase separation with cytosolic DNA. Molecular Cell. 2021;81:2688–2689. doi: 10.1016/j.molcel.2021.06.007. [DOI] [PubMed] [Google Scholar]
- Morehouse B.R., Govande A.A., Millman A., Keszei A.F.A., Lowey B., Ofir G., Shao S., Sorek R., Kranzusch P.J. STING cyclic dinucleotide sensing originated in bacteria. Nature. 2020;586:429–433. doi: 10.1038/s41586-020-2719-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morita M., Stamp G., Robins P., Dulic A., Rosewell I., Hrivnak G., Daly G., Lindahl T., Barnes D.E. Gene-targeted mice lacking the Trex1 (DNase III) 3'-->5' DNA exonuclease develop inflammatory myocarditis. Molecular and Cellular Biology. 2004;24:6719–6727. doi: 10.1128/MCB.24.15.6719-6727.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moriyama M., Koshiba T., Ichinohe T. Influenza A virus M2 protein triggers mitochondrial DNA-mediated antiviral immune responses. Nature Communications. 2019;10:4624. doi: 10.1038/s41467-019-12632-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Motwani M., Pawaria S., Bernier J., Moses S., Henry K., Fang T., Burkly L., Marshak-Rothstein A., Fitzgerald K.A. Hierarchy of clinical manifestations in SAVI N153S and V154M mouse models. Proceedings of the National Academy of Sciences of the United States of America. 2019;116:7941–7950. doi: 10.1073/pnas.1818281116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Motwani M., Pesiridis S., Fitzgerald K.A. DNA sensing by the cGAS-STING pathway in health and disease. Nature Reviews Genetics. 2019;20:657–674. doi: 10.1038/s41576-019-0151-1. [DOI] [PubMed] [Google Scholar]
- Mukai K., Konno H., Akiba T., Uemura T., Waguri S., Kobayashi T., Barber G.N., Arai H., Taguchi T. Activation of STING requires palmitoylation at the Golgi. Nature Communications. 2016;7 doi: 10.1038/ncomms11932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Munn D.H., Mellor A.L. Ido in the tumor microenvironment: Inflammation, counter-regulation, and tolerance. Trends in Immunology. 2016;37:193–207. doi: 10.1016/j.it.2016.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Munoz J., Rodiere M., Jeremiah N., Rieux-Laucat F., Oojageer A., Rice G.I., Rozenberg F., Crow Y.J., Bessis D. Stimulator of interferon genes-associated vasculopathy with onset in infancy: A mimic of childhood granulomatosis with polyangiitis. JAMA Dermatology. 2015;151:872–877. doi: 10.1001/jamadermatol.2015.0251. [DOI] [PubMed] [Google Scholar]
- Nandakumar R., Tschismarov R., Meissner F., Prabakaran T., Krissanaprasit A., Farahani E., Zhang B.C., Assil S., Martin A., Bertrams W., et al. Intracellular bacteria engage a STING-TBK1-MVB12b pathway to enable paracrine cGAS-STING signalling. Nature Microbiology. 2019;4:701–713. doi: 10.1038/s41564-019-0367-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ni G., Konno H., Barber G.N. Ubiquitination of STING at lysine 224 controls IRF3 activation. Science Immunology. 2017;2 doi: 10.1126/sciimmunol.aah7119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nishimura T., Baba M., Ogawa S., Kojima K., Takita T., Crouch R.J., Yasukawa K. Characterization of six recombinant human RNase H2 bearing Aicardi-Goutieres syndrome causing mutations. Journal of Biochemistry. 2019;166:537–545. doi: 10.1093/jb/mvz073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oda H., Nakagawa K., Abe J., Awaya T., Funabiki M., Hijikata A., Nishikomori R., Funatsuka M., Ohshima Y., Sugawara Y., et al. Aicardi-Goutieres syndrome is caused by IFIH1 mutations. The American Journal of Human Genetics. 2014;95:121–125. doi: 10.1016/j.ajhg.2014.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Oliveira Mann C.C., Orzalli M.H., King D.S., Kagan J.C., Lee A.S.Y., Kranzusch P.J. Modular architecture of the STING C-terminal tail allows interferon and NF-kappaB signaling adaptation. Cell Reports. 2019;27:1165–1175. doi: 10.1016/j.celrep.2019.03.098. e1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orzalli M.H., Broekema N.M., Diner B.A., Hancks D.C., Elde N.C., Cristea I.M., Knipe D.M. cGAS-mediated stabilization of IFI16 promotes innate signaling during herpes simplex virus infection. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E1773–E1781. doi: 10.1073/pnas.1424637112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan B.S., Perera S.A., Piesvaux J.A., Presland J.P., Schroeder G.K., Cumming J.N., Trotter B.W., Altman M.D., Buevich A.V., Cash B., et al. An orally available non-nucleotide STING agonist with antitumor activity. Science. 2020;369 doi: 10.1126/science.aba6098. [DOI] [PubMed] [Google Scholar]
- Pathare G.R., Decout A., Gluck S., Cavadini S., Makasheva K., Hovius R., Kempf G., Weiss J., Kozicka Z., Guey B., et al. Structural mechanism of cGAS inhibition by the nucleosome. Nature. 2020;587:668–672. doi: 10.1038/s41586-020-2750-6. [DOI] [PubMed] [Google Scholar]
- Pokatayev V., Hasin N., Chon H., Cerritelli S.M., Sakhuja K., Ward J.M., Morris H.D., Yan N., Crouch R.J. RNase H2 catalytic core Aicardi-Goutieres syndrome-related mutant invokes cGAS-STING innate immune-sensing pathway in mice. Journal of Experimental Medicine. 2016;213:329–336. doi: 10.1084/jem.20151464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pokatayev V., Yang K., Tu X., Dobbs N., Wu J., Kalb R.G., Yan N. Homeostatic regulation of STING protein at the resting state by stabilizer TOLLIP. Nature Immunology. 2020;21:158–167. doi: 10.1038/s41590-019-0569-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pommier A., Anaparthy N., Memos N., Kelley Z.L., Gouronnec A., Yan R., Auffray C., Albrengues J., Egeblad M., Iacobuzio-Donahue C.A., et al. Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases. Science. 2018;360 doi: 10.1126/science.aao4908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ray Chaudhuri A., Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nature Reviews Molecular Cell Biology. 2017;18:610–621. doi: 10.1038/nrm.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rice G.I., Bond J., Asipu A., Brunette R.L., Manfield I.W., Carr I.M., Fuller J.C., Jackson R.M., Lamb T., Briggs T.A., et al. Mutations involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of the innate immune response. Nature Genetics. 2009;41:829–832. doi: 10.1038/ng.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rice G.I., Kasher P.R., Forte G.M., Mannion N.M., Greenwood S.M., Szynkiewicz M., Dickerson J.E., Bhaskar S.S., Zampini M., Briggs T.A., et al. Mutations in ADAR1 cause Aicardi-Goutieres syndrome associated with a type I interferon signature. Nature Genetics. 2012;44:1243–1248. doi: 10.1038/ng.2414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodero M.P., Tesser A., Bartok E., Rice G.I., Della Mina E., Depp M., Beitz B., Bondet V., Cagnard N., Duffy D., et al. Type I interferon-mediated autoinflammation due to DNase II deficiency. Nature Communications. 2017;8:2176. doi: 10.1038/s41467-017-01932-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saldanha R.G., Balka K.R., Davidson S., Wainstein B.K., Wong M., Macintosh R., Loo C.K.C., Weber M.A., Kamath V., Circa, et al. A mutation outside the dimerization domain causing atypical STING-associated vasculopathy with onset in infancy. Frontiers in Immunology. 2018;9:1535. doi: 10.3389/fimmu.2018.01535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sansone P., Savini C., Kurelac I., Chang Q., Amato L.B., Strillacci A., Stepanova A., Iommarini L., Mastroleo C., Daly L., et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proceedings of the National Academy of Sciences of the United States of America. 2017;114:E9066–E9075. doi: 10.1073/pnas.1704862114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoggins J.W., MacDuff D.A., Imanaka N., Gainey M.D., Shrestha B., Eitson J.L., Mar K.B., Richardson R.B., Ratushny A.V., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo J., Kang J.A., Suh D.I., Park E.B., Lee C.R., Choi S.A., Kim S.Y., Kim Y., Park S.H., Ye M., et al. Tofacitinib relieves symptoms of stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy caused by 2 de novo variants in TMEM173. The Journal of Allergy and Clinical Immunology. 2017;139:1396–1399. doi: 10.1016/j.jaci.2016.10.030. e1312. [DOI] [PubMed] [Google Scholar]
- Seo G.J., Kim C., Shin W.J., Sklan E.H., Eoh H., Jung J.U. TRIM56-mediated monoubiquitination of cGAS for cytosolic DNA sensing. Nature Communications. 2018;9:613. doi: 10.1038/s41467-018-02936-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo G.J., Yang A., Tan B., Kim S., Liang Q., Choi Y., Yuan W., Feng P., Park H.S., Jung J.U. Akt kinase-mediated checkpoint of cGAS DNA sensing pathway. Cell Reports. 2015;13:440–449. doi: 10.1016/j.celrep.2015.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seth R.B., Sun L., Ea C.K., Chen Z.J. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell. 2005;122:669–682. doi: 10.1016/j.cell.2005.08.012. [DOI] [PubMed] [Google Scholar]
- 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. 2019;567:389–393. doi: 10.1038/s41586-019-0998-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shmuel-Galia L., Humphries F., Lei X., Ceglia S., Wilson R., Jiang Z., Ketelut-Carneiro N., Foley S.E., Pechhold S., Houghton J., et al. Dysbiosis exacerbates colitis by promoting ubiquitination and accumulation of the innate immune adaptor STING in myeloid cells. Immunity. 2021;54:1137–1153. doi: 10.1016/j.immuni.2021.05.008. e1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson S.R., Rego S.L., Harvey S.E., Liu M., Hemphill W.O., Venkatadri R., Sharma R., Grayson J.M., Perrino F.W. T cells produce IFN-alpha in the TREX1 D18N model of lupus-like autoimmunity. The Journal of Immunology. 2020;204:348–359. doi: 10.4049/jimmunol.1900220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sliter D.A., Martinez J., Hao L., Chen X., Sun N., Fischer T.D., Burman J.L., Li Y., Zhang Z., Narendra D.P., et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature. 2018;561:258–262. doi: 10.1038/s41586-018-0448-9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Song Z.M., Lin H., Yi X.M., Guo W., Hu M.M., Shu H.B. KAT5 acetylates cGAS to promote innate immune response to DNA virus. Proceedings of the National Academy of Sciences of the United States of America. 2020;117:21568–21575. doi: 10.1073/pnas.1922330117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srikanth S., Woo J.S., Wu B., El-Sherbiny Y.M., Leung J., Chupradit K., Rice L., Seo G.J., Calmettes G., Ramakrishna C., et al. The Ca(2+) sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum. Nature Immunology. 2019;20:152–162. doi: 10.1038/s41590-018-0287-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stetson D.B., Ko J.S., Heidmann T., Medzhitov R. Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell. 2008;134:587–598. doi: 10.1016/j.cell.2008.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sumner R.P., Harrison L., Touizer E., Peacock T.P., Spencer M., Zuliani-Alvarez L., Towers G.J. Disrupting HIV-1 capsid formation causes cGAS sensing of viral DNA. The EMBO Journal. 2020;39 doi: 10.15252/embj.2019103958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun W., Li Y., Chen L., Chen H., You F., Zhou X., Zhou Y., Zhai Z., Chen D., Jiang Z. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:8653–8658. doi: 10.1073/pnas.0900850106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X., Liu T., Zhao J., Xia H., Xie J., Guo Y., Zhong L., Li M., Yang Q., Peng C., et al. DNA-PK deficiency potentiates cGAS-mediated antiviral innate immunity. Nature Communications. 2020;11:6182. doi: 10.1038/s41467-020-19941-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Q., Sun L., Liu H.H., Chen X., Seth R.B., Forman J., Chen Z.J. The specific and essential role of MAVS in antiviral innate immune responses. Immunity. 2006;24:633–642. doi: 10.1016/j.immuni.2006.04.004. [DOI] [PubMed] [Google Scholar]
- 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. 2013;339:786–791. doi: 10.1126/science.1232458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun H., Zhang Q., Jing Y.Y., Zhang M., Wang H.Y., Cai Z., Liuyu T., Zhang Z.D., Xiong T.C., Wu Y., et al. USP13 negatively regulates antiviral responses by deubiquitinating STING. Nature Communications. 2017;8 doi: 10.1038/ncomms15534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takahashi M., Lio C.J., Campeau A., Steger M., Ay F., Mann M., Gonzalez D.J., Jain M., Sharma S. The tumor suppressor kinase DAPK3 drives tumor-intrinsic immunity through the STING-IFN-beta pathway. Nature Immunology. 2021;22:485–496. doi: 10.1038/s41590-021-00896-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takaoka A., Wang Z., Choi M.K., Yanai H., Negishi H., Ban T., Lu Y., Miyagishi M., Kodama T., Honda K., et al. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature. 2007;448:501–505. doi: 10.1038/nature06013. [DOI] [PubMed] [Google Scholar]
- Tan A.S., Baty J.W., Dong L.F., Bezawork-Geleta A., Endaya B., Goodwin J., Bajzikova M., Kovarova J., Peterka M., Yan B., et al. Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA. Cell Metabolism. 2015;21:81–94. doi: 10.1016/j.cmet.2014.12.003. [DOI] [PubMed] [Google Scholar]
- Tang C.H., Zundell J.A., Ranatunga S., Lin C., Nefedova Y., Del Valle J.R., Hu C.C. Agonist-mediated activation of STING induces apoptosis in malignant B cells. Cancer Research. 2016;76:2137–2152. doi: 10.1158/0008-5472.CAN-15-1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan X., Sun L., Chen J., Chen Z.J. Detection of microbial infections through innate immune sensing of nucleic acids. Annual Review of Microbiology. 2018;72:447–478. doi: 10.1146/annurev-micro-102215-095605. [DOI] [PubMed] [Google Scholar]
- Terai H., Kitajima S., Potter D.S., Matsui Y., Quiceno L.G., Chen T., Kim T.J., Rusan M., Thai T.C., Piccioni F., et al. ER stress signaling promotes the survival of cancer "persister cells" tolerant to EGFR tyrosine kinase inhibitors. Cancer Research. 2018;78:1044–1057. doi: 10.1158/0008-5472.CAN-17-1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomsen M.K., Nandakumar R., Stadler D., Malo A., Valls R.M., Wang F., Reinert L.S., Dagnaes-Hansen F., Hollensen A.K., Mikkelsen J.G., et al. Lack of immunological DNA sensing in hepatocytes facilitates hepatitis B virus infection. Hepatology. 2016;64:746–759. doi: 10.1002/hep.28685. [DOI] [PubMed] [Google Scholar]
- Toso A., Revandkar A., Di Mitri D., Guccini I., Proietti M., Sarti M., Pinton S., Zhang J., Kalathur M., Civenni G., et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Reports. 2014;9:75–89. doi: 10.1016/j.celrep.2014.08.044. [DOI] [PubMed] [Google Scholar]
- Tsuchida T., Zou J., Saitoh T., Kumar H., Abe T., Matsuura Y., Kawai T., Akira S. The ubiquitin ligase TRIM56 regulates innate immune responses to intracellular double-stranded DNA. Immunity. 2010;33:765–776. doi: 10.1016/j.immuni.2010.10.013. [DOI] [PubMed] [Google Scholar]
- Unterholzner L., Keating S.E., Baran M., Horan K.A., Jensen S.B., Sharma S., Sirois C.M., Jin T., Latz E., Xiao T.S., et al. IFI16 is an innate immune sensor for intracellular DNA. Nature Immunology. 2010;11:997–1004. doi: 10.1038/ni.1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C., Guan Y., Lv M., Zhang R., Guo Z., Wei X., Du X., Yang J., Li T., Wan Y., et al. Manganese increases the sensitivity of the cGAS-STING pathway for double-stranded DNA and is required for the host defense against DNA viruses. Immunity. 2018;48:675–687. doi: 10.1016/j.immuni.2018.03.017. e677. [DOI] [PubMed] [Google Scholar]
- Wang Y., Lian Q., Yang B., Yan S., Zhou H., He L., Lin G., Lian Z., Jiang Z., Sun B. TRIM30alpha is a negative-feedback regulator of the intracellular DNA and DNA virus-triggered response by targeting STING. PLoS Pathogens. 2015;11 doi: 10.1371/journal.ppat.1005012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q., Liu X., Cui Y., Tang Y., Chen W., Li S., Yu H., Pan Y., Wang C. The E3 ubiquitin ligase AMFR and INSIG1 bridge the activation of TBK1 kinase by modifying the adaptor STING. Immunity. 2014;41:919–933. doi: 10.1016/j.immuni.2014.11.011. [DOI] [PubMed] [Google Scholar]
- Wang Y., Wang M., Djekidel M.N., Chen H., Liu D., Alt F.W., Zhang Y. eccDNAs are apoptotic products with high innate immunostimulatory activity. Nature. 2021;599:308–314. doi: 10.1038/s41586-021-04009-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C., Wang X., Veleeparambil M., Kessler P.M., Willard B., Chattopadhyay S., Sen G.C. EGFR-mediated tyrosine phosphorylation of STING determines its trafficking route and cellular innate immunity functions. The EMBO Journal. 2020;39 doi: 10.15252/embj.2019104106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warner J.D., Irizarry-Caro R.A., Bennion B.G., Ai T.L., Smith A.M., Miner C.A., Sakai T., Gonugunta V.K., Wu J., Platt D.J., et al. STING-associated vasculopathy develops independently of IRF3 in mice. Journal of Experimental Medicine. 2017;214:3279–3292. doi: 10.1084/jem.20171351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whiteley A.T., Eaglesham J.B., de Oliveira Mann C.C., Morehouse B.R., Lowey B., Nieminen E.A., Danilchanka O., King D.S., Lee A.S.Y., Mekalanos J.J., et al. Bacterial cGAS-like enzymes synthesize diverse nucleotide signals. Nature. 2019;567:194–199. doi: 10.1038/s41586-019-0953-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White M.J., McArthur K., Metcalf D., Lane R.M., Cambier J.C., Herold M.J., van Delft M.F., Bedoui S., Lessene G., Ritchie M.E., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woo S.R., Fuertes M.B., Corrales L., Spranger S., Furdyna M.J., Leung M.Y., Duggan R., Wang Y., Barber G.N., Fitzgerald K.A., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J., Chen Y.J., Dobbs N., Sakai T., Liou J., Miner J.J., Yan N. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. Journal of Experimental Medicine. 2019;216:867–883. doi: 10.1084/jem.20182192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu M.Z., Cheng W.C., Chen S.F., Nieh S., O'Connor C., Liu C.L., Tsai W.W., Wu C.J., Martin L., Lin Y.S., et al. miR-25/93 mediates hypoxia-induced immunosuppression by repressing cGAS. Nature Cell Biology. 2017;19:1286–1296. doi: 10.1038/ncb3615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J., Dobbs N., Yang K., Yan N. Interferon-independent activities of mammalian STING mediate antiviral response and tumor immune evasion. Immunity. 2020;53:115–126. doi: 10.1016/j.immuni.2020.06.009. e115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu X., Wu F.H., Wang X., Wang L., Siedow J.N., Zhang W., Pei Z.M. Molecular evolutionary and structural analysis of the cytosolic DNA sensor cGAS and STING. Nucleic Acids Research. 2014;42:8243–8257. doi: 10.1093/nar/gku569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia T., Konno H., Ahn J., Barber G.N. Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis. Cell Reports. 2016;14:282–297. doi: 10.1016/j.celrep.2015.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia T., Konno H., Barber G.N. Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis. Cancer Research. 2016;76:6747–6759. doi: 10.1158/0008-5472.CAN-16-1404. [DOI] [PubMed] [Google Scholar]
- Xiao N., Wei J., Xu S., Du H., Huang M., Zhang S., Ye W., Sun L., Chen Q. cGAS activation causes lupus-like autoimmune disorders in a TREX1 mutant mouse model. Journal of Autoimmunity. 2019;100:84–94. doi: 10.1016/j.jaut.2019.03.001. [DOI] [PubMed] [Google Scholar]
- Xia P., Wang S., Ye B., Du Y., Li C., Xiong Z., Qu Y., Fan Z. A circular RNA protects dormant hematopoietic stem cells from DNA sensor cGAS-mediated exhaustion. Immunity. 2018;48:688–701. doi: 10.1016/j.immuni.2018.03.016. e687. [DOI] [PubMed] [Google Scholar]
- Xia P., Ye B., Wang S., Zhu X., Du Y., Xiong Z., Tian Y., Fan Z. Glutamylation of the DNA sensor cGAS regulates its binding and synthase activity in antiviral immunity. Nature Immunology. 2016;17:369–378. doi: 10.1038/ni.3356. [DOI] [PubMed] [Google Scholar]
- Xia T., Yi X.M., Wu X., Shang J., Shu H.B. PTPN1/2-mediated dephosphorylation of MITA/STING promotes its 20S proteasomal degradation and attenuates innate antiviral response. Proceedings of the National Academy of Sciences of the United States of America. 2019;116:20063–20069. doi: 10.1073/pnas.1906431116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie W., Lama L., Adura C., Tomita D., Glickman J.F., Tuschl T., Patel D.J. Human cGAS catalytic domain has an additional DNA-binding interface that enhances enzymatic activity and liquid-phase condensation. Proceedings of the National Academy of Sciences of the United States of America. 2019;116:11946–11955. doi: 10.1073/pnas.1905013116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing J., Zhang A., Zhang H., Wang J., Li X.C., Zeng M.S., Zhang Z. TRIM29 promotes DNA virus infections by inhibiting innate immune response. Nature Communications. 2017;8:945. doi: 10.1038/s41467-017-00101-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu G., Liu C., Zhou S., Li Q., Feng Y., Sun P., Feng H., Gao Y., Zhu J., Luo X., et al. Viral tegument proteins restrict cGAS-DNA phase separation to mediate immune evasion. Molecular Cell. 2021;81:2823–2837. doi: 10.1016/j.molcel.2021.05.002. e2829. [DOI] [PubMed] [Google Scholar]
- Xu L.G., Wang Y.Y., Han K.J., Li L.Y., Zhai Z., Shu H.B. VISA is an adapter protein required for virus-triggered IFN-beta signaling. Molecular Cell. 2005;19:727–740. doi: 10.1016/j.molcel.2005.08.014. [DOI] [PubMed] [Google Scholar]
- Xu J.Y., Zhang C., Wang X., Zhai L., Ma Y., Mao Y., Qian K., Sun C., Liu Z., Jiang S., et al. Integrative proteomic characterization of human lung adenocarcinoma. Cell. 2020;182:245–261. doi: 10.1016/j.cell.2020.05.043. e217. [DOI] [PubMed] [Google Scholar]
- Yamashiro L.H., Wilson S.C., Morrison H.M., Karalis V., Chung J.J., Chen K.J., Bateup H.S., Szpara M.L., Lee A.Y., Cox J.S., et al. Interferon-independent STING signaling promotes resistance to HSV-1 in vivo. Nature Communications. 2020;11:3382. doi: 10.1038/s41467-020-17156-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H., Lee W.S., Kong S.J., Kim C.G., Kim J.H., Chang S.K., Kim S., Kim G., Chon H.J., Kim C. STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade. Journal of Clinical Investigation. 2019;129:4350–4364. doi: 10.1172/JCI125413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Q., Liu T.T., Lin H., Zhang M., Wei J., Luo W.W., Hu Y.H., Zhong B., Hu M.M., Shu H.B. TRIM32-TAX1BP1-dependent selective autophagic degradation of TRIF negatively regulates TLR3/4-mediated innate immune responses. PLoS Pathogens. 2017;13 doi: 10.1371/journal.ppat.1006600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H., Wang H., Ren J., Chen Q., Chen Z.J. cGAS is essential for cellular senescence. Proceedings of the National Academy of Sciences of the United States of America. 2017;114:E4612–E4620. doi: 10.1073/pnas.1705499114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye L., Zhang Q., Liuyu T., Xu Z., Zhang M.X., Luo M.H., Zeng W.B., Zhu Q., Lin D., Zhong B. USP49 negatively regulates cellular antiviral responses via deconjugating K63-linked ubiquitination of MITA. PLoS Pathogens. 2019;15 doi: 10.1371/journal.ppat.1007680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin Q., Tian Y., Kabaleeswaran V., Jiang X., Tu D., Eck M.J., Chen Z.J., Wu H. Cyclic di-GMP sensing via the innate immune signaling protein STING. Molecular Cell. 2012;46:735–745. doi: 10.1016/j.molcel.2012.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yum S., Li M., Fang Y., Chen Z.J. TBK1 recruitment to STING activates both IRF3 and NF-kappaB that mediate immune defense against tumors and viral infections. Proceedings of the National Academy of Sciences of the United States of America. 2021;118 doi: 10.1073/pnas.2100225118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu X., Zhang L., Shen J., Zhai Y., Jiang Q., Yi M., Deng X., Ruan Z., Fang R., Chen Z., et al. The STING phase-separator suppresses innate immune signalling. Nature Cell Biology. 2021;23:330–340. doi: 10.1038/s41556-021-00659-0. [DOI] [PubMed] [Google Scholar]
- Zhang X., Bai X.C., Chen Z.J. Structures and mechanisms in the cGAS-STING innate immunity pathway. Immunity. 2020;53:43–53. doi: 10.1016/j.immuni.2020.05.013. [DOI] [PubMed] [Google Scholar]
- Zhang J., Hu M.M., Wang Y.Y., Shu H.B. TRIM32 protein modulates type I interferon induction and cellular antiviral response by targeting MITA/STING protein for K63-linked ubiquitination. Journal of Biological Chemistry. 2012;287:28646–28655. doi: 10.1074/jbc.M112.362608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B.C., Nandakumar R., Reinert L.S., Huang J., Laustsen A., Gao Z.L., Sun C.L., Jensen S.B., Troldborg A., Assil S., et al. Author correction: STEEP mediates STING ER exit and activation of signaling. Nature Immunology. 2020;21:1468–1469. doi: 10.1038/s41590-020-0803-5. [DOI] [PubMed] [Google Scholar]
- Zhang C., Shang G., Gui X., Zhang X., Bai X.C., Chen Z.J. Structural basis of STING binding with and phosphorylation by TBK1. Nature. 2019;567:394–398. doi: 10.1038/s41586-019-1000-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Q., Tang Z., An R., Ye L., Zhong B. USP29 maintains the stability of cGAS and promotes cellular antiviral responses and autoimmunity. Cell Research. 2020;30:914–927. doi: 10.1038/s41422-020-0341-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L., Wei N., Cui Y., Hong Z., Liu X., Wang Q., Li S., Liu H., Yu H., Cai Y., et al. The deubiquitinase CYLD is a specific checkpoint of the STING antiviral signaling pathway. PLoS Pathogens. 2018;14 doi: 10.1371/journal.ppat.1007435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X., Wu J., Du F., Xu H., Sun L., Chen Z., Brautigam C.A., Zhang X., Chen Z.J. The cytosolic DNA sensor cGAS forms an oligomeric complex with DNA and undergoes switch-like conformational changes in the activation loop. Cell Reports. 2014;6:421–430. doi: 10.1016/j.celrep.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z.D., Xiong T.C., Yao S.Q., Wei M.C., Chen M., Lin D., Zhong B. RNF115 plays dual roles in innate antiviral responses by catalyzing distinct ubiquitination of MAVS and MITA. Nature Communications. 2020;11:5536. doi: 10.1038/s41467-020-19318-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y., Yeruva L., Marinov A., Prantner D., Wyrick P.B., Lupashin V., Nagarajan U.M. The DNA sensor, cyclic GMP-AMP synthase, is essential for induction of IFN-beta during Chlamydia trachomatis infection. The Journal of Immunology. 2014;193:2394–2404. doi: 10.4049/jimmunol.1302718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z., Yuan B., Bao M., Lu N., Kim T., Liu Y.J. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nature Immunology. 2011;12:959–965. doi: 10.1038/ni.2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M., Zhang M.X., Zhang Q., Zhu G.F., Yuan L., Zhang D.E., Zhu Q., Yao J., Shu H.B., Zhong B. USP18 recruits USP20 to promote innate antiviral response through deubiquitinating STING/MITA. Cell Research. 2016;26:1302–1319. doi: 10.1038/cr.2016.125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao L., Ching L.M., Kestell P., Baguley B.C. The antitumour activity of 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in TNF receptor-1 knockout mice. British Journal of Cancer. 2002;87:465–470. doi: 10.1038/sj.bjc.6600479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao B., Du F., Xu P., Shu C., Sankaran B., Bell S.L., Liu M., Lei Y., Gao X., Fu X., et al. A conserved PLPLRT/SD motif of STING mediates the recruitment and activation of TBK1. Nature. 2019;569:718–722. doi: 10.1038/s41586-019-1228-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao Z., Ma Z., Wang B., Guan Y., Su X.D., Jiang Z. Mn(2+) directly activates cGAS and structural analysis suggests Mn(2+) induces a noncanonical catalytic synthesis of 2'3'-cGAMP. Cell Reports. 2020;32 doi: 10.1016/j.celrep.2020.108053. [DOI] [PubMed] [Google Scholar]
- Zhao B., Xu P., Rowlett C.M., Jing T., Shinde O., Lei Y., West A.P., Liu W.R., Li P. The molecular basis of tight nuclear tethering and inactivation of cGAS. Nature. 2020;587:673–677. doi: 10.1038/s41586-020-2749-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhong L., Hu M.M., Bian L.J., Liu Y., Chen Q., Shu H.B. Phosphorylation of cGAS by CDK1 impairs self-DNA sensing in mitosis. Cell Discovery. 2020;6:26. doi: 10.1038/s41421-020-0162-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhong B., Yang Y., Li S., Wang Y.Y., Li Y., Diao F., Lei C., He X., Zhang L., Tien P., et al. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity. 2008;29:538–550. doi: 10.1016/j.immuni.2008.09.003. [DOI] [PubMed] [Google Scholar]
- Zhong B., Zhang L., Lei C., Li Y., Mao A.P., Yang Y., Wang Y.Y., Zhang X.L., Shu H.B. The ubiquitin ligase RNF5 regulates antiviral responses by mediating degradation of the adaptor protein MITA. Immunity. 2009;30:397–407. doi: 10.1016/j.immuni.2009.01.008. [DOI] [PubMed] [Google Scholar]
- Zhou W., Whiteley A.T., de Oliveira Mann C.C., Morehouse B.R., Nowak R.P., Fischer E.S., Gray N.S., Mekalanos J.J., Kranzusch P.J. Structure of the human cGAS-DNA complex reveals enhanced control of immune surveillance. Cell. 2018;174:300–311. doi: 10.1016/j.cell.2018.06.026. e311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zierhut C., Funabiki H. Regulation and consequences of cGAS activation by self-DNA. Trends in Cell Biology. 2020;30:594–605. doi: 10.1016/j.tcb.2020.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]



