Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jan 30.
Published in final edited form as: Immunity. 2023 Sep 1;56(9):1991–2005.e9. doi: 10.1016/j.immuni.2023.08.006

The virus-induced cyclic dinucleotide 2′3′-c-di-GMP mediates STING-dependent antiviral immunity in Drosophila

Hua Cai 1,6,*, Lihua Li 1,6, Kailey M Slavik 2,3,6, Jingxian Huang 1,6, Ting Yin 1, Xianlong Ai 1, Léna Hédelin 4, Gabrielle Haas 4, Zhangmin Xiang 5, Yunyun Yang 5, Xiaoyan Li 1, Yuqiang Chen 1, Ziming Wei 1, Huimin Deng 1, Di Chen 1, Renjie Jiao 1, Nelson Martins 4,7, Carine Meignin 4, Philip J Kranzusch 2,3,*, Jean-Luc Imler 1,4
PMCID: PMC11781366  NIHMSID: NIHMS2046904  PMID: 37659413

Abstract

In mammals, the enzyme cGAS senses the presence of cytosolic DNA and synthesizes the cyclic dinucleotide (CDN) 2′3′-cGAMP, which triggers STING-dependent immunity. In Drosophila melanogaster, two cGAS-like receptors (cGLRs) produce 3′2′-cGAMP and 2′3′-cGAMP to activate STING. We explored CDN-mediated immunity in 14 Drosophila species covering 50 million years of evolution and found that 2′3′-cGAMP and 3′2′-cGAMP failed to control infection by Drosophila C virus in D. serrata and two other species. We discovered diverse CDNs produced in a cGLR-dependent manner in response to viral infection in D. melanogaster, including 2′3′-c-di-GMP. This CDN was a more potent STING agonist than cGAMP in D. melanogaster and it also activated a strong antiviral transcriptional response in D. serrata. Our results shed light on the evolution of cGLRs in flies and provide a basis for the understanding of the function and regulation of this emerging family of pattern recognition receptors in animal innate immunity.

Introduction

Innate immunity is the first line of host-defense against infections. It involves a set of pattern recognition receptors (PRRs), which sense conserved microbial products and activate signaling to mount an efficient antimicrobial response. The model organism of the fruit fly Drosophila melanogaster has played an important role in the identification of the first characterized family of PRRs, the Toll-like receptors13. In Drosophila, however, Toll functions as a receptor for the cytokine Spaetzle, rather than as a PRR4. Sensing of bacterial and fungal infection occurs upstream of Toll, and is mediated by members of two families of PRRs, peptidoglycan recognition proteins (PGRPs) and Gram-negative binding proteins (GNBPs), which detect lysine-type peptidoglycan or β-glucans57. Other members of the PGRP family sense DAP-type peptidoglycan to activate the IMD pathway811.

Viral infections have long been thought to be controlled solely by RNA interference in insects, even though induction of gene expression in virus-infected flies has been noted (reviewed in12,13). A transcriptional response to viral infection involving the evolutionarily conserved protein STING has indeed been recently characterized in flies and silkworms1416. In mammals, STING is activated upon binding a second messenger, the cyclic dinucleotide (CDN) 2′3′-cGAMP, and triggers the transcription factors IRF3 and NF-κB to induce expression of genes encoding interferons and other antiviral molecules (reviewed in17). 2′3′-cGAMP is produced by the enzyme cyclic GMP-AMP synthase (cGAS), which acts as a PRR sensing the presence of double stranded (ds)DNA in the cytosol18. In flies and silkworms, STING regulates Relish, a member of the NF-κB family, to induce expression of genes that function in concert to control of viral infections14,15,19. The pathway is activated by two cGAS-like receptors (cGLRs), which, together with cGAS, define a previously unrecognized family of PRRs in Drosophila20,21.

cGAS-STING signaling originated in bacteria, where it plays a critical role in the control of phage infections (reviewed in22). A large collection of cGAS/DncV-like nucleotidyltransferases (CD-NTases) are activated upon phage infection and catalyze the production of cyclic di- or trinucleotides as part of a cyclic oligonucleotide-based antiphage signaling system (CBASS)23,24. These phage-induced cyclic oligonucleotides bind to a variety of effector proteins, including homologs of STING, activating them to oppose phage replication, either by triggering cell death or by degrading phage nucleic acids25,26. Thus, it appears that animals acquired the components of the cGAS-STING pathway from prokaryotes, a hypothesis supported by the recent demonstration that STING signaling is activated and participates in the response to infection in invertebrates beyond insects, e.g., the sea anemone Nematostella vectensis27, and even in the choanoflagellate Monosiga brevicollis, a unicellular close relative of metazoans28.

In D. melanogaster, two cGLRs have been shown to produce CDNs that activate STING signaling and their initial characterization revealed differences compared to mammalian cGAS20,21. First, the activity of cGLR1 can be triggered in vitro or in transfected cells by dsRNA rather than dsDNA. The ligand activating cGLR2 is still unknown, although mutation of conserved residues critical for DNA or RNA binding in cGAS and oligoadenylate synthetase decrease the activity of cGLR2, suggesting allosteric activation by a nucleic acid20,29. Second, although cGLR2 also produces 2′3′-cGAMP, both cGLRs were found to produce another potent STING agonist, the CDN 3′2′-cGAMP. Third, Drosophila genomes encode a set of putative cGLRs, with between 2 and 7 cGAS homologues per species. Of note, cGLR1 and cGLR2 have so far mostly been molecularly characterized in vitro with recombinant proteins or using transfected cells. Here, we exploit the biodiversity of the Drosophila genus to gain insight on the contribution of 2′3′-cGAMP and 3′2′-cGAMP in the control of viral infection and on the function of cGLRs in vivo. Our results reveal a rapidly evolving family of PRRs which participate in host-defense through the production of diverse nucleotide signals.

Results

3′2′-cGAMP induces antiviral immunity in most, but not all, Drosophila species

To investigate the role of 3′2′-cGAMP signaling in antiviral immunity in the Drosophila genus, we selected 14 species covering 50 million years of evolution (Figure 1A). We injected them with 3′2′-cGAMP and 2′3′-cGAMP, the ancestral signaling molecule in metazoans, as well as 2′3′-c-di-AMP, identified as a minor product of cGLR120,21. The flies were subsequently challenged with Drosophila C virus (DCV, Dicistroviridae family), a natural Drosophila pathogen that is sensitive to CDN signaling in D. melanogaster, and viral RNA load was monitored two- and three-days post-infection (Figure 1B). As with 3′2′-cGAMP and 2′3′-cGAMP, injection of 2′3′-c-di-AMP resulted in significant reduction of DCV RNA in D. melanogaster, revealing that the production of this CDN in vitro by recombinant cGLR1 might be physiologically relevant (Figure 1C)20,21. Injection of the three CDNs also resulted in decreased viral RNA loads in 8 other fly species (Figure 1B,D). However, neither 2′3′-cGAMP nor 2′3′-c-di-AMP reduced DCV load in D. sechellia, D. serrata, D. kikkawai, D. pseudoobscura and D. mojavensis (Figure 1B,E,F). The injection of 3′2′-cGAMP led to reduced DCV replication in 11 of the 14 species, including D. melanogaster, and in most cases the decrease in viral RNAs was stronger than in flies injected with 2′3′-c-di-AMP or 2′3′-cGAMP. Yet, neither 3′2′-cGAMP nor the other tested CDNs affected DCV RNA levels in D. sechellia, D. serrata or D. mojavensis (Figure 1B,F). Overall, these results confirm the importance of 3′2′-cGAMP in antiviral immunity in Drosophila and raise the possibility that other types of CDNs mediate antiviral protection in some Drosophila species that respond poorly to 3′2′-cGAMP, 2′3′-cGAMP and 2′3′-c-di-AMP.

Figure 1: 3′2′-cGAMP induces antiviral immunity in most, but not all, Drosophila species.

Figure 1:

A. Phylogeny of the 14 Drosophila species used. Drosophila and Sophophora subgenus are indicated. B. Summary of the antiviral effect of the indicated CDNs in the Drosophila species. Heatmap showing the log2 fold change of DCV RNA load at two- and three-days post-infection of male flies that had been pre-injected with the indicated CDNs or Tris. Significant changes (adjusted p value ≤0.05; pairwise permutation test with FDR method) are highlighted with dots. C-F. Relative DCV RNA loads at two or three days post DCV infection in flies from the indicated species. Data are from at least three independent experiments, each performed in biological triplicates and shown with boxplot with scatter plot. Analysis using pairwise permutation test with FDR method upon CDN- compared to Tris-injection is shown (ns= non significant, *<0.05, **<0.01 and ***<0.001).

cGLR1 and cGLR2 produce several CDNs in vivo

To detect CDNs produced by D. melanogaster cGLR1 and cGLR2 in vivo, we took advantage of transgenic fly lines. We previously reported that the STING pathway is activated in the absence of viral infection in flies expressing wild-type cGLR1 or 2, but not catalytically inactive mutant versions, suggesting that CDNs are produced20. To monitor the presence of CDNs, we designed a bioassay based on the injection of hemolymph from cGLR-overexpressing flies into naïve flies, followed by analysis of expression of STING-regulated genes (srg)19 (Figure S1A). Injection of hemolymph extracted from flies ectopically expressing cGLR1, but not the inactive cGLR1AFA mutant, into wild-type flies resulted in significant upregulation of srg2 and srg3 expression. Similar trends for increased expression were observed for the genes Drosophila STING (dSTING) itself and srg1 (Figure S1B-E). Comparable results were observed for the transfer of hemolymph from flies ectopically expressing cGLR2. No such induction was observed when the hemolymph was transferred into dSTING mutant flies (Figure S1BE). Overall, these results indicate that a STING agonist is produced and circulating in the cGLR-overexpressing flies.

We next developed a liquid chromatography tandem mass spectrometry (LC MS/MS) method using high resolution mass spectrometry for identification and quantification of CDNs in the hemolymph and whole fly lysates20,30. In contrast to collected hemolymph, fly extracts are complex mixtures, which required optimization of the CDN extraction, purification and solid phase separation procedures. The CDNs were first identified by targeted mass analysis for exact masses and formulae for all possible CDNs in extracts prepared from lysates of flies ectopically expressing catalytically active cGLR2. Mass-to-charge ratio targeted to cGAMP, but also two other CDNs, namely c-di-AMP and c-di-GMP, were detected. These CDNs were not detected in flies expressing the catalytically inactive version, although a minor peak for c-di-AMP was noted in two out of six samples (Figure S2A, B, C). The progeny ions of the selected parent ion for each of the three CDNs were detected by tandem MS, which confirmed the identification of cGAMP, c-di-AMP and c-di-GMP (Figure S2D,E,F). Of note, the ratio of progeny ion for c-di-GMP in fly extracts resembled that of 2'3'-c-di-GMP rather than 3'3'-c-di-GMP (Figure S2F). To determine the isomers of the CDNs produced by cGLR2 in a less complex system, we analyzed extracts from human HEK293T cells transfected with a cGLR2-expressing vector20 and observed the production of cGAMP, c-di-GMP and, albeit in lower quantities, c-di-AMP (Figure S3A-D). Spiking of chemically synthesized isomers of cGAMP, c-di-AMP and c-di-GMP confirmed that ectopically expressed cGLR2 induced the production of 3'2'-cGAMP, 2'3'-c-GAMP, 2'3'-c-di-GMP and low amount of 2'3'-c-di-AMP (Figure S3E-G).

Next, we established LC-MS/MS using a triple quadrupole mass spectrometer to quantify the two cGAMP isomers (3'2'-cGAMP and 2'3'-c-GAMP), 2'3'-c-di-GMP and 2'3'-c-di-AMP, both in the hemolymph and in fly extracts. The two cGAMP isomers were quantified by two quantifier ions with m/z=136.0 and 524.0, which produced the strongest signal in 2'3'-cGAMP and 3'2'-cGAMP, respectively (Figure S4). The ions with m/z=524.0 and 540.0 were used for the quantification of 2'3'-c-di-AMP and 2'3'-c-di-GMP, respectively (Figure S4). All the CDNs, including both cGAMP isomers, 2'3'-c-di-GMP and 2'3'-c-di-AMP, were detected in the hemolymph of flies ectopically expressing cGLR2 (Figure 2A). In contrast, only 2'3'-cGAMP and 3'2'-cGAMP were detected in the hemolymph of flies ectopically expressing cGLR1. None of these CDNs were present in the hemolymph of flies expressing the catalytically inactive version of cGLR1 or cGLR2 (Figure 2A). 2'3'-c-di-GMP, in addition to the cGAMP isomers and 2'3'-c-di-AMP, was also present in extracts prepared from lysates of flies ectopically expressing catalytically active cGLR2 or from human HEK293T cells transfected with a cGLR2-expressing vector (Figure 2B,C). Note that 2'3'-c-di-AMP, together with some 3'3'-c-di-AMP, was also detected in control flies expressing the inactive enzyme or GFP. In summary, we have established a method to detect CDNs in vivo in Drosophila. This revealed the production of 2′3′-c-di-GMP, which has until now only been characterized as a synthetic analog of 3′3′-c-di-GMP31.

Figure 2: cGLR1 and cGLR2 produce several CDNs in D. melanogaster flies.

Figure 2:

A-B. Concentration of CDNs measured by LC-MS in the hemolymph (A) or in whole fly lysates (B) collected from male transgenic flies ectopically expressing cGLR1, cGLR2, their inactive cGLRAFA versions or a control transgene (GFP). C. Concentration of CDNs measured by LC-MS in the lysate of HEK293T cells overexpressing cGLR2 or GFP. Data are representative of one (A), three (B) or four (C) independent experiments and shown with dot plots with median and quartile. nd, non-detected. Concentration of 2'3'-c-di-AMP in whole flies was analysed by pairwise permutation test with FDR method (***<0.001). D. Thin-layer chromatography (TLC) analysis of α-32P labeled Dp-cGLR2 reaction products and treatment with P1 nuclease, which cleaves 3′–5′ phosphodiester linkages. Specific labeling reveals that only G nucleobases are incorporated in the cGLR2 product and P1 treatment supports that the cGLR2 product contains one 3′–5′ bond and one P1-insensitive bond. Data are representative of n= 3 independent experiments. E. HPLC chromatogram showing C18 elution profile of Dp-cGLR2 reaction; dashed lined indicates retention times of synthetic nucleotide standards. Data are representative of n= 3 independent experiments. F. High-resolution mass spectrometry confirms the major Dp-cGLR2 product as 2′3′-c-di-GMP. See also Figures S1, S2, S3, S4 and S5.

In our experiments the production of 2′3′-c-di-GMP in flies and transfected cells correlated exclusively with the expression of catalytically active cGLR2 (Figure 2A,B). To determine if cGLR2 produced 2′3′-c-di-GMP as a nucleotide second messenger, we investigated the in vitro activity of this enzyme. We first performed a systematic screen of cGLR2 homologs encoded in 13 Drosophila species (Figure S5A-D) and discovered that D. pseudoananassae (Dp) cGLR2 and D. bipectinata (Db) cGLR2 synthesizes CDN products in vitro (Figures 2D and S5E). Targeted mutation of the Dp-cGLR2 metal-coordinating residue D82 confirmed that CDN synthesis was dependent on the conserved cGLR catalytic site (Figure S6A,B)32. Using nucleobase specific labeling and nuclease digestion to analyze the cGLR2 product, we determined that this CDN is produced exclusively from GTP substrates and contains a P1-insensitive phosphodiester linkage (Figures 2D and S5E) indicating the presence of a 2′–5′ bond. We next analyzed cGLR2 reactions using high-performance liquid chromatography (HPLC) and observed that the major cGLR2 product exhibits a C18 chromatography profile identical to a 2′3′-c-di-GMP synthetic standard (Figures 2E and S5D,F). Using LC-MS/MS analysis in comparison to the synthetic standard, we confirmed that 2′3′-c-di-GMP is major product of Drosophila cGLR2 (Figures 2F and S5G,H). We observed that cGLR2 exclusively synthesizes 2′3′-c-di-GMP in vitro, in contrast to the production of both 3′2′-cGAMP and 2′3′-c-di-AMP by cGLR121. We note that both cGLR1 and cGLR2 exhibit more specific CDN synthesis activity in vitro than upon overexpression in flies.

DCV infection triggers cGLR-dependent production of CDNs in Drosophila.

We next monitored production of CDNs in virus-infected flies. We used dSTING mutant flies in these experiments to maximize the ability to detect free CDNs33. Only 2′3′-c-di-AMP could be detected in the extracts from non-infected flies (note that 3′3′-c-di-AMP was also present in the extracts). However, systemic DCV infection resulted in the induction of 2′3′-cGAMP, 3′2′-cGAMP and 2′3′-c-di-GMP. The quantity of 2′3′-c-di-AMP also significantly increased in DCV-infected flies (Figure 3A). In addition, 2′3′-cGAMP, 3′2′-cGAMP, 2′3′-c-di-GMP and 2′3′-c-di-AMP were detected in the hemolymph of DCV-infected dSTING mutant flies (Figure 3B). Expression of the four CDNs was also upregulated following enteric infection with another natural Drosophila pathogen, Nora virus34 (Figure 3C). To assess the contribution of cGLRs to this response, we repeated the experiment using cGLR1/2 mutant flies recombined in dSTING null mutant background. Production of 2′3′-cGAMP and 3′2′-cGAMP in response to DCV infection decreased significantly when both cGLR1 and cGLR2 were absent but was not affected in single mutant flies (Figure 3A). A small but significant decrease of 2′3′-c-di-GMP was observed in cGLR2 mutant flies. The concentration of 2′3′-c-di-GMP further dropped when both cGLR1 and cGLR2 were mutated, revealing that cGLR1 contributes to the production of 2′3′-c-di-GMP in the context of a viral infection in flies, although it does not when it is ectopically expressed in transgenic flies (Figure 2A). We hypothesize that the lack of a physiologically relevant ligand to fully activate cGLR1 in transgenic, uninfected flies results in partial activity of the receptor. We note that production of these three CDNs could still be detected in the cGLR1/2 double mutant flies. In contrast to 2′3′-cGAMP, 3′2′-cGAMP and 2′3′-c-di-GMP, the upregulation of 2′3′-c-di-AMP by DCV infection was still observed in the absence of cGLR1 and cGLR2 (Figure 3A).

Figure 3: DCV infection triggers cGLR-dependent production of CDNs in D. melanogaster.

Figure 3:

A. Concentration of CDNs measured by LC-MS in whole fly lysates from Tris- or DCV- injected flies. Data are from at least four independent experiments. B. Concentration of CDNs in the hemolymph of Tris- or DCV-injected dSTING knockout flies. Data are from three independent experiments. Each experiment involves 1000 flies. C. Concentration of CDNs in whole fly lysates from Nora-infected dSTING knockout flies. Data are from four independent experiments, each involving 500 flies. All data are shown with boxplot with scatter plot and were analyzed using permutation test with FDR method (ns= non significant, *<0.05, **<0.01, ***<0.001 and ****<0.0001), non-detected (nd). See also Figure S4.

dsRNA binding of Drosophila cGLR2 facilitates its production of 2′3′-c-di-GMP

We previously determined that Drosophila cGLR1 functions as a dsRNA sensor, consistent with its role in CDN production during RNA virus infection20,21. To determine how cGLR2 is activated to produce CDNs, we monitored in vitro CDN synthesis in the presence of different nucleic acid ligands (Figure 4A,B). We found that Dp- and Db-cGLR2 synthesize a low amount of 2′3′-c-di-GMP in the absence of exogenous nucleic acid ligands and that activation is enhanced most strongly in the presence of dsRNA (Figures 4A and S6C). We additionally used HPLC to monitor Dp-cGLR2 reactions and verified that dsRNA facilitates 2′3′-c-di-GMP synthesis (Figure 4B).

Figure 4: dsRNA binding of Drosophila cGLR2 facilitates its production of 2'3'-c-di-GMP.

Figure 4:

A. TLC analysis and quantification of GTP conversion to 2′3′-c-di-GMP by Dp-cGLR2 in the presence of different nucleic acid ligands. Data are mean +/- s.e.m. of n= 3 individual experiments. B. HPLC chromatogram showing 2′3′-c-di-GMP synthesis by Dp-cGLR2 in the presence of dsRNA mimic Poly I:C or a buffer control. Data are representative of n= 3 independent experiments. C. Surface electrostatic view of predicted Dp-cGLR2 structure modeled with a 19 bp dsRNA ligand, highlighting predicted interacting residues selected for mutagenesis. D. TLC analysis and quantification of wildtype (WT) and mutant Dp-cGLR2 activity in the absence and presence of dsRNA mimic Poly I:C. For quantification, % GTP conversion to 2′3′-c-di-GMP for each reaction was normalized to the wildtype control mean value for either buffer or Poly I:C reactions. Data are mean +/- s.e.m. of n= 3 individual experiments. See also Figures S5 and S6.

To understand how cGLR2 recognizes nucleic acid ligands we modeled Dp-cGLR2 with a short dsRNA ligand and designed charge-swap mutations to conserved basic residues in the predicted interaction surface (Figures 4C and S6D). Many of these residues are conserved in cGAS and Drosophila cGLR1, providing a structural explanation for the role of cGLR2 as a nucleic acid sensor20,21,3537 (Figure S5B). To understand the role of the highly basic, disordered N-terminus of Dp-cGLR2, we additionally generated a truncation mutant starting at residue N11. Dp-cGLR2 truncation and charge-swap mutants variably disrupted CDN synthesis by cGLR2 (Figure 4D). Truncation of the Dp-cGLR2 N-terminus resulted in ligand-independent nucleotide product synthesis, suggesting that the disordered N-terminus negatively regulates enzymatic activity in the absence of dsRNA. A charge swap mutation to R29 similarly resulted in robust, ligand-independent product synthesis by cGLR2. Similar to cGAS and Drosophila cGLR1, basic residues R50, K255, and K263 were critical for cGLR2 activity, supporting a common role for these positions in nucleic acid sensing by cGLR enzymes21,3537. Together our mutational analysis of Dp-cGLR2 supports that the shared cGLR ligand binding surface is critical for enzymatic activity of cGLR2 and that conserved basic residues control dsRNA-induced CDN synthesis.

2′3′-c-di-GMP triggers a potent antiviral response in flies.

We previously reported that injection of 2′3′-cGAMP and 3′2′-cGAMP, but not 3′3′-c-di-GMP, primes antiviral immunity, such that injected flies display increased resistance to DCV19,21. Here, we compared the effects of all four CDNs detected as cGLR1/2 products and found that injection of 2′3′-c-di-AMP also efficiently protected flies against systemic infection by DCV. However, the strongest protection was achieved following injection of 2′3′-c-di-GMP (Figure 5A). No protection was observed in dSTING mutant flies, whatever the CDN injected, confirming that the four CDNs act as STING agonists. Injection of 2′3′-c-di-GMP also resulted in better protection against the rhabdovirus Vesicular Stomatitis Virus (VSV), as attested by reduced viral RNA load 4- and 5-days post-infection (Figure 5B). Accordingly, 2′3′-c-di-GMP was a potent inducer of STING signaling, as illustrated by the strong induction of dSTING, srg1, srg2 and srg3 (Figure 5C). Genome wide analysis of the transcriptional response of flies injected with 3′2′-cGAMP or 2′3′-c-di-GMP revealed that although the two CDNs upregulated the same set of genes, the response was markedly enhanced when 2′3′-c-di-GMP was used (Figure 5D). We conclude that 2′3′-c-di-GMP is a potent agonist of STING in Drosophila, with functional relevance for the induction of antiviral immunity.

Figure 5: 2'3'-c-di-GMP triggers a potent antiviral response in D. melanogaster.

Figure 5:

A. Survival of control (isogenic w1118) and dSTING knockout flies pretreated with the indicated CDNs before systemic DCV infection. Data are from four independent experiments, each with three independent groups of around ten flies and analyzed by Log-rank test (ns= non significant and ****<0.0001). B. Control flies pre-injected with the indicated CDNs or Tris were challenged with VSV and viral RNA load was monitored after 4- or 5-days post injection. Data are from four independent experiments, each performed in biological triplicates (n = 12). Data are shown with boxplot with scatter plot and were analyzed using permutation test with FDR method (ns= non significant, *<0.05, **<0.01, ***<0.001 and ****<0.0001). C. Relative gene expression of the STING-regulated genes dSTING, srg1, srg2 and srg3 seven days after injection of the indicated CDNs in control and dSTING knockout flies. Samples were collected from three independent experiments, each involving three groups of six flies including three males and three females. Data are shown with boxplot with scatter plot and were analysed using permutation test with FDR method (ns= non significant, *<0.05, **<0.01 and ***<0.001). D. Expression profiles of control male D. melanogaster (isogenic w1118) seven days after injection with Tris, 3′2′-cGAMP or 2′3′-c-di-GMP. All differentially expressed genes (DEGs) including upregulated and downregulated genes are shown in log2(FPKM+1). Data are from three independent experiments.

We next tested the capacity of 2′3′-c-di-GMP to trigger antiviral immunity in the three species in which we failed to see an anti-DCV effect with 2′3′-cGAMP, 2′3′-c-di-AMP or 3′2′-cGAMP (Figure 1B). Injection of 2′3′-c-di-GMP, but not 3′2′-cGAMP, resulted in significant inhibition of DCV replication in D. serrata, D. sechellia and D. mojavensis, 2- and 3-days post-infection (Figure 6AC). Accordingly, genome wide analysis of the transcriptome of D. serrata flies injected with CDNs revealed that, while 3′2′-cGAMP did not induce large scale gene expression compared to injection of Tris buffer, 2′3′-c-di-GMP induced a robust transcriptional response (Figure 6D). This effect was specific to Drosophila and we only observed a minor effect of 2′3′-c-di-GMP on STING signaling in human cells, as previously reported31. One potential explanation for the distinct signaling ability of CDNs in different Drosophila species could be species-specific expression of nucleases that degrade CDN signals. To investigate this possibility, we analyzed the relative stability of 2′3′-c-di-AMP, 2′3′-c-di-GMP, 2′3′-cGAMP and 3′2′-cGAMP in the presence of lysates from a panel of Drosophila species (Figure S7 AD). We found that all four CDNs are highly stable in each of the tested species and conclude that the relative stability of each molecule cannot explain species-specific differences in immune signaling.

Figure 6: 2'3'-c-di-GMP triggers antiviral immunity in D. serrata, D. sechellia and D. mojavensis.

Figure 6:

A-C. Relative DCV RNA loads at two or three days post-DCV infection in D. serrata (A), D. sechellia (B) and D. mojavensis (C) male flies pre-injected with Tris or the indicated CDNs. Data are from four independent experiments. Data are shown with boxplot with scatter plot and were analysed using pairwise permutation test with FDR method (ns= non significant, *<0.05, **<0.01 and ***<0.001). D. Expression profiles of D. serrata male flies seven days after injection with Tris, 3′2′-cGAMP or 2′3′-c-di-GMP. All differentially expressed genes (DEGs) including upregulated and downregulated genes are shown in log2(FPKM+1). Data are from three independent experiments. E-F. Analysis of the change in thermophoresis in dSTING::eGFP fusion proteins upon incubation with increasing doses of the indicated CDNs in lysates from S2 cells transfected with expression vectors for the D. melanogaster (E) or D. serrata (F) proteins. The three data points corresponding to the most concentrated doses of 2′3′-c-di-GMP were not taken into account for the calculation of the Kd. Data are from 2 to 4 independent experiments. See also Figure S7.

To understand how Drosophila STING recognizes 2′3′-c-di-GMP as a potent agonist, we modeled dSTING–2′3′-c-di-GMP interactions based on the dSTING–3′2′-cGAMP structure21. Our predictive modeling indicated that there are no steric clashes between dSTING residues and 2′3′-c-di-GMP in the CDN binding pocket and that the specific contacts which control 3′2′-cGAMP recognition also have the potential to mediate 2′3′-c-di-GMP binding (Figure S7E). Specifically, our model predicted that each nucleobase of 2′3′-c-di-GMP stacks between Y164 and R234 residues extending from each protomer of the STING dimer and N159 coordinates the free 3′-OH of one ribose. In the dSTING–3′2′-cGAMP complex, E257 from one protomer contacts the guanosine N2 position. In our modeled dSTING–2′3′-c-di-GMP complex, this contact was conserved and E257 from the opposing protomer was predicted to additionally readout the N2 position of the second guanosine. We also observed that in our predicted dSTING–2′3′-c-di-GMP complex, T260 from one protomer coordinates the N2 position of one guanosine nucleobase, forming an additional contact that could potentially increase the specificity of dSTING for 2′3′-c-di-GMP. Residues N159, Y164, R234, E257, and T260 are highly conserved throughout the Drosophila genus, suggesting that 2′3′-c-di-GMP has the potential to serve as a potent agonist in most fly species (Figure S7F). We note that 2′3′-c-di-GMP adopts a compact regiochemical conformation similar to other CDNs with mixed 2′−5′ and 3′−5′ phosphodiester linkages, including 3′2′-cGAMP and 2′3′-cGAMP, supporting the formation of a tightly closed STING dimer critical for downstream immune signaling38,39. Residues controlling the specific recognition of 3′3′- linked CDNs by bacterial and animal STINGs26,40,41 are absent in Drosophila STING, explaining the inability of the 3′3′-c-di-GMP isomer to trigger STING antiviral immunity in flies. We finally note that our analysis of Drosophila STING does not suggest a structural explanation for the inability of 3′2′-cGAMP to induce antiviral immunity in certain species, as the residues controlling specific recognition of this molecule are widely conserved21 (Figure S7F).

We next used microscale thermophoresis to monitor the affinity of STING for 3′2′-cGAMP and 2′3′-c-di-GMP in cell extracts42. These experiments revealed that STING proteins from D. melanogaster and D. serrata bind 2′3′-c-di-GMP with higher affinity (Kd=28.2μM and 36.7μM, respectively) than 3′2′-cGAMP (Kd=1mM and 0.4mM, respectively) (Figure 6E, F). Of note, we observed a biphasic curve when the ligand was 2′3′-c-di-GMP, which may reflect dimerization of the protein at high CDN concentration43. This experiment also revealed a lower thermophoresis signal amplitude for 3′2′-cGAMP for the STING protein from D. serrata compared to the STING protein from D. melanogaster. This may indicate a more extensive conformational change triggered by 3′2′-cGAMP in the D. melanogaster protein, possibly related to signaling efficiency44.

cGLR-STING signaling evolved rapidly in Drosophila.

Drosophila species possess a variable set of cGLR genes, and we previously reported that individual species are predicted to encode as many as seven enzymes21. In total, we identified 207 genes encoding cGLRs in Drosophila species, including 61 cGLR genes in the 14 species we focused on for our in vivo analysis (Figure 7A,B). These cGLRs belong to three major clades, with evidence of multiple gene duplication and loss events. Both Dm-cGLR1 and Dm-cGLR2 belong to a first clade in which they define two subclades. The cGLR1 subclade contains 16 genes, one in each of the 14 species, with a duplication in the Drosophila subgenus (D. mojavensis and D. hydei). The cGLR2 subclade results from a duplication of cGLR1 in the Sophophora subgenus and contains 10 genes (Figures 1A and 7A). The major clade, with 26 genes in the 14 species, includes the D. melanogaster gene CG7194, the closest homolog of cGAS in Drosophila which we refer to as “cGLR3”. Members from this clade are present in all of the species we focused on in this study, except D. virilis, with a number of genes varying between 1 (D. melanogaster, D. simulans, D. sechellia, D. mojavensis and D. hydei) and 4 (D. serrata, D. kikkawai, and D. suzukii), and evidence for multiple duplications and losses. Finally, members of the third clade are present in 9 of our species of interest, revealing independent losses in different branches (Figure 7A). Of note, enzymes from this clade exhibit a low predicted isoelectric point (pI), unlike the other cGLRs and mammalian cGAS, which have a high pI reflecting the presence of a long positively charged surface accommodating nucleic acid ligands (Figure 7A).

Figure 7: Rapid evolution of cGLR-STING signaling in Drosophila.

Figure 7:

A. Phylogenetic tree of the cGLRs identified in Drosophila genomes (n= 207). Sequences of predicted cGLR were aligned using MAFFT and the aligned sequences were used to construct the tree. The names of the cGLRs from the 14 Drosophila species are indicated and those able to activate STING signaling are labelled in red while the inactive ones are in black. The red stars indicate cGLRs from D. melanogaster. The 8 cGLRs in grey were not tested. Isoelectric point (pI) annotated by color gradient where acidic pIs are shown in red and basic pIs are shown in blue. B. The predicted cGLRs encoded in the genome of the 14 Drosophila species. The cGLRs able to activate STING signaling are labelled in red. C. Heatmap showing log2 fold change intensities in dSTING luciferase reporter gene activity in S2 cells expressing the indicated 52 candidate cGLRs in comparison to control. Four conditions were tested with empty vector, STING, cGLR and cGLR+STING from the same species. 15 positive candidates highlighted with dots were selected by mixed-effect model (RELM) with p value ≤0.05 (Tukey correction for multiple hypothesis testing) and with fold change > 2 in cGLR or cGLR with STING conditions. Data are from 2 to 4 independent experiments. D. Concentration of 2′3′-c-di-GMP measured by LC-MS/MS in lysates from dSTING knockout S2 cells ectopically expressing the indicated cGLRs. Data are from 5 independent experiments.

Expression vectors for 52 cGLR genes from the 14 species were constructed and transfected in S2 cells together with vectors expressing STING from the corresponding species and a dSTING-luciferase reporter plasmid, to monitor activation of the pathway15. We identified 15 cGLRs, including D. melanogaster cGLR1 and cGLR2, that were able to activate STING signaling in S2 cells (Figure 7C). Most of them (13/15) belong to the cGLR1/2 clade. The two other active cGLRs, both from the species D. takahashii, belong to the same clade as the D. melanogaster gene CG7194 (cGLR3) (Figure 7A). Of note, several of them, including the cGLR1 orthologs from D. sechellia and D. serrata, produced 2′3′-c-di-GMP when transfected into S2 cells (Figure 7D). Overall, our findings confirm that cGLRs from different clades share the property to activate STING signaling and point to a dynamic evolution of these receptors in the Drosophila lineage, with 2′3′-c-di-GMP playing a major role at least in some species.

Discussion

We show here that CDNs are produced in D. melanogaster flies in response to virus infection, thus connecting sensing of viral infection to the enzymatic activity of cGLRs in vivo. We also report that a set of CDNs, rather than a single molecular species, are produced during infection. Of these molecules, the most potent STING agonist in D. melanogaster is a previously unknown CDN, 2′3′-c-di-GMP. This may explain our previous observation that transgenic flies ectopically expressing cGLR2, which produces this CDN, are better protected against DCV infection than flies ectopically expressing cGLR1, which does not produce 2′3′-c-di-GMP20. 2′3′-c-di-GMP also triggers a potent antiviral response in D. serrata, D. sechellia and D. mojavensis. Whether this can be generalized to all Drosophila species remains to be tested, although the differences observed between the potency of 2′3′-cGAMP, 2′3′-c-di-AMP and 3′2′-cGAMP according to the species (see Figure 1A) suggest that exceptions will be likely.

Our results contrast with the situation in mammals, where cGAS primarily synthesize 2′3′-cGAMP30,33, and highlights similarities with the CBASS system in bacteria, where CD-NTase enzymes can produce a broad diversity of cyclic di- or tri-nucleotides23,25,45,46. This diversity has been proposed to give an edge to bacteria to escape viral nucleases targeting them. Indeed, 3′2′-cGAMP is resistant to both poxin and Acb1 nucleases, which are produced respectively by insect DNA viruses and phages and readily cleave 2′3′- and 3′3′-cGAMP, respectively21,4750. An alternative explanation for the diversity of CDNs produced in Drosophila may be the existence of additional sensors, besides STING, which may preferentially recognize some species of CDNs. Indeed, the only known CDN receptor in mammals, besides STING, is RECON, which preferentially binds 3′–5′-linked CDNs and CTNs, rather than 2′3′-cGAMP23,51. However, these alternative sensors for CDNs remain unknown in Drosophila, and we note that the antiviral protection induced by the four CDNs we identified is fully dependent on STING. A third possibility could be related to extracellular functions of CDNs in flies. Indeed, we and others had previously shown that injection of CDNs into the fly body cavity results in potent STING-dependent antiviral protection, pointing to the existence of import mechanisms for CDNs16,19,21. We now show that CDNs are found circulating in the hemolymph upon viral infection, suggesting that CDN export mechanisms may operate in virus-infected cells. This would provide an efficient means to trigger a systemic response to infection and one may hypothesize that some CDNs are more efficiently transported in and out of cells and function as immuno-transmitters, whereas others function as bona fide second messengers in the cells in which they are produced. The observation that 3′3′-c-di-GMP protects D. melanogaster against enteric infection when fed to flies16 but has no effect on systemic antiviral immunity when it is injected in the body cavity16,19, supports the hypothesis that regulation of the transport across cellular membranes plays a role in the biology of CDNs. Finally, an additional possibility is that the diversity of CDNs produced reflects the number of cGLRs that produce them, possibly in different tissues.

The genes encoding cGLRs in Drosophila have experienced multiple duplication and losses, resulting in a number of receptors ranging from 2 to 7 according to the species and defining three major clades. Some 30% of the cGLRs tested (15 out of 52) can produce STING agonists in Drosophila S2 cells and most of the active enzymes we identified belong to the clade encompassing D. melanogaster cGLR1 and 2, with only two active enzymes identified in the clade encompassing the third cGLR in D. melanogaster, cGLR3. Our data reveal that the activity of D. pseudoananassae cGLR2, like that of D. melanogaster cGLR1, can be activated in vitro by dsRNA, confirming that cGLRs function as PRRs and that the clade to which cGLR1 and cGLR2 belong is primarily dedicated to sensing of viral RNAs. The observation that significant amounts of CDNs are still induced in response to DCV infection in cGLR1 or cGLR2 D. melanogaster mutant flies, confirm that both receptors can be independently activated by the virus and contribute to the control of the infection. We noted residual production of CDNs in DCV-infected cGLR1/2 double mutant flies, which suggests that cGLR3 (CG7194) may also be activated upon sensing viral infection in D. melanogaster. This hypothesis is supported by the observation that two members of the cGLR3 clade in D. takahashii can activate STING in transfected cells and that one of them produces 2′3′-c-di-GMP. Altogether, these findings raise the question of the biological relevance of the multiplicity of cGLRs encoded in the genome of Drosophila flies. In the case of D. melanogaster, we previously reported that induction of Sting, srg1, srg2 and srg3 in response to DCV infection was severely impaired or abolished in cGLR1 mutant flies, but was not affected in cGLR2 mutant flies, suggesting that cGLR1 plays a major role in the sensing of DCV infection. Yet, a stronger phenotype for survival to DCV infection and increased viral load was observed when both cGLR1 and cGLR2 were mutated (compare Figures 2d and 2a,c in ref.20). This latter result is fully consistent with the production of CDNs in response to DCV infection reported here. Overall, the genetic data at hand in D. melanogaster suggests that the CDNs produced by cGLR2 control DCV infection independently from the induction of the canonical STING-regulated genes. Further experiments are required to determine if the CDNs produced by cGLR2 contribute to the control of DCV infection through the induction of a different set of genes, possibly in a tissue-specific manner, or through a mechanism independent from transcription, e.g., autophagy28,52,53.

cGLRs within the cGLR1/2 clade and the cGLR3 clade are defined by characteristic positive residues in the ligand binding surface that control the recognition of negatively charged nucleic acids20,21. These residues contribute to an overall basic pI, consistent with other nucleic acid sensing cGLRs including cGAS41. We find that a third clade of Drosophila cGLRs have calculated pIs <6 and are predicted to have neutral and acidic ligand binding surfaces, suggesting they may not recognize nucleic acid ligands. No representative members of this clade have been characterized, and we predict these proteins could function as PRRs to detect non-nucleic acid pathogen associated molecules. Whether the lack of activity of the other cGLRs we tested here reflects the absence of activating ligands in S2 cells or the fact that their products cannot bind to and activate STING remains an open question, the answer to which will provide important insight on the biology of cGLRs. Similar species-specific differences between the activity of cGAS from different mammals, including within the primate lineage, have recently been described and shown to result, at least in part, from differences of subcellular localization of the receptor54.

Our findings nicely complement a recent study that identified more than 3,000 cGLRs in the genomes of most metazoans and characterized a subset of them as PRRs responding to dsRNA or dsDNA41. While most metazoan cGLR synthesize 2′3′-cGAMP, some cGLRs also produce alternative nucleotide signals including isoforms of c-UMP–AMP with pyrimidine bases, raising the possibility that some Drosophila cGLRs synthesize still additional CDNs besides the four we identified here. Overall, our results create a foundation to better understanding of the biology of cGLRs using the Drosophila model. Further experiments using cGLR1/2 double mutant flies or even cGLR1/2/3 triple mutant flies challenged by a panel of biotic or abiotic stresses may provide some clue regarding their function in a physiological setting. In addition, genetic complementation of cGLR mutant D. melanogaster with genes encoding cGLRs from other species is expected to shed light on their function, with possible implications for the function of uncharacterized cGLRs with complete catalytic sites encoded in human and other vertebrates (e.g., MB21D2)21.

Limitations of the study

This study reports the identification and quantification of CDNs produced in vivo in response to viral infection and the discovery of 2′3′-c-diGMP, which acts as a highly potent STING agonist in Drosophila. One limitation of our study is the variability in retention time between CDNs observed in the complex matrix of fly lysates compared to chemically synthesized CDNs, which may affect the accuracy of identification. In addition, the LC-MS/MS method using a triple quadrupole mass spectrometer lacks accuracy in distinguishing the isomers for some CDNs. Specifically, the amount of cGAMP isomers produced was monitored simultaneously using the quantifier ions m/z=136.0 and m/z=524.0 that produced the strongest signal in 2′3′-cGAMP and 3′2′-cGAMP, respectively. To address these limitations, future research should continue to focus on optimizing CDN extraction, improving the separation of CDNs using liquid chromatography, and utilizing high-resolution mass spectrometry to enhance the accuracy and specificity of CDN identification. Finally, 2′3′-c-di-GMP binds with higher affinity than 3′2′-cGAMP to STING from D. melanogaster and D. serrata, but our data do not explain why 3′2′-cGAMP is a poor STING agonist in D. serrata. Additional structural biology experiments will be required to address this point.

STAR METHODS

RESOURCE AVAILABILITY

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Hua Cai (chjorbe@hotmail.com).

Materials availability

Mutant fly strains and cell lines generated in this study are available upon request.

Data and code availability

  • The RNAseq data for D. melanogaster and D. serrata are available in the Genome Sequence Archive, National Genomics Data Center, China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (https://ngdc.cncb.ac.cn/gsa) and are publicly available from the date of publication. . Accession numbers are listed in the Key Resources Table. Raw data are accessible at Mendeley Data repository (https://data.mendeley.com/) (DOI: 10.17632/kcgwhnghvx.1).

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
     
     
     
     
     
Bacterial and virus strains
E. coli BL21-DE3 RIL Agilent Cat# 230245
E. coli TOP 10 Invitrogen Cat# C404006
Drosophila C virus Goto et al., 201815 N/A
Vesicular Stomatitis Virus Goto et al., 201815 N/A
Nora virus This study N/A
Biological samples    
     
     
     
     
     
Chemicals, peptides, and recombinant proteins
Ni-NTA Agarose Qiagen Cat# 30250
HiTrap Heparin HP column Cytiva Cat# 17040701
Zorbax Bonus-RP Agilent Cat# 863668–901
Alkaline Phosphatase, Quick CIP New England Biolabs Cat# M0525S
Nuclease P1 from Penicillium citrinum Sigma-Aldrich Cat# N8630
PEI-Cellulose F TLC plate EMD Biosciences Cat# EM1.05579.0001
ATP, [α-32P], 3000 Ci/mmol 10 mCi/ml Perkin Elmer Cat# BLU003H250UC
GTP, [α-32P], 3000 Ci/mmol 10 mCi/ml Perkin Elmer Cat# BLU006H250UC
ATP, GTP New England Biolabs Cat# N0450S
2′3′-c-di-GMP Biolog Life Science Institute Cat# C 182
Poly(I:C) (HMW) VacciGrade Invivogen Cat# vac-pic
2'3'-c-di-AMP Biolog Life Science Institute Cat# C 187
3′2′-cGAMP Biolog Life Science Institute Cat# C 238
2′3′-cGAMP Biolog Life Science Institute Cat# C 161
3′3′-c-di-GMP Biolog Life Science Institute Cat# C 057
Nucleodur Pyramid C18 column (3 μm, 50 x 3 mm) Macherey Nagel Cat# 760263.30
ZORBAX RRHD StableBond Aq column (1.8um, 2.1 x 100mm) Agilent Technologies Cat# 858700–914
     
Critical commercial assays
     
     
     
     
     
Deposited data
RNA-seq data for CDN-injected flies Genome Sequence Archive, National Genomics Data Center, China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (https://ngdc.cncb.ac.cn/gsa) Accession # CRA011428
Raw data Mendeley Data repository (https://data.mendeley.com/) DOI: 10.17632/kcgwhnghvx.1
     
     
     
Experimental models : Cell lines
Drosophila S2 cells Akira Goto et al., 201815 N/A
Drosophila STING-KO S2 cells This study N/A
HEK293T cells ATCC Cat# CRL-3216
     
     
Experimental models : Organisms/strains
DrosDel w 1118 Hua Cai et al., 202019 N/A
dSTING L76GfsTer11 Hua Cai et al., 202019 N/A
dSTINGRxn, Akira Goto et al., 201815 N/A
dSTING Control Akira Goto et al., 201815 N/A
iso : cGLR1 -/- ,dSTING -/- This study N/A
iso : cGLR2 -/- ,dSTING -/- This study N/A
iso : cGLR1+2 -/- ,dSTING -/- This study N/A
UAS-cGLR1 Andreas Holleufer et al., 202120 N/A
UAS-cGLR1 AFA Andreas Holleufer et al., 202120 N/A
UAS-cGLR2 Andreas Holleufer et al., 20215 N/A
UAS-cGLR2 AFA Andreas Holleufer et al., 202120 N/A
Act-GAL4/CyO ; TubG80[ts] IBMC N/A
D.willistoni Peking University, Laboratory of Jian Lu N/A
D.yakuba Peking University, Laboratory of Jian Lu N/A
D.simulans Peking University, Laboratory of Jian Lu N/A
D.virilis Peking University, Laboratory of Jian Lu N/A
D.takahashii Peking University, Laboratory of Jian Lu N/A
D.suzukii Chinese Academy of Sciences, Laboratory of Binyan Lu N/A
D.hydei Chinese Academy of Sciences, Laboratory of Binyan Lu N/A
D.serrata South China Agricultural University, Laboratory of Shuoyang Wen N/A
D.kikkawai South China Agricultural University, Laboratory of Shuoyang Wen N/A
D.pseudoobscura Chinese Academy of Sciences, Laboratory of Wei Wu N/A
D.mojavensis Southeast University, Laboratory of Yufeng Pan N/A
D.sechellia Lüdwig Maximilian University, Laboratory of Nicolas Gompel N/A
D.santomea Lüdwig Maximilian University, Laboratory of Nicolas Gompel N/A
     
Oligonucleotides
Oligonucleotide primers for qPCR, See Table S6 This study N/A
     
     
Recombinant DNA
pAC-Actin5C-Renilla luciferase E. Santiago, CNRS, Strasbourg, France N/A
pGL3/-200bp-dSTING promoter Akira Goto et al., 201815 N/A
pAC5.1 vector ThermoFischer Cat# V411020
pAC5.1/GFP IBMC N/A
pAC5.1-Dmel-STING This study N/A
pAC5.1-Dsim-STING This study N/A
pAC5.1-Dwil-STING IGE biotechnology N/A
pAC5.1-Dyak-STING IGE biotechnology N/A
pAC5.1-Dmoj-STING IGE biotechnology N/A
pAC5.1-Dvir-STING IGE biotechnology N/A
pAC5.1-Dhyd-STING IGE biotechnology N/A
pAC5.1-Dsan-STING IGE biotechnology N/A
pAC5.1-Dsec-STING IGE biotechnology N/A
pAC5.1-Dtak-STING IGE biotechnology N/A
pAC5.1-Dpse-STING IGE biotechnology N/A
pAC5.1-Dsuz-STING IGE biotechnology N/A
pAC5.1-Dser-STING IGE biotechnology N/A
pAC5.1-Dkik-STING IGE biotechnology N/A
pAC5.1-Dhyd-LOC111595620 This study N/A
pAC5.1-Dhyd-LOC111605403 This study N/A
pAC5.1-Dhyd-LOC111592833 This study N/A
pAC5.1-Dhyd-LOC111601518 This study N/A
pAC5.1-Dmoj-LOC6579490 This study N/A
pAC5.1-Dmoj-LOC6582322 This study N/A
pAC5.1-Dmoj-LOC6574034 This study N/A
pAC5.1-Dmoj-LOC6580632 This study N/A
pAC5.1-Dvir-LOC6625054 This study N/A
pAC5.1-Dvir-LOC6625472 This study N/A
pAC5.1-Dwil-LOC6638395 This study N/A
pAC5.1-Dwil-LOC6646488 This study N/A
pAC5.1-Dwil-LOC6638458 This study N/A
pAC5.1-Dpse-LOC6899115 This study N/A
pAC5.1-Dpse-LOC6900849 This study N/A
pAC5.1-Dpse-LOC6900403 This study N/A
pAC5.1-Dpse-LOC4803562 This study N/A
pAC5.1-Dtak-LOC108054184 This study N/A
pAC5.1-Dtak-LOC108066267 This study N/A
pAC5.1-Dtak-LOC108054321 This study N/A
pAC5.1-Dtak-LOC108067387 This study N/A
pAC5.1-Dtak-LOC108055687 This study N/A
pAC5.1-Dsuz-LOC108018091 This study N/A
pAC5.1-Dsuz-LOC108012585 This study N/A
pAC5.1-Dsuz-LOC108013198 This study N/A
pAC5.1-Dsuz-LOC108006881 This study N/A
pAC5.1-Dsuz-LOC108006833 This study N/A
pAC5.1-Dsuz-LOC108009543 This study N/A
pAC5.1-Dsuz-LOC108008397 This study N/A
pAC5.1-Dsec-LOC6609335 This study N/A
pAC5.1-Dsec-LOC116801260 This study N/A
pAC5.1-Dsim-LOC6734663 This study N/A
pAC5.1-Dsim-LOC6737253 This study N/A
pAC5.1-Dmel-cGLR1 This study N/A
pAC5.1-Dmel-cGLR2 This study N/A
pAC5.1-Dyak-LOC6531519 This study N/A
pAC5.1-Dyak-LOC6532544 This study N/A
pAC5.1-Dyak-LOC6530290 This study N/A
pAC5.1-Dyak-LOC6532149 This study N/A
pAC5.1-Dsan-LOC120447203 This study N/A
pAC5.1-Dsan-LOC120447932 This study N/A
pAC5.1-Dsan-LOC120445453 This study N/A
pAC5.1-Dser-LOC110178317 This study N/A
pAC5.1-Dser-LOC110191302 This study N/A
pAC5.1-Dser-LOC110179302 This study N/A
pAC5.1-Dser-LOC110184043 This study N/A
pAC5.1-Dkik-LOC108082553 This study N/A
pAC5.1-Dkik-LOC108071893 This study N/A
pAC5.1-Dkik-LOC108080761 This study N/A
pAC5.1-Dkik-LOC108078272 This study N/A
pAC5.1-Dkik-LOC108078001 This study N/A
pAC5.1-Dkik-LOC108072236 This study N/A
pAC5.1-Dmel-sting-eGFP This study N/A
pAC5.1-Dser-sting-eGFP This study N/A
pcDNA3.1-Dmel-cGLR2 IGE biotechnology N/A
     
     
     
     
     
     
     
Software and algorithms
MAFFT Katoh and Yamada56 https://mafft.cbrc.jp/alignment/software/
Geneious Prime (v2022.1.1) Biomatters, Ltd https://www.geneious.com/
iTOL Letunic and Bork59 https://itol.embl.de/
Phenix 1.13–2998 Liebschner et al.65 https://www.phenixonline.org/
Coot 0.8.9 Emsley and Cowtan66 https://www2.mrclmb.cam.ac.uk/personal/pemsley/coot/
PyMOL (v 2.4.2) Schrödinger, LLC https://pymol.org/
Xcalibur 2.2 Thermo Fisher Scientific, San Jose, CA, USA https://www.thermofisher.cn/order/catalog/product/OPTON-30967
Agilent MassHunter Acquisition (ver. B.08.00) Agilent Technologies, Santa Clara, CA, USA https://www.agilent.com.cn/zh-cn/product/software-informatics/mass-spectrometry-software/data-analysis
MO.Affinity Analysis(version 2.3) NanoTemper Technologies https://nanotempertech.com
R studio (version 4.1.2) The R project http://www.rstudio.com/products/rstudio
Coin package http://coin.r-forge.r-project.org/ https://cran.r-project.org/web/packages/coin/index.html
GraphPad Prism9 (version 9.2.0) GraphPad Software, LLC www.graphpad.com
Other
     
     
     
     

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Cell lines

HEK293T female cells (ATCC) were cultured in Dulbecco’s modified Eagle medium (DMEM) (Sigma-Aldrich) supplemented with 10% fetal bovine serum (FBS) (Excell Bio), 100 U mL−1 penicillin (Sigma-Aldrich) and 100 µg mL−1 streptomycin (Sigma-Aldrich) at 37°C and 5% CO2. Schneider 2 (S2) male cells (Invivogen) were cultured in Schneider’s Drosophila Medium (Thermal Fisher Scientific) supplemented with 10% FBS, 100 U mL−1 penicillin (Sigma-Aldrich) and 100 µg mL−1 streptomycin (Sigma-Aldrich) at 27°C. To generate Sting knockout cells by CRISPR-Cas9, three CRISPR RNAs targeting the first translated exon in the D. melanogaster Sting gene were cloned into the pAc-sgRNA-Casp vector20. After transfection, S2 cells were grown under selection with 5 µg mL−1 puromycin (Invitrogen) for two weeks. Knockout of Sting was verified by Sanger sequencing.

Drosophila strains

Fly stocks were raised on standard cornmeal agar medium at 25°C and were free of Wolbachia. All fly stocks used (DrosDel w1118, dSTINGL76GfsTer11, dSTINGRxn, dSTINGControl, cGLR1 or cGLR2 single knockout or double knockout flies, the transgenic lines of wild-type and AFA mutant versions of cGLR1 and cGLR2, and the GFP control) have been described previously15,19,20. The different species of Drosophila were kindly provided by Prof. Jian Lu, Peking University (D.willistoni, D.yakuba, D.simulans, D.virilis and D.takahashii); Prof. Binyan Lu, Chinese Academy of Sciences (D.suzukii and D. hydei); Prof. Shuoyang Wen, South China Agricultural University (D.serrata and D.kikkawai); Prof. Wei Wu, Chinese Academy of Sciences (D.pseudoobscura); Prof. Yufeng Pan, Southeast University (D.mojavensis) and Prof. Nicolas Gompel, Lüdwig Maximilian University (D.sechellia and D.santomea). A 50:50 mix of male and female adult flies collected three- to five-days post ecclosion was used in experiments, unless stated otherwise in the figure legends.

METHOD DETAILS

Plasmids

Coding sequences of predicted cGLRs were PCR-amplified from the cDNA prepared from flies from the different Drosophila species using 2×EasyTaq PCR SuperMix (TransGen Biotech, China) and cloned into the pAc5.1 vector using ClonExpress MultiS (Vazyme, China). The coding sequences of STING from fourteen species of Drosophila were synthesized by the IGE biotechnology company (China) and sub-cloned to pAc5.1 vector.

Antiviral effect of cyclic di-nucleotides in fourteen species of Drosophila

Cyclic dinucleotides including 3′2′-cGAMP (Biolog C238), 2′3′-cGAMP (Biolog C161), 2′3′-c-di-AMP (Biolog C187), 2′3′-c-di-GMP (Biolog C182) or 3′3′-c-di-GMP (Biolog C057) were dissolved in 10 mM Tris-HCl pH 7.5 and diluted to the indicated concentrations. 3–5 days old male flies were injected with 69 nL of cyclic dinucleotide solution or 10 mM Tris-HCl pH 7.5 (negative control) by intrathoracic injection using a Nanoject II apparatus (Drummond Scientific). 7 days post CDN injection, the flies were injected with 4.6 nL of DCV (50 PFU for all the tested species except D. melanogaster and D. yakuba, for which the dose was 5 PFU) in 10 mM Tris-HCl pH 7.5. Flies were collected 48 h and 72 h later in pools of 6 males and homogenized for RNA extraction and RT-qPCR analysis, as described.

Fly hemolymph and lysate preparation for Liquid Chromatography Mass Spectrometry (LC-MS)

For hemolymph collection, anesthesized adult flies were punctured using a tungsten needle and transferred to a 0.6 mL microcentrifuge tube (12 flies/tube) with a hole pierced in the bottom. The tube with flies was then transferred to a 1.5 mL Eppendorf tube. The two-tube assembly was then centrifuged for 10 min at 2,300 g at 4°C. The procedure was repeated until the hemolymph of one thousand flies was collected. Hemolymph was stored at −80°C.

For lysate preparation, 500 adult flies were collected in five 2 mL microfuge tubes (100 flies/ tube). 10 small zirconia beads were added per tube together with 100 μL precooled extraction reagent (2/2/1 [v/v/v] methanol, acetonitrile, water mixture) with 10 ng mL−1 3′3′-cAIMP as an internal standard in the LC-MS/MS analysis. The tubes with flies were frozen in liquid nitrogen and then homogenized using Precellys Evolution homogenizer (4°C, 5800 rpm, 2*30 s pause 12 s). The tubes were then centrifuged for 10 min at 12,000 g at 4°C. The supernatant was collected into a new microfuge tube, which was centrifuged again. The collected supernatant was transferred into 2 mL safe-lock tubes and heated at 95°C for 10 min, before cooling on ice. 500 µL chloroform was then added to the tubes, which were vortexed and placed at −20°C for 10 min. Subsequently, the tubes were centrifuged at 20,000 x g for 15 min, and the supernatants was transferred into fresh microfuge tubes. Next, the lysate was loaded on HLB SPE columns. The eluents were collected and then concentrated by evaporation and resuspended in 200 μL 0.1% formic acid water for LC-MS/MS analyses.

Identification of CDN using LC-MS

To detect the CDNs production in the lysate of DCV infected flies, high resolution LC-MS analysis was performed using a Ultimate3000 (ThermoFisher Scientific) coupled to a Quadrupole-Orbitrap Hybrid mass spectrometer (Q-Exactive, ThermoFisher Scientific). A volume of 5 μL sample was injected into a ZORBAX RRHD StableBond Aq column (1.8 μm, 2.1 x 100 mm; Agilent Technologie) maintained at 40°C. The mobile phase consisted of 5 mM ammonium carbonate (A) and acetonitrile (B). The following HPLC gradient was used: 0–14% B in 5.0 min, 14–25% B in 7.0 min, 25–100% B in 7.1 min, 100% B in 10.8 min, 100–0% B in 11.0 min, 0% B in 14.0 min; 0.300 mL min−1. Mass spectra were recorded using positive ion full scan mode with m/z from 300 to 1100. Accurate mass measurement was accomplished by Orbitrap-MS with a mass resolution of 70,000. The optimized MS parameters were set as follows: capillary temperature 350°C, maximum inject time 100ms, AGC target of 1.00E+06, and S-lens RF level 55. The target ions were sequentially isolated using high energy collision dissociation (HCD) fragmentation and progeny ions were detected with dd-MS2 mode. The parent ion was isolated with an isolation window of 2 m/z units, fragmented (Resolution = 17,500, nce=20, Maximum Inject Time: 50ms, Loop count: 5, TopN: 5). Identification of CDNs was performed by targeted mass analysis for exact masses and formulae for all possible CDNs. Xcalibur 2.2 (Thermo Fisher Scientific, San Jose, CA, USA) software was used for equipment control and data acquisition.

Measurement of CDN in the hemolymph or lysate

For measurement of 2'3'-cGAMP, 3'2'-cGAMP, 2'3'-c-di-AMP and 2'3'-c-di-GMP in the hemolymph, samples were analyzed using an Agilent 1290 System coupled to an Agilent 6470. A volume of 5 μL was injected into a Nucleodur Pyramid C18 column (3 μm, 50 x 3 mm; Macherey Nagel, Duren, Germany) maintained at 40°C. The mobile phase consisted of 0.1% formic acid in acetonitrile (A) and 0.1% formic acid in water (B). HPLC gradient are described in Table S1. The analytes were ionized by means of electro spray ionization in positive mode with the Delta EMV at 820 V. The source parameters are described in Table S2. For each CDN, the MRM transition(s) (m/z), Dwell, Frag (V) and CE (V) Cell Acc (V) are described in Table S3.

For measurement of 2'3'-cGAMP, 3'2'-cGAMP, 2'3'-c-di-AMP, 2'3'-c-di-GMP in whole fly lysates, the samples were analyzed using an Agilent 1290 System coupled to an Agilent 6470. A volume of 5 μL was injected into a Nucleodur Pyramid C18 column (50 x 3 mm; 3 μm Macherey Nagel, Duren, Germany) maintained at 40°C. The mobile phase consisted of 0.1% formic acid in acetonitrile (A) and 0.1% formic acid in water (B). The HPLC gradient and flow rate are described in Table S4. The analytes were ionized by means of electro spray ionization in positive mode with the Delta EMV at 820 V. The source parameters were described in Table S2. For each CDN, the MRM transition(s) (m/z), Dwell, Frag (V) and CE (V) Cell Acc (V) were described in Table S5.

LC-MS/MS analysis of in vitro cGLR2 reaction products

cGLR2 reaction products were generated in a 100 μL reaction with 5 μM cGLR enzyme, 200 μM GTP, 10 μg poly I:C, 1 mM MnCl2, and 50 mM Tris-HCl pH 7.5 and were analyzed by the commercial company MS-Omics using LC-MS/MS, as previously described41. Briefly, analysis was carried out using a Vanquish Horizon UHPLC System coupled to Orbitrap Exploris 240 Mass Spectrometer (Thermo Fisher Scientific, US).

First, UHPLC was performed using an Infinity Lab PoroShell 120 HILIC-Z PEEK lined column with the dimension of 2.1 × 150mm and particle size of 2.7 µm (Agilent Technologies). Mobile phase A was composed of 10 mM ammonium acetate, pH 9 in 90% Acetonitrile LC-MS grade (VWR Chemicals, Leuven) and 10% Ultra-pure water from Direct-Q® 3 UV Water Purification System with LC-Pak® Polisher (Merck KGaA, Darmstadt). Mobile phase B was composed of 10 mM ammonium acetate, pH 9 in ultra-pure water with 5 µM medronic acid (InfinityLab Deactivator additive, Agilent Technologies). The UHPLC column temperature was set at 30 °C and samples were analyzed at an injection volume of 5 µl. UHPLC was run using a flow rate kept at 250 µl mL−1 consisting of a 2 min hold at 10% B, increased to 40% B at 14 min, held till 15 min, decreased to 10% B at 16 min and held for 8 min.

For MS analysis, a heated electrospray ionization interface was used as ionization source and the analysis was performed in positive ionization mode from m/z 300 to 1500 at a mass resolution of 120000. Ion source parameters used: Sheath gas flow rate, 20 (arbitrary units); auxiliary gas flow rate, 5 (arbitrary units); Sweep gas flow rate, 1 (arbitrary units), capillary temperature, 350°C; S-lens radiofrequency level 70; automatic gain control (AGC) target, 1E6 (Standard); maximum injection time, 100 ms; spray voltage 3.5 kV in positive. MS2 spectra was acquired using data dependent acquisition (DDA) with the following parameters: mass resolution 45000, isolation window m/z 0.4 and normalized collision energy 20, 40 and 60 eV. Freestyle 1.4 (Thermo Fisher Scientific) was used to analyze data and generate MS/MS spectra.

CDN Stability Assay

Sf9 cell lysates were prepared from S. frugiperda Sf9 cells (Expression Systems) cultured in ESF 921 media. Pelleted cells were washed with 1× PBS, resuspended in 250 µL of a lysis buffer containing 20 mM HEPES-KOH pH 7.5, 150 mM NaCl, 5 mM MgCl2, 1 mM MnCl2, 1 mM DTT, 10% glycerol, and 1% NP-40, and incubated at room temperature for 30 min with occasional vortexing. Drosophila lysates were prepared in a similar manner from adult male flies of each respective species (55–120, depending on their sizes) using the same lysis buffer and grinding in Cryolis tubes. The total protein content of lysate samples was determined by Bradford assay and each sample was adjusted to 7.5 µg/µL. α-32P-labeled CDNs were synthesized by the following cGLR enzymes: 2′3′-cGAMP, Tc-cGLR; 3′2′-cGAMP and 2′3′-c-di-AMP, Ds-cGLR1; 2′3′-c-di-GMP, Db-cGLR2, as described above. 15 µL reactions containing 2.5 µM CDN and 1.5 µL Sf9 or Drosophila lysate in a buffer containing 50 mM HEPES-KOH 7.5, 10 mM KCl, 5 mM MgCl2, 1 mM MnCl2, and 1 mM TCEP were incubated at 37°C overnight and analyzed by TLC to determine CDN stability.

D. melanogaster cyclic dinucleotide injection and signaling analysis

Cyclic dinucleotides including 2'3'-c-di-GMP (Biolog C182), 2'3'-c-di-AMP (Biolog C187), 3′2′-cGAMP (Biolog C238), 2′3′-cGAMP (Biolog C161) and 3′3′-c-di-GMP (Biolog C057) were dissolved in 10 mM Tris-HCl pH 7.5 and diluted to the indicated concentrations. 3–5-day old adult flies were injected with 69 nL of cyclic dinucleotide solution or 10 mM Tris-HCl pH 7.5 (negative control) by intrathoracic injection using a Nanoject II apparatus (Drummond Scientific). Flies were collected 7 days later in pools of 6 individuals (3 males and 3 females) and homogenized for RNA extraction and quantitative PCR with reverse transcription (RT–qPCR) analysis, as described.

Bioinformatics and Drosophila cGLR and STING sequence analysis

Building on previous analyses21,23,41,55, Drosophila cGLRs were identified using the amino acid sequence of D. melanogaster cGLR1 (NP_788360.2) to seed a position-specific iterative BLAST (PSI-BLAST) search of Drosophila genomes (taxid:7215) in the NCBI non-redundant protein database. The PSI-BLAST search was performed with an E value cutoff 0.005 for inclusion into the next search round, BLOSUM62 scoring matrix, gap costs settings existence 11 and extension 1, and using conditional compositional score matrix adjustment. Iterative PSI-BLAST search was performed for 5 rounds and candidate Drosophila cGLR sequences were collected. Putative cGLR protein sequences were aligned using MAFFT (FFT-NS-i iterative refinement method)56; this alignment was used to construct a phylogenetic tree in Geneious Prime v2022.0.1 using the neighbor-joining method and Jukes-Cantor genetic distance model with no outgroup. Candidate proteins were analyzed by clade and selected for known cGLR domain organization and predicted structural homology to T. castaneum cGLR (PBD: 7LT2), including the presence of a conserved nucleotidyltransferase domain with a G[S/G] activation loop and [E/D]h[E/D] X50–90 [E/D] catalytic triad32. Manual analysis and curation of candidate cGLR sequences was performed based on alignments and predictive structural homology using Phyre257 and AlphaFold58. Manual refinement was also used to exclude duplicate sequences, gene isoforms, and proteins less than 250 residues. All cGLR sequences in the final tree were accessed from NCBI March 26th, 2023. The D. bipectinata cGLR2 sequence shown in Figure S5A,B and used for all biochemistry experiments was accessed from NCBI in January, 2020 under accession code XP_017096409.1. NCBI available genomes from 49 species in the Drosophila genus are represented in the final tree. iTOL was used for tree visualization and annotation59, including annotation of all cGLRs identified in fourteen Drosophila species of interest to this study. Isoelectric point was predicted by Geneious Prime software. Clustering of sequences in the final unrooted tree was used to define clades related to D. melanogaster cGLR1 (NP_788360.2), cGLR2 (A8DYP7.2), and cGLR3 (CG7194; AAF50449.1). The cGLR2-related clade was identified by the presence of D. melanogaster cGLR2 (A8DYP7.2) and extracted from the full cGLR tree for alignment using MAFFT (FFT-NS-i iterative refinement method), shown in Figure S5A. cGLR2 sequences of interest in Figure S5B were separately aligned and the secondary structure of D. pseudoananassae cGLR2 was generated based on the AlphaFold predicted structure.

Drosophila STING proteins were identified as a subset of eukaryotic STING proteins identified in ref.41. STING sequences were accessed from NCBI May 2022. Drosophila STING protein sequences from fourteen species of interest were aligned using MAFFT (FFT-NS-i iterative refinement method)56. Alignment in Figure S7F is shown in relation to the secondary structure of the D. eugracilis−3′2′-cGAMP complex (PDB: 7MWZ)21.

Screening of cGLRs based on cellular STING signaling assays

To test the activation of STING promoter luciferase reporter by predicted cGLRs, 96-well tissue culture plates were seeded with 2.5 x 105 S2 cells per well. After 3h, each well was transfected with 200ng pGL3 plasmid expressing firefly luciferase under transcriptional control of the Sting promoter, 25ng pAc5.1 plasmid constitutively expressing Renilla luciferase, 75ng pAc5.1 plasmid expressing predicted cGLRs, 25ng pAc5.1 plasmid expressing Sting, finally empty Ac5.1 plasmid to reach a total amount of 325ng plasmid for each well. All transfections of S2 cells were performed using lipofectamine 3000 (Invitrogen, Thermo Fisher Scientific) according to the manufacturer’s instructions. After 48 hours of transfection, cells were lysed in 100µL Lysis Buffer (Promega) per well. Firefly and Renilla luciferase activity was measured on 75µL lysate using the Dual-Luciferase® Reporter Assay System (Promega).

RNA-Sequencing of D. melanogaster injected with CDNs

Male flies of D. melanogaster or D. serrata were injected with 69 nL/fly of either 10 mM Tris (pH 7.5), 2'3'-cGAMP (0.9 mg mL−1) or 2'3'-cdi-GMP (0.9 mg mL−1) by intrathoracic injection (Nanoject II apparatus) in three independent experiments. Injected flies were collected in pools of 6 individuals at 7 days post injection. Total RNA was isolated from injected flies using TRIzol Reagent (Invitrogen), according to the manufacturer's protocol. RNA quality was assessed on an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA, USA) and checked using RNase free agarose gel electrophoresis. After total RNA was extracted, eukaryotic mRNA was enriched by Oligo(dT) beads. Then the enriched mRNA was fragmented into short fragments using fragmentation buffer and reverse transcribed into cDNA by using NEBNext Ultra RNA Library Prep Kit for Illumina (NEB #7530, New England Biolabs, Ipswich, MA, USA). The purified double-stranded cDNA fragments were end repaired, A base added, and ligated to Illumina sequencing adapters. The ligation reaction was purified with the AMPure XP Beads (1.0X). Ligated fragments were subjected to size selection by agarose gel electrophoresis and polymerase chain reaction (PCR) amplified. The resulting cDNA library was sequenced using Illumina Novaseq6000 by Gene Denovo Biotechnology Co. (Guangzhou, China).

Transcriptome analysis

After filtering by fastp60 (version 0.18.0), reads were mapped using HISAT2. 2.461 with “-rna-strandness RF” and other parameters set as a default to the genome of D. melanogaster (Ensembl_release102) and D. serrata (GCF_002093755.1). RNAs differential expression analysis was performed by DESeq262 software between two different groups (and by edgeR between two samples). The genes/transcripts with the parameter of false discovery rate (FDR) below 0.05 and absolute fold change≥2 were considered differentially expressed genes/transcripts. The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive (Genomics, Proteomics & Bioinformatics 2021) in National Genomics Data Center (Nucleic Acids Res 2022), China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (GSA: CRA011428) that are publicly accessible at https://ngdc.cncb.ac.cn/gsa63,64.

Protein expression and purification

Recombinant cGLR proteins were expressed and purified using methods previously optimized for human cGAS and Drosophila cGLRs, as described previously21,37. Full length cGLR coding sequences were codon-optimized for expression in E. coli and cloned from synthetic constructs (GeneArt or Integrated DNA Technologies) into a custom pET16 expression vector with an N-terminal 6×His-MBP fusion tag. Briefly, transformed BL21-CodonPlus (DE3)-RIL E. coli (Agilent) were grown in MDG media overnight prior to inoculation of M9ZB media at an OD600 of 0.0475. M9ZB cultures were grown to OD600 of 2.5 (approximately 5 h at 37°C with shaking at 230 rpm) followed by cooling on ice for 20 min. Cultures were induced with 500 μM IPTG prior to incubation at 16°C overnight with shaking at 230 rpm. Cultures were pelleted the following day and either flash frozen in liquid nitrogen for storage at −80°C or directly lysed for purification.

For large-scale protein purification, proteins were expressed with a 6×His-MBP fusion tag and grown as ~4–8× 1 L cultures in M9ZB media. Pellets were lysed by sonication in lysis buffer (20 mM HEPES pH 7.5, 400 mM NaCl, 30 mM imidazole, 10% glycerol and 1 mM DTT) and clarified by centrifugation at ~47,850 × g for 30 min at 4°C and subsequent filtration through glass wool. Recombinant protein was purified by gravity-flow over NiNTA resin (Qiagen). Resin was washed with lysis buffer supplemented to 1 M NaCl and then eluted with 20 mL of lysis buffer supplemented to 300 mM imidazole. MBP-tagged fusion proteins were buffer exchanged into lysis buffer with 4% glycerol and no imidazole to optimize conditions for overnight cleavage by recombinant TEV protease at ~10°C. cGLR proteins were next purified by ion exchange chromatography using 5 mL HiTrap Heparin HP columns (GE Healthcare) and eluted across a 150–1000 mM NaCl gradient in buffer with 10% glycerol. Target protein fractions were pooled concentrated to ~10–30 mg mL−1 and flash-frozen with liquid nitrogen and stored at −80°C for biochemistry experiments.

Nucleotide product synthesis analysis

cGLR nucleotide synthesis activity was analyzed by thin-layer chromatography as previously described21,23. For all biochemistry reactions analyzed by TLC, 5 μM recombinant protein preparations were incubated in 5 or 10 μL reactions containing 0.5 μL α-32P labeled ATP or GTP, 200 μM unlabeled NTPs, and 1 mM MnCl2 in a final reaction buffer of 50 mM Tris-HCl pH 7.5, ~50 mM KCl (final KCl = 100 mM), 1 mM TCEP. Reactions were additionally supplemented with ~1 μg poly I:C or 5 μM nucleic acid ligands, as indicated. Besides poly I:C all nucleic acids used in this study are 40 nucleotide (nt) or base pairs (bp) in length. Reactions were incubated at 37°C for two hours and subsequently treated with 1 μL Quick CIP phosphatase (New England Biolabs) for 20 min at 37°C to remove unreacted phosphate signal. 0.5 μL of each reaction was spotted on a 20-cm × 20-cm PEI-cellulose thin-layer chromatography plate. Plates were run with 1.5 M KH2PO4 solvent until ~2.5 cm from top of the plate, dried at room-temperature, and exposed to a phosphor-screen prior to signal detection with a Typhoon Trio Variable Mode Imager System (GE Healthcare). TLC images were adjusted for contrast using FIJI40 and quantified using ImageQuant (8.2.0). GTP conversion to nucleotide product formation was measured according to the ratio of product to total signal for each reaction.

Nuclease P1 cleavage analysis was performed using cGLR2 reactions labeled with either α-32P-ATP or α-32P-GTP as previously described21,23. Briefly, radiolabeled nucleotide products were incubated with Nuclease P1 (80 mU, Sigma N8630) in buffer (30 mM NaOAc pH 5.3, 5 mM ZnSO4, 50 mM NaCl) for 30 min in the presence of Quick CIP (NEB).

Nucleotide purification and HPLC analysis

Enzymatic synthesis of cGLR nucleotide products for HPLC analysis was performed using 100 μL reactions containing 5 μM cGLR enzyme, 100 μM ATP, 100 μM GTP, 10 μg poly I:C, 1 mM MnCl2, and 50 mM Tris-HCl pH 7.5. KCl was adjusted to a final concentration of 100 mM. Reactions were incubated at 37°C for 2 h and then terminated by 2 min incubation at 95°C. Nucleotide product was recovered by filtering reactions through a 30-kDa cutoff concentrator (Amicon) to remove protein. Nucleotide products were separated on an Agilent 1200 Infinity Series LC system using a C18 column (Zorbax Bonus-RP 4.6 × 150 mm, 3.5 μm) at 40°C. Products were eluted at a flow rate of 1 mL min−1 with a buffer of 50 mM NaH2PO4 pH 6.8 supplemented with 3% acetonitrile.

Synthetic cyclic dinucleotide standards

Synthetic nucleotide standards used for HPLC analysis and mass-spectrometry analysis were purchased from Biolog Life Science Institute: 2′3′-cGAMP (cat no. C 161), 3′2′-cGAMP (cat no. C 238), 2′3′-c-di-AMP (cat no. C 187) and 2′3′-c-di-GMP (cat no. C 182).

dSTING and 2′3′-c-di-GMP structural modeling

Coordinate and cif restraint files for 2′3′-c-di-GMP were generated by the eLBOW program on PHENIX65 using the ChemDraw v.20.0.38 generated SMILES for this ligand. The resulting files were used to model the 2′3′-c-d-GMP ligand in complex with D. eugracilis STING by fitting to the 3′2′-cGAMP ligand density in the dSTING3′2′-cGAMP complex density map (PDB 7MWZ) using Coot66.

MicroScale thermophoresis measurement

S2 cells seeded in 10 cm plates at a density of 10 million cells per plate and transfected with 10μg of plasmid DNA via calcium chloride for three days. The cells were harvested by centrifuging them in a 15 mL tube at 135g and 4°C for 5 minutes. The supernatant was discarded, and the cell pellet was resuspended in 5 mL of PBS and centrifuged again at for 5 minutes. 300μL of RIPA buffer (1% NP-40, 0.5% deoxycholate, 0.1% SDS) containing protease inhibitors was added to the cells and the samples were incubated on ice with gentle agitation for 15 minutes. The samples were then centrifuged at 21,000g, 4°C for 45 minutes. The supernatant was carefully collected in a fresh Eppendorf tube and stored at 4°C. The protein lysate was adjusted to 400 fluorescence units (FI units) after a pretest. Dilution was performed using PBS-T buffer (1X PBS containing 0.05% Tween 20). Measurements using a Monolith NT.115 (NanoTemper Technologies) were performed at 25°C using 90% Excitation power and Medium MST power. Data analyses were performed using MO.Affinity Analysis software6769.

Statistical analysis

All statistical analyses were performed in R (version 4.1.2) and GraphPad Prism9. To perform the qPCR analysis, 2∆Ct values calculated for each sample were tested by permutation test using coin package. In addition, survival analyses using Log-rank tests were performed to compare each condition to control lines. cGLRs from different species were analysed using mixed-effect model (RELM) with Tukey correction of the log transformed data. Fold change is determined in comparison to the transfected control (pAc5.1). 15 cGLRs from different species were selected with p value ≤ 0.05 with fold change ≥ 2.

Supplementary Material

Supplemental Information

Acknowledgements

We thank Joao Marques for critical reading of the manuscript, Hunter Toyoda for help purifying proteins, Jiewei Deng for LC-MS analysis and members of the Kranzusch, Imler and Cai labs for discussions and helpful comments. J.L.I. acknowledges financial support from CNRS, the Hoffmann Infinitus Program and grants from ANR (ANR-11-EQPX-0022, ANR-22-CE15–0019), IdEx Unistra (ANR-10-IDEX-0002), SFRI-STRAT’US (ANR-20-SFRI0012) and EUR IMCBio (ANR-17-EURE-0023). H.C. was supported by the Natural Science Foundation (32000662) , Guangdong Provincial Science Fund for Distinguished Young Scholars (2023B1515020098), Guangdong Provincial Young Scholars academic exchange program (2022A0505030018), Youth Talent Support Programme of Guangdong Provincial Association for Science and Technology (SKXRC202229), Science and Technology Program of Guangzhou (202102020090). D.C. was supported by Natural Science Foundation of China (NSFC, 32200578). R.J. was supported by the National Key R&D Program of China (2021YFA0805800, 2020YFA0803202), the National Natural Science Foundation of China (31970538) and the Local Innovative and Research Teams Project of Guangdong Perl River Talents Program (2017BT01S155). J.L.I., D.C. and R.J. acknowledge support by the Chinese National Overseas Expertise Introduction Center for Discipline Innovation (Project ‘111’ (D18010). P.J.K. acknowledges financial support from the Pew Biomedical Scholars program, the Burroughs Wellcome Fund PATH program, The G. Harold and Leila Y. Mathers Charitable Foundation, The Mark Foundation for Cancer Research, the Cancer Research Institute, the Parker Institute for Cancer Immunotherapy, and the National Institutes of Health (1DP2GM146250–01). K.M.S. is supported as an NCI F99 Graduate Fellow NIH 1F99CA274660–01.

Footnotes

Declaration of interests

The authors declare no competing interests.

References

  • 1.Lemaitre B, Nicolas E, Michaut L, Reichhart JM, and Hoffmann JA (1996). The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell 86, 973–983. 10.1016/s0092-8674(00)80172-5. [DOI] [PubMed] [Google Scholar]
  • 2.Poltorak A, He X, Smirnova I, Liu M-Y, Huffel CV, Du X, Birdwell D, Alejos E, Silva M, Galanos C, et al. (1998). Defective LPS Signaling in C3H/HeJ and C57BL/10ScCr Mice: Mutations in Tlr4 Gene. Science 282, 2085–2088. 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 3.Medzhitov R, Preston-Hurlburt P, and Janeway CA (1997). A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388, 394–397. 10.1038/41131. [DOI] [PubMed] [Google Scholar]
  • 4.Weber ANR, Tauszig-Delamasure S, Hoffmann JA, Lelièvre E, Gascan H, Ray KP, Morse MA, Imler J-L, and Gay NJ (2003). Binding of the Drosophila cytokine Spätzle to Toll is direct and establishes signaling. Nat. Immunol 4, 794–800. 10.1038/ni955. [DOI] [PubMed] [Google Scholar]
  • 5.Michel T, Reichhart JM, Hoffmann JA, and Royet J (2001). Drosophila Toll is activated by Gram-positive bacteria through a circulating peptidoglycan recognition protein. Nature 414, 756–759. 10.1038/414756a. [DOI] [PubMed] [Google Scholar]
  • 6.Gottar M, Gobert V, Matskevich AA, Reichhart J-M, Wang C, Butt TM, Belvin M, Hoffmann JA, and Ferrandon D (2006). Dual detection of fungal infections in Drosophila via recognition of glucans and sensing of virulence factors. Cell 127, 1425–1437. 10.1016/j.cell.2006.10.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gobert V, Gottar M, Matskevich AA, Rutschmann S, Royet J, Belvin M, Hoffmann JA, and Ferrandon D (2003). Dual Activation of the Drosophila Toll Pathway by Two Pattern Recognition Receptors. Science 302, 2126–2130. 10.1126/science.1085432. [DOI] [PubMed] [Google Scholar]
  • 8.Gottar M, Gobert V, Michel T, Belvin M, Duyk G, Hoffmann JA, Ferrandon D, and Royet J (2002). The Drosophila immune response against Gram-negative bacteria is mediated by a peptidoglycan recognition protein. Nature 416, 640–644. 10.1038/nature734. [DOI] [PubMed] [Google Scholar]
  • 9.Rämet M, Manfruelli P, Pearson A, Mathey-Prevot B, and Ezekowitz RAB (2002). Functional genomic analysis of phagocytosis and identification of a Drosophila receptor for E. coli. Nature 416, 644–648. 10.1038/nature735. [DOI] [PubMed] [Google Scholar]
  • 10.Choe K-M, Werner T, Stöven S, Hultmark D, and Anderson KV (2002). Requirement for a peptidoglycan recognition protein (PGRP) in Relish activation and antibacterial immune responses in Drosophila. Science 296, 359–362. 10.1126/science.1070216. [DOI] [PubMed] [Google Scholar]
  • 11.Takehana A, Yano T, Mita S, Kotani A, Oshima Y, and Kurata S (2004). Peptidoglycan recognition protein (PGRP)-LE and PGRP-LC act synergistically in Drosophila immunity. EMBO J 23, 4690–4700. 10.1038/sj.emboj.7600466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Guo Z, Li Y, and Ding S-W (2019). Small RNA-based antimicrobial immunity. Nat. Rev. Immunol. 19, 31–44. 10.1038/s41577-018-0071-x. [DOI] [PubMed] [Google Scholar]
  • 13.Schneider J, and Imler J-L (2020). Sensing and signalling viral infection in drosophila. Dev Comp Immunol 117, 103985. 10.1016/j.dci.2020.103985. [DOI] [PubMed] [Google Scholar]
  • 14.Hua X, Li B, Song L, Hu C, Li X, Wang D, Xiong Y, Zhao P, He H, Xia Q, et al. (2018). Stimulator of interferon genes (STING) provides insect antiviral immunity by promoting Dredd caspase-mediated NF-κB activation. J. Biol. Chem 293, 11878–11890. 10.1074/jbc.RA117.000194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Goto A, Okado K, Martins N, Cai H, Barbier V, Lamiable O, Troxler L, Santiago E, Kuhn L, Paik D, et al. (2018). The Kinase IKKβ Regulates a STING- and NF-κB-Dependent Antiviral Response Pathway in Drosophila. Immunity 49, 225–234.e4. 10.1016/j.immuni.2018.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Segrist E, Dittmar M, Gold B, and Cherry S (2021). Orally acquired cyclic dinucleotides drive dSTING-dependent antiviral immunity in enterocytes. Cell Reports 37, 110150. 10.1016/j.celrep.2021.110150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ablasser A, and Chen ZJ (2019). cGAS in action: Expanding roles in immunity and inflammation. Science 363. 10.1126/science.aat8657. [DOI] [PubMed] [Google Scholar]
  • 18.Sun L, Wu J, Du F, Chen X, and Chen ZJ (2013). Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791. 10.1126/science.1232458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cai H, Holleufer A, Simonsen B, Schneider J, Lemoine A, Gad HH, Huang J, Huang J, Chen D, Peng T, et al. (2020). 2’3’-cGAMP triggers a STING- and NF-κB-dependent broad antiviral response in Drosophila. Sci Signal 13. 10.1126/scisignal.abc4537. [DOI] [PubMed] [Google Scholar]
  • 20.Holleufer A, Winther KG, Gad HH, Ai X, Chen Y, Li L, Wei Z, Deng H, Liu J, Frederiksen NA, et al. (2021). Two cGAS-like receptors induce antiviral immunity in Drosophila. Nature 597, 114–118. 10.1038/s41586-021-03800-z. [DOI] [PubMed] [Google Scholar]
  • 21.Slavik KM, Morehouse BR, Ragucci AE, Zhou W, Ai X, Chen Y, Li L, Wei Z, Bähre H, König M, et al. (2021). cGAS-like receptors sense RNA and control 3’2’-cGAMP signalling in Drosophila. Nature 597, 109–113. 10.1038/s41586-021-03743-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Duncan-Lowey B, and Kranzusch PJ (2022). CBASS phage defense and evolution of antiviral nucleotide signaling. Current Opinion in Immunology 74, 156–163. 10.1016/j.coi.2022.01.002. [DOI] [PubMed] [Google Scholar]
  • 23.Whiteley AT, Eaglesham JB, de Oliveira Mann CC, Morehouse BR, Lowey B, Nieminen EA, Danilchanka O, King DS, Lee ASY, Mekalanos JJ, et al. (2019). Bacterial cGAS-like enzymes synthesize diverse nucleotide signals. Nature 567, 194–199. 10.1038/s41586-019-0953-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cohen D, Melamed S, Millman A, Shulman G, Oppenheimer-Shaanan Y, Kacen A, Doron S, Amitai G, and Sorek R (2019). Cyclic GMP-AMP signalling protects bacteria against viral infection. Nature 574, 691–695. 10.1038/s41586-019-1605-5. [DOI] [PubMed] [Google Scholar]
  • 25.Lowey B, Whiteley AT, Keszei AFA, Morehouse BR, Mathews IT, Antine SP, Cabrera VJ, Kashin D, Niemann P, Jain M, et al. (2020). CBASS Immunity Uses CARF-Related Effectors to Sense 3’-5’- and 2’-5’-Linked Cyclic Oligonucleotide Signals and Protect Bacteria from Phage Infection. Cell 182, 38–49.e17. 10.1016/j.cell.2020.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Morehouse BR, Govande AA, Millman A, Keszei AFA, Lowey B, Ofir G, Shao S, Sorek R, and Kranzusch PJ (2020). STING cyclic dinucleotide sensing originated in bacteria. Nature 586, 429–433. 10.1038/s41586-020-2719-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Margolis SR, Dietzen PA, Hayes BM, Wilson SC, Remick BC, Chou S, and Vance RE (2021). The cyclic dinucleotide 2′3′-cGAMP induces a broad antibacterial and antiviral response in the sea anemone Nematostella vectensis. PNAS 118. 10.1073/pnas.2109022118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Woznica A, Kumar A, Sturge CR, Xing C, King N, and Pfeiffer JK (2021). STING mediates immune responses in the closest living relatives of animals. eLife 10, e70436. 10.7554/eLife.70436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hornung V, Hartmann R, Ablasser A, and Hopfner K-P (2014). OAS proteins and cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. Nat. Rev. Immunol 14, 521–528. 10.1038/nri3719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Annibal A, Ripa R, Ballhysa E, Latza C, Hochhard N, and Antebi A (2021). Mass spectrometric characterization of cyclic dinucleotides (CDNs) in vivo. Anal Bioanal Chem 413, 6457–6468. 10.1007/s00216-021-03628-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, Woo S-R, Lemmens E, Banda T, Leong JJ, et al. (2015). Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Reports 11, 1018–1030. 10.1016/j.celrep.2015.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kranzusch PJ (2019). cGAS and CD-NTase enzymes: structure, mechanism, and evolution. Curr Opin Struct Biol 59, 178–187. 10.1016/j.sbi.2019.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wu J, Sun L, Chen X, Du F, Shi H, Chen C, and Chen ZJ (2013). Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830. 10.1126/science.1229963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Habayeb MS, Cantera R, Casanova G, Ekström J-O, Albright S, and Hultmark D (2009). The Drosophila Nora virus is an enteric virus, transmitted via feces. J. Invertebr. Pathol 101, 29–33. 10.1016/j.jip.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 35.Gao P, Ascano M, Wu Y, Barchet W, Gaffney BL, Zillinger T, Serganov AA, Liu Y, Jones RA, Hartmann G, et al. (2013). Cyclic [G(2′,5′)pA(3′,5′)p] Is the Metazoan Second Messenger Produced by DNA-Activated Cyclic GMP-AMP Synthase. Cell 153, 1094–1107. 10.1016/j.cell.2013.04.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Civril F, Deimling T, de Oliveira Mann CC, Ablasser A, Moldt M, Witte G, Hornung V, and Hopfner K-P (2013). Structural mechanism of cytosolic DNA sensing by cGAS. Nature 498, 332–337. 10.1038/nature12305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou W, Whiteley AT, de Oliveira Mann CC, Morehouse BR, Nowak RP, Fischer ES, Gray NS, Mekalanos JJ, and Kranzusch PJ (2018). Structure of the Human cGAS–DNA Complex Reveals Enhanced Control of Immune Surveillance. Cell 174, 300–311.e11. 10.1016/j.cell.2018.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhang X, Shi H, Wu J, Zhang X, Sun L, Chen C, and Chen ZJ (2013). Cyclic GMP-AMP Containing Mixed Phosphodiester Linkages Is An Endogenous High-Affinity Ligand for STING. Molecular Cell 51, 226–235. 10.1016/j.molcel.2013.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kranzusch PJ, Wilson SC, Lee ASY, Berger JM, Doudna JA, and Vance RE (2015). Ancient Origin of cGAS-STING Reveals Mechanism of Universal 2’,3’ cGAMP Signaling. Mol. Cell 59, 891–903. 10.1016/j.molcel.2015.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Morehouse BR, Yip MCJ, Keszei AFA, McNamara-Bordewick NK, Shao S, and Kranzusch PJ (2022). Cryo-EM structure of an active bacterial TIR–STING filament complex. Nature 608, 803–807. 10.1038/s41586-022-04999-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Li Y, Slavik KM, Toyoda HC, Morehouse BR, Mann C.C. de O., Elek A, Levy S, Wang Z, Mears K, Liu J, Kashin D, Guo X, Mass T, Sebé-Pedros A, Schwede F, and Kranzusch PJ (2023). cGLRs are a diverse family of pattern recognition receptors in innate immunity. Cell 186, 3261–3276. 10.1016/j.cell.2023.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wienken CJ, Baaske P, Rothbauer U, Braun D, and Duhr S (2010). Protein-binding assays in biological liquids using microscale thermophoresis. Nat Commun 1, 100. 10.1038/ncomms1093. [DOI] [PubMed] [Google Scholar]
  • 43.Jerabek-Willemsen M, André T, Wanner R, Roth HM, Duhr S, Baaske P, and Breitsprecher D (2014). MicroScale Thermophoresis: Interaction analysis and beyond. Journal of Molecular Structure 1077, 101–113. 10.1016/j.molstruc.2014.03.009. [DOI] [Google Scholar]
  • 44.Seidel SAI, Dijkman PM, Lea WA, van den Bogaart G, Jerabek-Willemsen M, Lazic A, Joseph JS, Srinivasan P, Baaske P, Simeonov A, et al. (2013). Microscale thermophoresis quantifies biomolecular interactions under previously challenging conditions. Methods 59, 301–315. 10.1016/j.ymeth.2012.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Millman A, Melamed S, Amitai G, and Sorek R (2020). Diversity and classification of cyclic-oligonucleotide-based anti-phage signalling systems. Nat Microbiol 5, 1608–1615. 10.1038/s41564-020-0777-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Govande AA, Duncan-Lowey B, Eaglesham JB, Whiteley AT, and Kranzusch PJ (2021). Molecular basis of CD-NTase nucleotide selection in CBASS anti-phage defense. Cell Reports 35, 109206. 10.1016/j.celrep.2021.109206. [DOI] [PubMed] [Google Scholar]
  • 47.Fatma S, Chakravarti A, Zeng X, and Huang RH (2021). Molecular mechanisms of the CdnG-Cap5 antiphage defense system employing 3′,2′-cGAMP as the second messenger. Nat Commun 12, 6381. 10.1038/s41467-021-26738-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hobbs SJ, Wein T, Lu A, Morehouse BR, Schnabel J, Leavitt A, Yirmiya E, Sorek R, and Kranzusch PJ (2022). Phage anti-CBASS and anti-Pycsar nucleases subvert bacterial immunity. Nature 605, 522–526. 10.1038/s41586-022-04716-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Eaglesham JB, Pan Y, Kupper TS, and Kranzusch PJ (2019). Viral and metazoan poxins are cGAMP-specific nucleases that restrict cGAS-STING signalling. Nature 566, 259–263. 10.1038/s41586-019-0928-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Eaglesham JB, McCarty KL, and Kranzusch PJ (2020). Structures of diverse poxin cGAMP nucleases reveal a widespread role for cGAS-STING evasion in host-pathogen conflict. Elife 9. 10.7554/eLife.59753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.McFarland AP, Luo S, Ahmed-Qadri F, Zuck M, Thayer EF, Goo YA, Hybiske K, Tong L, and Woodward JJ (2017). Sensing of Bacterial Cyclic Dinucleotides by the Oxidoreductase RECON Promotes NF-κB Activation and Shapes a Proinflammatory Antibacterial State. Immunity 46, 433–445. 10.1016/j.immuni.2017.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gui X, Yang H, Li T, Tan X, Shi P, Li M, Du F, and Chen ZJ (2019). Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 567, 262–266. 10.1038/s41586-019-1006-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu Y, Gordesky-Gold B, Leney-Greene M, Weinbren NL, Tudor M, and Cherry S (2018). Inflammation-Induced, STING-Dependent Autophagy Restricts Zika Virus Infection in the Drosophila Brain. Cell Host Microbe 24, 57–68.e3. 10.1016/j.chom.2018.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mosallanejad K, Kennedy SN, Bahleda KM, Slavik KM, Zhou W, Govande AA, Hancks DC, Kranzusch PJ, and Kagan JC (2023). Species-specific self-DNA detection mechanisms by mammalian cyclic GMP-AMP synthases. Science Immunology 8, eabp9765. 10.1126/sciimmunol.abp9765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Burroughs AM, Zhang D, Schäffer DE, Iyer LM, and Aravind L (2015). Comparative genomic analyses reveal a vast, novel network of nucleotide-centric systems in biological conflicts, immunity and signaling. Nucleic Acids Research 43, 10633–10654. 10.1093/nar/gkv1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Katoh K, Rozewicki J, and Yamada KD (2019). MAFFT online service: multiple sequence alignment, interactive sequence choice and visualization. Briefings in Bioinformatics 20, 1160–1166. 10.1093/bib/bbx108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kelley LA, Mezulis S, Yates CM, Wass MN, and Sternberg MJE (2015). The Phyre2 web portal for protein modeling, prediction and analysis. Nat Protoc 10, 845–858. 10.1038/nprot.2015.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, Tunyasuvunakool K, Bates R, Žídek A, Potapenko A, et al. (2021). Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589. 10.1038/s41586-021-03819-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Letunic I, and Bork P (2021). Interactive Tree Of Life (iTOL) v5: an online tool for phylogenetic tree display and annotation. Nucleic Acids Research 49, W293–W296. 10.1093/nar/gkab301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chen S, Zhou Y, Chen Y, and Gu J (2018). fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890. 10.1093/bioinformatics/bty560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim D, Langmead B, and Salzberg SL (2015). HISAT: a fast spliced aligner with low memory requirements. Nat Methods 12, 357–360. 10.1038/nmeth.3317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Love MI, Huber W, and Anders S (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 1–21. 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chen T, Chen X, Zhang S, Zhu J, Tang B, Wang A, Dong L, Zhang Z, Yu C, Sun Y, et al. (2021). The Genome Sequence Archive Family: Toward Explosive Data Growth and Diverse Data Types. Genomics, Proteomics & Bioinformatics 19, 578–583. 10.1016/j.gpb.2021.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Members CNCB-NGDC and Partners (2022). Database Resources of the National Genomics Data Center, China National Center for Bioinformation in 2022. Nucleic Acids Research 50, D27–D38. 10.1093/nar/gkab951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Liebschner D, Afonine PV, Baker ML, Bunkóczi G, Chen VB, Croll TI, Hintze B, Hung L-W, Jain S, McCoy AJ, et al. (2019). Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Cryst D 75, 861–877. 10.1107/S2059798319011471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Emsley P, and Cowtan K (2004). Coot: model-building tools for molecular graphics. Acta Cryst D 60, 2126–2132. 10.1107/S0907444904019158. [DOI] [PubMed] [Google Scholar]
  • 67.Magnez R, Thiroux B, Taront S, Segaoula Z, Quesnel B, and Thuru X (2017). PD-1/PD-L1 binding studies using microscale thermophoresis. Sci Rep 7, 17623. 10.1038/s41598-017-17963-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liberelle M, Magnez R, Thuru X, Bencheikh Y, Ravez S, Quenon C, Drucbert A-S, Foulon C, Melnyk P, Van Seuningen I, et al. (2019). MUC4-ErbB2 Oncogenic Complex: Binding studies using Microscale Thermophoresis. Sci Rep 9, 16678. 10.1038/s41598-019-53099-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Casarotto PC, Girych M, Fred SM, Kovaleva V, Moliner R, Enkavi G, Biojone C, Cannarozzo C, Sahu MP, Kaurinkoski K, et al. (2021). Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell 184, 1299–1313.e19. 10.1016/j.cell.2021.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Information

Data Availability Statement

  • The RNAseq data for D. melanogaster and D. serrata are available in the Genome Sequence Archive, National Genomics Data Center, China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (https://ngdc.cncb.ac.cn/gsa) and are publicly available from the date of publication. . Accession numbers are listed in the Key Resources Table. Raw data are accessible at Mendeley Data repository (https://data.mendeley.com/) (DOI: 10.17632/kcgwhnghvx.1).

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
     
     
     
     
     
Bacterial and virus strains
E. coli BL21-DE3 RIL Agilent Cat# 230245
E. coli TOP 10 Invitrogen Cat# C404006
Drosophila C virus Goto et al., 201815 N/A
Vesicular Stomatitis Virus Goto et al., 201815 N/A
Nora virus This study N/A
Biological samples    
     
     
     
     
     
Chemicals, peptides, and recombinant proteins
Ni-NTA Agarose Qiagen Cat# 30250
HiTrap Heparin HP column Cytiva Cat# 17040701
Zorbax Bonus-RP Agilent Cat# 863668–901
Alkaline Phosphatase, Quick CIP New England Biolabs Cat# M0525S
Nuclease P1 from Penicillium citrinum Sigma-Aldrich Cat# N8630
PEI-Cellulose F TLC plate EMD Biosciences Cat# EM1.05579.0001
ATP, [α-32P], 3000 Ci/mmol 10 mCi/ml Perkin Elmer Cat# BLU003H250UC
GTP, [α-32P], 3000 Ci/mmol 10 mCi/ml Perkin Elmer Cat# BLU006H250UC
ATP, GTP New England Biolabs Cat# N0450S
2′3′-c-di-GMP Biolog Life Science Institute Cat# C 182
Poly(I:C) (HMW) VacciGrade Invivogen Cat# vac-pic
2'3'-c-di-AMP Biolog Life Science Institute Cat# C 187
3′2′-cGAMP Biolog Life Science Institute Cat# C 238
2′3′-cGAMP Biolog Life Science Institute Cat# C 161
3′3′-c-di-GMP Biolog Life Science Institute Cat# C 057
Nucleodur Pyramid C18 column (3 μm, 50 x 3 mm) Macherey Nagel Cat# 760263.30
ZORBAX RRHD StableBond Aq column (1.8um, 2.1 x 100mm) Agilent Technologies Cat# 858700–914
     
Critical commercial assays
     
     
     
     
     
Deposited data
RNA-seq data for CDN-injected flies Genome Sequence Archive, National Genomics Data Center, China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (https://ngdc.cncb.ac.cn/gsa) Accession # CRA011428
Raw data Mendeley Data repository (https://data.mendeley.com/) DOI: 10.17632/kcgwhnghvx.1
     
     
     
Experimental models : Cell lines
Drosophila S2 cells Akira Goto et al., 201815 N/A
Drosophila STING-KO S2 cells This study N/A
HEK293T cells ATCC Cat# CRL-3216
     
     
Experimental models : Organisms/strains
DrosDel w 1118 Hua Cai et al., 202019 N/A
dSTING L76GfsTer11 Hua Cai et al., 202019 N/A
dSTINGRxn, Akira Goto et al., 201815 N/A
dSTING Control Akira Goto et al., 201815 N/A
iso : cGLR1 -/- ,dSTING -/- This study N/A
iso : cGLR2 -/- ,dSTING -/- This study N/A
iso : cGLR1+2 -/- ,dSTING -/- This study N/A
UAS-cGLR1 Andreas Holleufer et al., 202120 N/A
UAS-cGLR1 AFA Andreas Holleufer et al., 202120 N/A
UAS-cGLR2 Andreas Holleufer et al., 20215 N/A
UAS-cGLR2 AFA Andreas Holleufer et al., 202120 N/A
Act-GAL4/CyO ; TubG80[ts] IBMC N/A
D.willistoni Peking University, Laboratory of Jian Lu N/A
D.yakuba Peking University, Laboratory of Jian Lu N/A
D.simulans Peking University, Laboratory of Jian Lu N/A
D.virilis Peking University, Laboratory of Jian Lu N/A
D.takahashii Peking University, Laboratory of Jian Lu N/A
D.suzukii Chinese Academy of Sciences, Laboratory of Binyan Lu N/A
D.hydei Chinese Academy of Sciences, Laboratory of Binyan Lu N/A
D.serrata South China Agricultural University, Laboratory of Shuoyang Wen N/A
D.kikkawai South China Agricultural University, Laboratory of Shuoyang Wen N/A
D.pseudoobscura Chinese Academy of Sciences, Laboratory of Wei Wu N/A
D.mojavensis Southeast University, Laboratory of Yufeng Pan N/A
D.sechellia Lüdwig Maximilian University, Laboratory of Nicolas Gompel N/A
D.santomea Lüdwig Maximilian University, Laboratory of Nicolas Gompel N/A
     
Oligonucleotides
Oligonucleotide primers for qPCR, See Table S6 This study N/A
     
     
Recombinant DNA
pAC-Actin5C-Renilla luciferase E. Santiago, CNRS, Strasbourg, France N/A
pGL3/-200bp-dSTING promoter Akira Goto et al., 201815 N/A
pAC5.1 vector ThermoFischer Cat# V411020
pAC5.1/GFP IBMC N/A
pAC5.1-Dmel-STING This study N/A
pAC5.1-Dsim-STING This study N/A
pAC5.1-Dwil-STING IGE biotechnology N/A
pAC5.1-Dyak-STING IGE biotechnology N/A
pAC5.1-Dmoj-STING IGE biotechnology N/A
pAC5.1-Dvir-STING IGE biotechnology N/A
pAC5.1-Dhyd-STING IGE biotechnology N/A
pAC5.1-Dsan-STING IGE biotechnology N/A
pAC5.1-Dsec-STING IGE biotechnology N/A
pAC5.1-Dtak-STING IGE biotechnology N/A
pAC5.1-Dpse-STING IGE biotechnology N/A
pAC5.1-Dsuz-STING IGE biotechnology N/A
pAC5.1-Dser-STING IGE biotechnology N/A
pAC5.1-Dkik-STING IGE biotechnology N/A
pAC5.1-Dhyd-LOC111595620 This study N/A
pAC5.1-Dhyd-LOC111605403 This study N/A
pAC5.1-Dhyd-LOC111592833 This study N/A
pAC5.1-Dhyd-LOC111601518 This study N/A
pAC5.1-Dmoj-LOC6579490 This study N/A
pAC5.1-Dmoj-LOC6582322 This study N/A
pAC5.1-Dmoj-LOC6574034 This study N/A
pAC5.1-Dmoj-LOC6580632 This study N/A
pAC5.1-Dvir-LOC6625054 This study N/A
pAC5.1-Dvir-LOC6625472 This study N/A
pAC5.1-Dwil-LOC6638395 This study N/A
pAC5.1-Dwil-LOC6646488 This study N/A
pAC5.1-Dwil-LOC6638458 This study N/A
pAC5.1-Dpse-LOC6899115 This study N/A
pAC5.1-Dpse-LOC6900849 This study N/A
pAC5.1-Dpse-LOC6900403 This study N/A
pAC5.1-Dpse-LOC4803562 This study N/A
pAC5.1-Dtak-LOC108054184 This study N/A
pAC5.1-Dtak-LOC108066267 This study N/A
pAC5.1-Dtak-LOC108054321 This study N/A
pAC5.1-Dtak-LOC108067387 This study N/A
pAC5.1-Dtak-LOC108055687 This study N/A
pAC5.1-Dsuz-LOC108018091 This study N/A
pAC5.1-Dsuz-LOC108012585 This study N/A
pAC5.1-Dsuz-LOC108013198 This study N/A
pAC5.1-Dsuz-LOC108006881 This study N/A
pAC5.1-Dsuz-LOC108006833 This study N/A
pAC5.1-Dsuz-LOC108009543 This study N/A
pAC5.1-Dsuz-LOC108008397 This study N/A
pAC5.1-Dsec-LOC6609335 This study N/A
pAC5.1-Dsec-LOC116801260 This study N/A
pAC5.1-Dsim-LOC6734663 This study N/A
pAC5.1-Dsim-LOC6737253 This study N/A
pAC5.1-Dmel-cGLR1 This study N/A
pAC5.1-Dmel-cGLR2 This study N/A
pAC5.1-Dyak-LOC6531519 This study N/A
pAC5.1-Dyak-LOC6532544 This study N/A
pAC5.1-Dyak-LOC6530290 This study N/A
pAC5.1-Dyak-LOC6532149 This study N/A
pAC5.1-Dsan-LOC120447203 This study N/A
pAC5.1-Dsan-LOC120447932 This study N/A
pAC5.1-Dsan-LOC120445453 This study N/A
pAC5.1-Dser-LOC110178317 This study N/A
pAC5.1-Dser-LOC110191302 This study N/A
pAC5.1-Dser-LOC110179302 This study N/A
pAC5.1-Dser-LOC110184043 This study N/A
pAC5.1-Dkik-LOC108082553 This study N/A
pAC5.1-Dkik-LOC108071893 This study N/A
pAC5.1-Dkik-LOC108080761 This study N/A
pAC5.1-Dkik-LOC108078272 This study N/A
pAC5.1-Dkik-LOC108078001 This study N/A
pAC5.1-Dkik-LOC108072236 This study N/A
pAC5.1-Dmel-sting-eGFP This study N/A
pAC5.1-Dser-sting-eGFP This study N/A
pcDNA3.1-Dmel-cGLR2 IGE biotechnology N/A
     
     
     
     
     
     
     
Software and algorithms
MAFFT Katoh and Yamada56 https://mafft.cbrc.jp/alignment/software/
Geneious Prime (v2022.1.1) Biomatters, Ltd https://www.geneious.com/
iTOL Letunic and Bork59 https://itol.embl.de/
Phenix 1.13–2998 Liebschner et al.65 https://www.phenixonline.org/
Coot 0.8.9 Emsley and Cowtan66 https://www2.mrclmb.cam.ac.uk/personal/pemsley/coot/
PyMOL (v 2.4.2) Schrödinger, LLC https://pymol.org/
Xcalibur 2.2 Thermo Fisher Scientific, San Jose, CA, USA https://www.thermofisher.cn/order/catalog/product/OPTON-30967
Agilent MassHunter Acquisition (ver. B.08.00) Agilent Technologies, Santa Clara, CA, USA https://www.agilent.com.cn/zh-cn/product/software-informatics/mass-spectrometry-software/data-analysis
MO.Affinity Analysis(version 2.3) NanoTemper Technologies https://nanotempertech.com
R studio (version 4.1.2) The R project http://www.rstudio.com/products/rstudio
Coin package http://coin.r-forge.r-project.org/ https://cran.r-project.org/web/packages/coin/index.html
GraphPad Prism9 (version 9.2.0) GraphPad Software, LLC www.graphpad.com
Other
     
     
     
     

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Cell lines

HEK293T female cells (ATCC) were cultured in Dulbecco’s modified Eagle medium (DMEM) (Sigma-Aldrich) supplemented with 10% fetal bovine serum (FBS) (Excell Bio), 100 U mL−1 penicillin (Sigma-Aldrich) and 100 µg mL−1 streptomycin (Sigma-Aldrich) at 37°C and 5% CO2. Schneider 2 (S2) male cells (Invivogen) were cultured in Schneider’s Drosophila Medium (Thermal Fisher Scientific) supplemented with 10% FBS, 100 U mL−1 penicillin (Sigma-Aldrich) and 100 µg mL−1 streptomycin (Sigma-Aldrich) at 27°C. To generate Sting knockout cells by CRISPR-Cas9, three CRISPR RNAs targeting the first translated exon in the D. melanogaster Sting gene were cloned into the pAc-sgRNA-Casp vector20. After transfection, S2 cells were grown under selection with 5 µg mL−1 puromycin (Invitrogen) for two weeks. Knockout of Sting was verified by Sanger sequencing.

Drosophila strains

Fly stocks were raised on standard cornmeal agar medium at 25°C and were free of Wolbachia. All fly stocks used (DrosDel w1118, dSTINGL76GfsTer11, dSTINGRxn, dSTINGControl, cGLR1 or cGLR2 single knockout or double knockout flies, the transgenic lines of wild-type and AFA mutant versions of cGLR1 and cGLR2, and the GFP control) have been described previously15,19,20. The different species of Drosophila were kindly provided by Prof. Jian Lu, Peking University (D.willistoni, D.yakuba, D.simulans, D.virilis and D.takahashii); Prof. Binyan Lu, Chinese Academy of Sciences (D.suzukii and D. hydei); Prof. Shuoyang Wen, South China Agricultural University (D.serrata and D.kikkawai); Prof. Wei Wu, Chinese Academy of Sciences (D.pseudoobscura); Prof. Yufeng Pan, Southeast University (D.mojavensis) and Prof. Nicolas Gompel, Lüdwig Maximilian University (D.sechellia and D.santomea). A 50:50 mix of male and female adult flies collected three- to five-days post ecclosion was used in experiments, unless stated otherwise in the figure legends.

METHOD DETAILS

Plasmids

Coding sequences of predicted cGLRs were PCR-amplified from the cDNA prepared from flies from the different Drosophila species using 2×EasyTaq PCR SuperMix (TransGen Biotech, China) and cloned into the pAc5.1 vector using ClonExpress MultiS (Vazyme, China). The coding sequences of STING from fourteen species of Drosophila were synthesized by the IGE biotechnology company (China) and sub-cloned to pAc5.1 vector.

Antiviral effect of cyclic di-nucleotides in fourteen species of Drosophila

Cyclic dinucleotides including 3′2′-cGAMP (Biolog C238), 2′3′-cGAMP (Biolog C161), 2′3′-c-di-AMP (Biolog C187), 2′3′-c-di-GMP (Biolog C182) or 3′3′-c-di-GMP (Biolog C057) were dissolved in 10 mM Tris-HCl pH 7.5 and diluted to the indicated concentrations. 3–5 days old male flies were injected with 69 nL of cyclic dinucleotide solution or 10 mM Tris-HCl pH 7.5 (negative control) by intrathoracic injection using a Nanoject II apparatus (Drummond Scientific). 7 days post CDN injection, the flies were injected with 4.6 nL of DCV (50 PFU for all the tested species except D. melanogaster and D. yakuba, for which the dose was 5 PFU) in 10 mM Tris-HCl pH 7.5. Flies were collected 48 h and 72 h later in pools of 6 males and homogenized for RNA extraction and RT-qPCR analysis, as described.

Fly hemolymph and lysate preparation for Liquid Chromatography Mass Spectrometry (LC-MS)

For hemolymph collection, anesthesized adult flies were punctured using a tungsten needle and transferred to a 0.6 mL microcentrifuge tube (12 flies/tube) with a hole pierced in the bottom. The tube with flies was then transferred to a 1.5 mL Eppendorf tube. The two-tube assembly was then centrifuged for 10 min at 2,300 g at 4°C. The procedure was repeated until the hemolymph of one thousand flies was collected. Hemolymph was stored at −80°C.

For lysate preparation, 500 adult flies were collected in five 2 mL microfuge tubes (100 flies/ tube). 10 small zirconia beads were added per tube together with 100 μL precooled extraction reagent (2/2/1 [v/v/v] methanol, acetonitrile, water mixture) with 10 ng mL−1 3′3′-cAIMP as an internal standard in the LC-MS/MS analysis. The tubes with flies were frozen in liquid nitrogen and then homogenized using Precellys Evolution homogenizer (4°C, 5800 rpm, 2*30 s pause 12 s). The tubes were then centrifuged for 10 min at 12,000 g at 4°C. The supernatant was collected into a new microfuge tube, which was centrifuged again. The collected supernatant was transferred into 2 mL safe-lock tubes and heated at 95°C for 10 min, before cooling on ice. 500 µL chloroform was then added to the tubes, which were vortexed and placed at −20°C for 10 min. Subsequently, the tubes were centrifuged at 20,000 x g for 15 min, and the supernatants was transferred into fresh microfuge tubes. Next, the lysate was loaded on HLB SPE columns. The eluents were collected and then concentrated by evaporation and resuspended in 200 μL 0.1% formic acid water for LC-MS/MS analyses.

Identification of CDN using LC-MS

To detect the CDNs production in the lysate of DCV infected flies, high resolution LC-MS analysis was performed using a Ultimate3000 (ThermoFisher Scientific) coupled to a Quadrupole-Orbitrap Hybrid mass spectrometer (Q-Exactive, ThermoFisher Scientific). A volume of 5 μL sample was injected into a ZORBAX RRHD StableBond Aq column (1.8 μm, 2.1 x 100 mm; Agilent Technologie) maintained at 40°C. The mobile phase consisted of 5 mM ammonium carbonate (A) and acetonitrile (B). The following HPLC gradient was used: 0–14% B in 5.0 min, 14–25% B in 7.0 min, 25–100% B in 7.1 min, 100% B in 10.8 min, 100–0% B in 11.0 min, 0% B in 14.0 min; 0.300 mL min−1. Mass spectra were recorded using positive ion full scan mode with m/z from 300 to 1100. Accurate mass measurement was accomplished by Orbitrap-MS with a mass resolution of 70,000. The optimized MS parameters were set as follows: capillary temperature 350°C, maximum inject time 100ms, AGC target of 1.00E+06, and S-lens RF level 55. The target ions were sequentially isolated using high energy collision dissociation (HCD) fragmentation and progeny ions were detected with dd-MS2 mode. The parent ion was isolated with an isolation window of 2 m/z units, fragmented (Resolution = 17,500, nce=20, Maximum Inject Time: 50ms, Loop count: 5, TopN: 5). Identification of CDNs was performed by targeted mass analysis for exact masses and formulae for all possible CDNs. Xcalibur 2.2 (Thermo Fisher Scientific, San Jose, CA, USA) software was used for equipment control and data acquisition.

Measurement of CDN in the hemolymph or lysate

For measurement of 2'3'-cGAMP, 3'2'-cGAMP, 2'3'-c-di-AMP and 2'3'-c-di-GMP in the hemolymph, samples were analyzed using an Agilent 1290 System coupled to an Agilent 6470. A volume of 5 μL was injected into a Nucleodur Pyramid C18 column (3 μm, 50 x 3 mm; Macherey Nagel, Duren, Germany) maintained at 40°C. The mobile phase consisted of 0.1% formic acid in acetonitrile (A) and 0.1% formic acid in water (B). HPLC gradient are described in Table S1. The analytes were ionized by means of electro spray ionization in positive mode with the Delta EMV at 820 V. The source parameters are described in Table S2. For each CDN, the MRM transition(s) (m/z), Dwell, Frag (V) and CE (V) Cell Acc (V) are described in Table S3.

For measurement of 2'3'-cGAMP, 3'2'-cGAMP, 2'3'-c-di-AMP, 2'3'-c-di-GMP in whole fly lysates, the samples were analyzed using an Agilent 1290 System coupled to an Agilent 6470. A volume of 5 μL was injected into a Nucleodur Pyramid C18 column (50 x 3 mm; 3 μm Macherey Nagel, Duren, Germany) maintained at 40°C. The mobile phase consisted of 0.1% formic acid in acetonitrile (A) and 0.1% formic acid in water (B). The HPLC gradient and flow rate are described in Table S4. The analytes were ionized by means of electro spray ionization in positive mode with the Delta EMV at 820 V. The source parameters were described in Table S2. For each CDN, the MRM transition(s) (m/z), Dwell, Frag (V) and CE (V) Cell Acc (V) were described in Table S5.

LC-MS/MS analysis of in vitro cGLR2 reaction products

cGLR2 reaction products were generated in a 100 μL reaction with 5 μM cGLR enzyme, 200 μM GTP, 10 μg poly I:C, 1 mM MnCl2, and 50 mM Tris-HCl pH 7.5 and were analyzed by the commercial company MS-Omics using LC-MS/MS, as previously described41. Briefly, analysis was carried out using a Vanquish Horizon UHPLC System coupled to Orbitrap Exploris 240 Mass Spectrometer (Thermo Fisher Scientific, US).

First, UHPLC was performed using an Infinity Lab PoroShell 120 HILIC-Z PEEK lined column with the dimension of 2.1 × 150mm and particle size of 2.7 µm (Agilent Technologies). Mobile phase A was composed of 10 mM ammonium acetate, pH 9 in 90% Acetonitrile LC-MS grade (VWR Chemicals, Leuven) and 10% Ultra-pure water from Direct-Q® 3 UV Water Purification System with LC-Pak® Polisher (Merck KGaA, Darmstadt). Mobile phase B was composed of 10 mM ammonium acetate, pH 9 in ultra-pure water with 5 µM medronic acid (InfinityLab Deactivator additive, Agilent Technologies). The UHPLC column temperature was set at 30 °C and samples were analyzed at an injection volume of 5 µl. UHPLC was run using a flow rate kept at 250 µl mL−1 consisting of a 2 min hold at 10% B, increased to 40% B at 14 min, held till 15 min, decreased to 10% B at 16 min and held for 8 min.

For MS analysis, a heated electrospray ionization interface was used as ionization source and the analysis was performed in positive ionization mode from m/z 300 to 1500 at a mass resolution of 120000. Ion source parameters used: Sheath gas flow rate, 20 (arbitrary units); auxiliary gas flow rate, 5 (arbitrary units); Sweep gas flow rate, 1 (arbitrary units), capillary temperature, 350°C; S-lens radiofrequency level 70; automatic gain control (AGC) target, 1E6 (Standard); maximum injection time, 100 ms; spray voltage 3.5 kV in positive. MS2 spectra was acquired using data dependent acquisition (DDA) with the following parameters: mass resolution 45000, isolation window m/z 0.4 and normalized collision energy 20, 40 and 60 eV. Freestyle 1.4 (Thermo Fisher Scientific) was used to analyze data and generate MS/MS spectra.

CDN Stability Assay

Sf9 cell lysates were prepared from S. frugiperda Sf9 cells (Expression Systems) cultured in ESF 921 media. Pelleted cells were washed with 1× PBS, resuspended in 250 µL of a lysis buffer containing 20 mM HEPES-KOH pH 7.5, 150 mM NaCl, 5 mM MgCl2, 1 mM MnCl2, 1 mM DTT, 10% glycerol, and 1% NP-40, and incubated at room temperature for 30 min with occasional vortexing. Drosophila lysates were prepared in a similar manner from adult male flies of each respective species (55–120, depending on their sizes) using the same lysis buffer and grinding in Cryolis tubes. The total protein content of lysate samples was determined by Bradford assay and each sample was adjusted to 7.5 µg/µL. α-32P-labeled CDNs were synthesized by the following cGLR enzymes: 2′3′-cGAMP, Tc-cGLR; 3′2′-cGAMP and 2′3′-c-di-AMP, Ds-cGLR1; 2′3′-c-di-GMP, Db-cGLR2, as described above. 15 µL reactions containing 2.5 µM CDN and 1.5 µL Sf9 or Drosophila lysate in a buffer containing 50 mM HEPES-KOH 7.5, 10 mM KCl, 5 mM MgCl2, 1 mM MnCl2, and 1 mM TCEP were incubated at 37°C overnight and analyzed by TLC to determine CDN stability.

D. melanogaster cyclic dinucleotide injection and signaling analysis

Cyclic dinucleotides including 2'3'-c-di-GMP (Biolog C182), 2'3'-c-di-AMP (Biolog C187), 3′2′-cGAMP (Biolog C238), 2′3′-cGAMP (Biolog C161) and 3′3′-c-di-GMP (Biolog C057) were dissolved in 10 mM Tris-HCl pH 7.5 and diluted to the indicated concentrations. 3–5-day old adult flies were injected with 69 nL of cyclic dinucleotide solution or 10 mM Tris-HCl pH 7.5 (negative control) by intrathoracic injection using a Nanoject II apparatus (Drummond Scientific). Flies were collected 7 days later in pools of 6 individuals (3 males and 3 females) and homogenized for RNA extraction and quantitative PCR with reverse transcription (RT–qPCR) analysis, as described.

Bioinformatics and Drosophila cGLR and STING sequence analysis

Building on previous analyses21,23,41,55, Drosophila cGLRs were identified using the amino acid sequence of D. melanogaster cGLR1 (NP_788360.2) to seed a position-specific iterative BLAST (PSI-BLAST) search of Drosophila genomes (taxid:7215) in the NCBI non-redundant protein database. The PSI-BLAST search was performed with an E value cutoff 0.005 for inclusion into the next search round, BLOSUM62 scoring matrix, gap costs settings existence 11 and extension 1, and using conditional compositional score matrix adjustment. Iterative PSI-BLAST search was performed for 5 rounds and candidate Drosophila cGLR sequences were collected. Putative cGLR protein sequences were aligned using MAFFT (FFT-NS-i iterative refinement method)56; this alignment was used to construct a phylogenetic tree in Geneious Prime v2022.0.1 using the neighbor-joining method and Jukes-Cantor genetic distance model with no outgroup. Candidate proteins were analyzed by clade and selected for known cGLR domain organization and predicted structural homology to T. castaneum cGLR (PBD: 7LT2), including the presence of a conserved nucleotidyltransferase domain with a G[S/G] activation loop and [E/D]h[E/D] X50–90 [E/D] catalytic triad32. Manual analysis and curation of candidate cGLR sequences was performed based on alignments and predictive structural homology using Phyre257 and AlphaFold58. Manual refinement was also used to exclude duplicate sequences, gene isoforms, and proteins less than 250 residues. All cGLR sequences in the final tree were accessed from NCBI March 26th, 2023. The D. bipectinata cGLR2 sequence shown in Figure S5A,B and used for all biochemistry experiments was accessed from NCBI in January, 2020 under accession code XP_017096409.1. NCBI available genomes from 49 species in the Drosophila genus are represented in the final tree. iTOL was used for tree visualization and annotation59, including annotation of all cGLRs identified in fourteen Drosophila species of interest to this study. Isoelectric point was predicted by Geneious Prime software. Clustering of sequences in the final unrooted tree was used to define clades related to D. melanogaster cGLR1 (NP_788360.2), cGLR2 (A8DYP7.2), and cGLR3 (CG7194; AAF50449.1). The cGLR2-related clade was identified by the presence of D. melanogaster cGLR2 (A8DYP7.2) and extracted from the full cGLR tree for alignment using MAFFT (FFT-NS-i iterative refinement method), shown in Figure S5A. cGLR2 sequences of interest in Figure S5B were separately aligned and the secondary structure of D. pseudoananassae cGLR2 was generated based on the AlphaFold predicted structure.

Drosophila STING proteins were identified as a subset of eukaryotic STING proteins identified in ref.41. STING sequences were accessed from NCBI May 2022. Drosophila STING protein sequences from fourteen species of interest were aligned using MAFFT (FFT-NS-i iterative refinement method)56. Alignment in Figure S7F is shown in relation to the secondary structure of the D. eugracilis−3′2′-cGAMP complex (PDB: 7MWZ)21.

Screening of cGLRs based on cellular STING signaling assays

To test the activation of STING promoter luciferase reporter by predicted cGLRs, 96-well tissue culture plates were seeded with 2.5 x 105 S2 cells per well. After 3h, each well was transfected with 200ng pGL3 plasmid expressing firefly luciferase under transcriptional control of the Sting promoter, 25ng pAc5.1 plasmid constitutively expressing Renilla luciferase, 75ng pAc5.1 plasmid expressing predicted cGLRs, 25ng pAc5.1 plasmid expressing Sting, finally empty Ac5.1 plasmid to reach a total amount of 325ng plasmid for each well. All transfections of S2 cells were performed using lipofectamine 3000 (Invitrogen, Thermo Fisher Scientific) according to the manufacturer’s instructions. After 48 hours of transfection, cells were lysed in 100µL Lysis Buffer (Promega) per well. Firefly and Renilla luciferase activity was measured on 75µL lysate using the Dual-Luciferase® Reporter Assay System (Promega).

RNA-Sequencing of D. melanogaster injected with CDNs

Male flies of D. melanogaster or D. serrata were injected with 69 nL/fly of either 10 mM Tris (pH 7.5), 2'3'-cGAMP (0.9 mg mL−1) or 2'3'-cdi-GMP (0.9 mg mL−1) by intrathoracic injection (Nanoject II apparatus) in three independent experiments. Injected flies were collected in pools of 6 individuals at 7 days post injection. Total RNA was isolated from injected flies using TRIzol Reagent (Invitrogen), according to the manufacturer's protocol. RNA quality was assessed on an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA, USA) and checked using RNase free agarose gel electrophoresis. After total RNA was extracted, eukaryotic mRNA was enriched by Oligo(dT) beads. Then the enriched mRNA was fragmented into short fragments using fragmentation buffer and reverse transcribed into cDNA by using NEBNext Ultra RNA Library Prep Kit for Illumina (NEB #7530, New England Biolabs, Ipswich, MA, USA). The purified double-stranded cDNA fragments were end repaired, A base added, and ligated to Illumina sequencing adapters. The ligation reaction was purified with the AMPure XP Beads (1.0X). Ligated fragments were subjected to size selection by agarose gel electrophoresis and polymerase chain reaction (PCR) amplified. The resulting cDNA library was sequenced using Illumina Novaseq6000 by Gene Denovo Biotechnology Co. (Guangzhou, China).

Transcriptome analysis

After filtering by fastp60 (version 0.18.0), reads were mapped using HISAT2. 2.461 with “-rna-strandness RF” and other parameters set as a default to the genome of D. melanogaster (Ensembl_release102) and D. serrata (GCF_002093755.1). RNAs differential expression analysis was performed by DESeq262 software between two different groups (and by edgeR between two samples). The genes/transcripts with the parameter of false discovery rate (FDR) below 0.05 and absolute fold change≥2 were considered differentially expressed genes/transcripts. The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive (Genomics, Proteomics & Bioinformatics 2021) in National Genomics Data Center (Nucleic Acids Res 2022), China National Center for Bioinformation / Beijing Institute of Genomics, Chinese Academy of Sciences (GSA: CRA011428) that are publicly accessible at https://ngdc.cncb.ac.cn/gsa63,64.

Protein expression and purification

Recombinant cGLR proteins were expressed and purified using methods previously optimized for human cGAS and Drosophila cGLRs, as described previously21,37. Full length cGLR coding sequences were codon-optimized for expression in E. coli and cloned from synthetic constructs (GeneArt or Integrated DNA Technologies) into a custom pET16 expression vector with an N-terminal 6×His-MBP fusion tag. Briefly, transformed BL21-CodonPlus (DE3)-RIL E. coli (Agilent) were grown in MDG media overnight prior to inoculation of M9ZB media at an OD600 of 0.0475. M9ZB cultures were grown to OD600 of 2.5 (approximately 5 h at 37°C with shaking at 230 rpm) followed by cooling on ice for 20 min. Cultures were induced with 500 μM IPTG prior to incubation at 16°C overnight with shaking at 230 rpm. Cultures were pelleted the following day and either flash frozen in liquid nitrogen for storage at −80°C or directly lysed for purification.

For large-scale protein purification, proteins were expressed with a 6×His-MBP fusion tag and grown as ~4–8× 1 L cultures in M9ZB media. Pellets were lysed by sonication in lysis buffer (20 mM HEPES pH 7.5, 400 mM NaCl, 30 mM imidazole, 10% glycerol and 1 mM DTT) and clarified by centrifugation at ~47,850 × g for 30 min at 4°C and subsequent filtration through glass wool. Recombinant protein was purified by gravity-flow over NiNTA resin (Qiagen). Resin was washed with lysis buffer supplemented to 1 M NaCl and then eluted with 20 mL of lysis buffer supplemented to 300 mM imidazole. MBP-tagged fusion proteins were buffer exchanged into lysis buffer with 4% glycerol and no imidazole to optimize conditions for overnight cleavage by recombinant TEV protease at ~10°C. cGLR proteins were next purified by ion exchange chromatography using 5 mL HiTrap Heparin HP columns (GE Healthcare) and eluted across a 150–1000 mM NaCl gradient in buffer with 10% glycerol. Target protein fractions were pooled concentrated to ~10–30 mg mL−1 and flash-frozen with liquid nitrogen and stored at −80°C for biochemistry experiments.

Nucleotide product synthesis analysis

cGLR nucleotide synthesis activity was analyzed by thin-layer chromatography as previously described21,23. For all biochemistry reactions analyzed by TLC, 5 μM recombinant protein preparations were incubated in 5 or 10 μL reactions containing 0.5 μL α-32P labeled ATP or GTP, 200 μM unlabeled NTPs, and 1 mM MnCl2 in a final reaction buffer of 50 mM Tris-HCl pH 7.5, ~50 mM KCl (final KCl = 100 mM), 1 mM TCEP. Reactions were additionally supplemented with ~1 μg poly I:C or 5 μM nucleic acid ligands, as indicated. Besides poly I:C all nucleic acids used in this study are 40 nucleotide (nt) or base pairs (bp) in length. Reactions were incubated at 37°C for two hours and subsequently treated with 1 μL Quick CIP phosphatase (New England Biolabs) for 20 min at 37°C to remove unreacted phosphate signal. 0.5 μL of each reaction was spotted on a 20-cm × 20-cm PEI-cellulose thin-layer chromatography plate. Plates were run with 1.5 M KH2PO4 solvent until ~2.5 cm from top of the plate, dried at room-temperature, and exposed to a phosphor-screen prior to signal detection with a Typhoon Trio Variable Mode Imager System (GE Healthcare). TLC images were adjusted for contrast using FIJI40 and quantified using ImageQuant (8.2.0). GTP conversion to nucleotide product formation was measured according to the ratio of product to total signal for each reaction.

Nuclease P1 cleavage analysis was performed using cGLR2 reactions labeled with either α-32P-ATP or α-32P-GTP as previously described21,23. Briefly, radiolabeled nucleotide products were incubated with Nuclease P1 (80 mU, Sigma N8630) in buffer (30 mM NaOAc pH 5.3, 5 mM ZnSO4, 50 mM NaCl) for 30 min in the presence of Quick CIP (NEB).

Nucleotide purification and HPLC analysis

Enzymatic synthesis of cGLR nucleotide products for HPLC analysis was performed using 100 μL reactions containing 5 μM cGLR enzyme, 100 μM ATP, 100 μM GTP, 10 μg poly I:C, 1 mM MnCl2, and 50 mM Tris-HCl pH 7.5. KCl was adjusted to a final concentration of 100 mM. Reactions were incubated at 37°C for 2 h and then terminated by 2 min incubation at 95°C. Nucleotide product was recovered by filtering reactions through a 30-kDa cutoff concentrator (Amicon) to remove protein. Nucleotide products were separated on an Agilent 1200 Infinity Series LC system using a C18 column (Zorbax Bonus-RP 4.6 × 150 mm, 3.5 μm) at 40°C. Products were eluted at a flow rate of 1 mL min−1 with a buffer of 50 mM NaH2PO4 pH 6.8 supplemented with 3% acetonitrile.

Synthetic cyclic dinucleotide standards

Synthetic nucleotide standards used for HPLC analysis and mass-spectrometry analysis were purchased from Biolog Life Science Institute: 2′3′-cGAMP (cat no. C 161), 3′2′-cGAMP (cat no. C 238), 2′3′-c-di-AMP (cat no. C 187) and 2′3′-c-di-GMP (cat no. C 182).

dSTING and 2′3′-c-di-GMP structural modeling

Coordinate and cif restraint files for 2′3′-c-di-GMP were generated by the eLBOW program on PHENIX65 using the ChemDraw v.20.0.38 generated SMILES for this ligand. The resulting files were used to model the 2′3′-c-d-GMP ligand in complex with D. eugracilis STING by fitting to the 3′2′-cGAMP ligand density in the dSTING3′2′-cGAMP complex density map (PDB 7MWZ) using Coot66.

MicroScale thermophoresis measurement

S2 cells seeded in 10 cm plates at a density of 10 million cells per plate and transfected with 10μg of plasmid DNA via calcium chloride for three days. The cells were harvested by centrifuging them in a 15 mL tube at 135g and 4°C for 5 minutes. The supernatant was discarded, and the cell pellet was resuspended in 5 mL of PBS and centrifuged again at for 5 minutes. 300μL of RIPA buffer (1% NP-40, 0.5% deoxycholate, 0.1% SDS) containing protease inhibitors was added to the cells and the samples were incubated on ice with gentle agitation for 15 minutes. The samples were then centrifuged at 21,000g, 4°C for 45 minutes. The supernatant was carefully collected in a fresh Eppendorf tube and stored at 4°C. The protein lysate was adjusted to 400 fluorescence units (FI units) after a pretest. Dilution was performed using PBS-T buffer (1X PBS containing 0.05% Tween 20). Measurements using a Monolith NT.115 (NanoTemper Technologies) were performed at 25°C using 90% Excitation power and Medium MST power. Data analyses were performed using MO.Affinity Analysis software6769.

Statistical analysis

All statistical analyses were performed in R (version 4.1.2) and GraphPad Prism9. To perform the qPCR analysis, 2∆Ct values calculated for each sample were tested by permutation test using coin package. In addition, survival analyses using Log-rank tests were performed to compare each condition to control lines. cGLRs from different species were analysed using mixed-effect model (RELM) with Tukey correction of the log transformed data. Fold change is determined in comparison to the transfected control (pAc5.1). 15 cGLRs from different species were selected with p value ≤ 0.05 with fold change ≥ 2.

RESOURCES