Significance
The immune system strikes a careful balance between launching a robust response to threats and avoiding overactivation. The molecule cGAMP is an immunotransmitter that activates innate immunity and signals extracellularly, where it is subject to degradation by the enzyme ENPP1. Here, we engineer ENPP1 to lose activity toward cGAMP but not other substrates, thus creating a biochemically precise tool to understand how ENPP1 regulates extracellular cGAMP and thus innate immunity. We uncover that ENPP1's degradation of extracellular cGAMP has a long evolutionary history, and that this mechanism is critical for controlling diverse immune threats, including viral infection and inflammation.
Keywords: cGAMP, ENPP1, STING, extracellular cGAMP, immunotransmitter
Abstract
The metazoan innate immune second messenger 2′3′-cGAMP is present both inside and outside cells. However, only extracellular cGAMP can be negatively regulated by the extracellular hydrolase ENPP1. Here, we determine whether ENPP1’s regulation of extracellular cGAMP is a ubiquitous mechanism of attenuating stimulator of interferon genes (STING) signaling. We identified ENPP1H362A, a point mutation that cannot degrade the 2′-5′ linkage in cGAMP while maintaining otherwise normal function. The selectivity of this histidine is conserved down to bacterial nucleotide pyrophosphatase/phosphodiesterase (NPP), allowing structural analysis and suggesting an unexplored ancient history of 2′-5′ cyclic dinucleotides. Enpp1H362A mice demonstrated that extracellular cGAMP is not responsible for the devastating phenotype in ENPP1-null humans and mice but is responsible for antiviral immunity and systemic inflammation. Our data define extracellular cGAMP as a pivotal STING activator, identify an evolutionarily critical role for ENPP1 in regulating inflammation, and suggest a therapeutic strategy for viral and inflammatory conditions by manipulating ENPP1 activity.
Since its discovery 9 y ago, 2′3′-cyclic-GMP-AMP (cGAMP) quickly became the intense focus of many biochemistry, immunology, and cell biology laboratories (1–3). Its significance was immediately apparent, being a new addition to the relatively small group of known second messengers. cGAMP is synthesized by the enzyme cyclic-GMP-AMP synthase (cGAS) in response to self and pathogenic double-stranded DNA in the cytosol. cGAMP then binds its endoplasmic reticulum membrane-localized receptor stimulator of interferon genes (STING) to activate downstream transcription of interferon (IFN) and other cytokines, triggering powerful antiviral (1, 4) and anticancer (5–9) defense mechanisms. However, it also triggers devastating inflammation in neurodegenerative diseases (10, 11), myocardial infarction (12), and autoimmune diseases (13–16) when not properly regulated.
cGAMP’s regulation depends on its cellular localization. cGAMP was originally thought to function as an intracellular signal that is synthesized by cGAS and sensed by STING within the same cell. cGAMP was subsequently found to spread to neighboring cells through gap junctions (17–19) and exosomes (20, 21), both of which do not permit access to the extracellular space. The prevailing view that cGAMP is exclusively an intracellular signal was challenged by the recent discovery that cGAMP is secreted into the extracellular space by cancer cells and functions as a paracrine immunotransmitter by entering and activating host immune cells (19, 22–26). Intracellular cGAMP signaling is primarily regulated at the level of STING activation, which requires the formation of a large signaling complex (27–29) that has a high activation threshold but is irreversible once formed (27), explaining its role in uncontrolled inflammation. Extracellular cGAMP signaling is controlled by many additional molecular and cellular mechanisms. cGAMP is transported by cell-type–specific transporters (25, 26, 30–33), allowing for cell-specific secretion and uptake. Additionally, extracellular cGAMP is degraded by the extracellular hydrolase ENPP1, while no intracellular cGAMP hydrolase has been reported (22, 34).
The studies that established cancer-to-host cGAMP immunotransmission were enabled by experimental techniques that are only possible in cancer models, including genetic manipulation of cancer cells (19, 22, 23, 26) and local extracellular cGAMP depletion through intratumoral injections of a depleting agent (22, 25). ENPP1 was subsequently shown to be a negative regulator of anticancer innate immunity, enabling the development of ENPP1 inhibitors (patents WO2020160333A1, WO2019023635A1, WO2018119328A1) as investigative new drugs for cancer therapy. Whether ENPP1-regulated extracellular cGAMP signaling is a ubiquitous mechanism of STING signaling remains a major unsolved question, with the potential to impact therapies targeting ENPP1 in broad disease contexts.
Results
Mutations of Guanosine-Adjacent Residues in ENPP1 Do Not Inhibit cGAMP Hydrolysis.
To understand the role that extracellular cGAMP plays as an immunotransmitter in diseases beyond cancer, we sought to develop a universal tool that can systemically manipulate extracellular cGAMP levels. We hypothesized that the cGAMP hydrolase ENPP1 was a promising candidate to build such a tool, since ENPP1 degrades extracellular but not intracellular cGAMP (22). However, ENPP1 also degrades extracellular ATP to AMP and pyrophosphate (PPi), the latter of which is critical in regulating calcium homeostasis (35). Organisms lacking ENPP1 exhibit severe systemic calcification and a significantly shortened lifespan (36–38). In addition, extracellular ATP and its downstream degradation products AMP and adenosine are all immunomodulatory molecules. Therefore, we sought to identify substrate-selective ENPP1 mutations to specifically study the role of extracellular cGAMP.
We first compared the cocrystal structures of mouse ENPP1 bound to AMP (39) and pApG, the intermediate after the first phosphodiester bond cleavage of cGAMP (40). ENPP1 has a tight nucleotide-binding site, which the AMP portions of ATP and cGAMP occupy, and a secondary site, which the guanine base of cGAMP occupies (guanosine-adjacent site) (Fig. 1 A and B). Between the two sites, a cluster of residues chelate two zinc ions, which position the α-phosphate of the substrates for nucleophilic attack by the catalytic residue T238. We validated our previously developed enzymatic assay using ENPP1WT and ENPP1T238A (SI Appendix, Fig. S1A) as positive and negative controls for both substrates. We generated whole-cell lysates by overexpressing FLAG-tagged ENPP1 variants in 293T ENPP1−/− cells (SI Appendix, Fig. S1B) as a fast and robust source of mutant enzymes. [32P]-cGAMP degradation was monitored using thin-layer chromatography separation and autoradiography, and ATP degradation was monitored using a luciferase-based assay. Using these assays, we confirmed that ENPP1WT is able to degrade both ATP and cGAMP, while ENPP1T238A is not (SI Appendix, Fig. S1 C and D).
Inactivating the nucleotide-binding site is known to prevent both cGAMP (41) and ATP hydrolysis (39) (SI Appendix, Fig. S1E). Therefore, we sought to identify residues that are required for cGAMP but not ATP hydrolysis by individually mutating 10 residues with sidechains within 5 Å of the guanine base of cGAMP (Fig. 1C and SI Appendix, Fig. S1 F and G) and measuring their initial rates of substrate hydrolysis (Fig. 1D and SI Appendix, Fig. S1 H–R). Surprisingly, most of these mutations had only a modest effect on cGAMP degradation. ATP activity usually tracked with cGAMP activity, suggesting that these mutations led to general destabilization. Mutation of N259, a highly conserved residue that forms a hydrogen bond with the nonbridging phosphoryl oxygen of the substrates (42, 43), inhibited activity for both substrates. The K237A mutation achieved the opposite of our goal by abolishing ATP degradation but preserving cGAMP degradation. It is possible that K237 is important for stabilizing the β- and γ-phosphates of ATP (44), but is too far away from the phosphodiester of cGAMP to affect catalysis.
Discovery and Characterization of ENPP1H362A, a Mutation That Degrades ATP but Not cGAMP.
We next made alanine mutations of the six aspartate and histidine zinc-binding residues that comprise the catalytic core of ENPP1 (Fig. 1 E and F and SI Appendix, Fig. S2A). All of these mutations were inactive toward both substrates except for the H362A mutation (ENPP1H362A). Remarkably, this mutation recapitulated ENPP1WT activity toward ATP but had no detectable activity toward cGAMP (Fig. 1G and SI Appendix, Fig. S2 B–J). We then mutated H362 to the other 18 amino acids, all of which were inactive toward cGAMP, indicating that histidine is essential at that position for cGAMP degradation (Fig. 1H and SI Appendix, Fig. S3 A–E). Finally, the purified ENPP1H362A enzyme also had no detectable cGAMP hydrolysis activity (Fig. 1I and SI Appendix, Fig. S3F) but had identical kcat/Km values to ENPP1WT for ATP hydrolysis (Fig. 1J). As expected, ENPP1H362A was also fully active toward the other NTP substrates of ENPP1 (GTP, UTP, and CTP) (SI Appendix, Fig. S3G).
Bacterial NPP Selectively Cleaves 2′-5′ Linkages in Cyclic Dinucleotides Using the Conserved Histidine.
Since ENPP1 is part of the highly conserved nucleotide pyrophosphatase/phosphodiesterase (NPP) protein family, we investigated the evolutionary conservation of the analogous histidine residue and its requirement for cGAMP degradation. This histidine is 100% conserved in the 998 eukaryotic species and 99.9% conserved in the 1,000 bacterial species that we investigated (Fig. 2A and SI Appendix, Fig. S4A). We then selected a small pool of eukaryotic and prokaryotic NPP sequences to investigate their cellular localization. All the sequences we investigated had a predicted signal peptide (SI Appendix, Fig. S4B), suggesting extracellular localization in eukaryotes or periplasmic localization in bacteria. However, the physiological substrates and the role of NPP in bacteria remain unknown. One hypothesis is that bacterial NPP in the periplasm hydrolyzes nucleotide triphosphates, such as ATP, to salvage the base products (45). We therefore tested the ability of bacterial Xanthomonas axonopodis pv. citri (Xac) NPP (42, 46) to degrade ATP. We found that Xac NPPWT degrades ATP and that Xac NPPH214A (which corresponds to the mouse H362A mutation) degrades ATP several-fold faster than NPPWT (Fig. 2B), suggesting that H214 is not conserved for the purpose of scavenging base products from ATP.
We then tested the hypothesis that the conserved histidine is important for cleaving cyclic dinucleotides. Indeed, Xac NPPWT degrades the metazoan 2′3′-cGAMP while the NPPH214A mutant does not, mirroring our findings with mouse ENPP1H362A (Fig. 2C). A different 2′-5′–linked cGAMP, 3′2′-cGAMP, was recently discovered as a second messenger in a bacterial antiviral system (47). We found that Xac NPPWT readily degrades 3′2′-cGAMP (Fig. 2D). In addition, Xac NPPWT degrades other synthetic cyclic dinucleotides with 2′-5′/3′-5′ mixed linkages much faster than their counterparts with two 3′-5′ linkages (Fig. 2 E and F and SI Appendix, Fig. S4 C–F). Interestingly, H214 is required to cleave cyclic dinucleotides with 2′-5′ linkages (2′3′-cGAMP, 3′2′-cGAMP, 2′3′-CDA, 2′3′-CDG), but is not required for cyclic dinucleotides with only 3′-5′ linkages (3′3′-cGAMP, 3′3-CDA, or 3′3-CDG) (Fig. 2 D–F and SI Appendix, Fig. S4 G–I). This substrate preference and requirement for the remarkably conserved histidine suggest that bacterial NPP’s canonical substrates may be 2′-5′–linked cyclic dinucleotides or oligonucleotides, including the recently discovered 3′2′-cGAMP (48).
Structure-Guided Elucidation of the Substrate-Selective Degradation Mechanism of ENPP1H362A.
We then sought to use structural analysis to explain the mechanism of the exquisite substrate selectivity of Xac NPPH214A and mouse ENPP1H362A toward nucleotide triphosphates and 3′-5′ phosphodiester bonds, but not 2′-5′ phosphodiester bonds. As mouse ENPP1 is technically challenging to crystallize, we took advantage of the conservation between Xac NPP and mouse ENPP1 to solve the cocrystal structure of Xac NPP in complex with cGAMP. To prevent cGAMP hydrolysis, we mutated the catalytic threonine to alanine (Xac NPP T90A), crystalized it with cGAMP, and obtained a 1.9 Å crystal structure (SI Appendix, Tables S1 and S2). Similar to the previous mouse ENPP1 T238A structure (40), we found a linear 3′-5′–linked pApG intermediate bound to NPP instead of intact cGAMP (Fig. 3A). In the previous structure, the guanine ring was within stacking distance of the conserved histidine (∼4.5 Å). However, the guanine ring in our structure is ∼8 Å away from the histidine and rotated 140° from the previous structure (Fig. 3B). This discrepancy suggests that the guanine ring is flexible and mobile, which explains why mutating the guanosine-adjacent site did not disrupt cGAMP hydrolysis. Using cGAMP as a competitive inhibitor of the ENPP1-ATP reaction, we determined that ENPP1WT and ENPP1H362A have very similar Ki values, demonstrating that ENPP1H362A binds cGAMP equally well as ENPP1WT (Fig. 3C). Together, our structural and biochemical data demonstrate that H362 is not necessary for binding cGAMP.
Two zinc ions are required for NPP’s catalytic activity, with Zn1 being particularly important for stabilizing the leaving group, whereas Zn2 activates the threonine nucleophile (42, 49). Since the histidine (H362 in mouse ENPP1 and H214 in Xac NPP) is one of three residues that chelates Zn1, we hypothesized that the loss of the histidine prevents Zn1 binding. Indeed, in contrast to Xac NPPWT, which binds two zincs, our 2.0 Å crystal structure revealed that Xac NPPH214A was missing Zn1 (Fig. 3 D and E). We then followed up this structural insight with biochemical experiments using mouse ENPP1. Increasing the zinc concentration from 1 μM to 10 μM sped up the rate of ATP hydrolysis for ENPP1H362A, but had no effect on ENPP1WT, suggesting that the missing histidine lowers the affinity for Zn1 (Fig. 3F). However, we were unable to rescue cGAMP activity even with 1 mM zinc, which is 100-fold above physiological conditions (10 to 20 μM) (Fig. 3G).
To explain this discrepancy in how the two substrates are differentially impacted by zinc concentrations, we hypothesize that ATP is able to replace the histidine to chelate Zn1, as ATP in solution is usually chelated to metals (50–52) and binds to zinc with a Kd of ∼10 to 100 μM, depending on the solution conditions (52). We tested this hypothesis by modeling zinc-bound ATP in mouse ENPP1H362A (Fig. 3H). In our model, the α-, β-, and γ-phosphates of ATP all chelated Zn1 in conjunction with H517 and D200. In fact, the γ-phosphate oxygen was in a very similar position as the H362 nitrogen that coordinates Zn1 in ENPP1WT (Fig. 3I). We also docked 3′3′-cGAMP and 2′3′-cGAMP into our H214A Xac NPP structure. Similar to ATP, the free 2′-OH group in 3′3′-cGAMP can also take the place of the missing histidine to chelate Zn1 while the free 3′-OH group in 2′3′-cGAMP is too far away from Zn1 (Fig. 3 J and K). We propose that ATP and 3′-5′–linked cyclic dinucleotides are able to compensate for the loss of histidine with their γ-phosphate oxygen and 2′-OH group, respectively, explaining why H362 in mouse ENPP1 and H214 in Xac NPP are only required for cleaving 2′-5′–linked phosphodiester bonds.
Enpp1H362A Mice Can Degrade ATP but Not cGAMP.
We next created homozygous Enpp1H362A mice using CRISPR-based homologous recombination (SI Appendix, Fig. S5 A and B). After verifying the genotype of the mice (SI Appendix, Fig. S5C), we sought to confirm that these mice exhibited the expected enzymatic activity: intact ATP hydrolysis but defective cGAMP hydrolysis. We compared tissue lysates from Enpp1WT and Enpp1H362A mice to those from Enpp1asj mice, which harbor a point mutation that abolishes all ENPP1 activity (36). While tissue lysates from Enpp1WT mice rapidly degraded [32P]-cGAMP, tissue lysates from Enpp1H362A mice did not, mirroring the deficiency seen in Enpp1asj mice (Fig. 4A). In addition to the ENPP1 tethered to the surface of cells, ENPP1 is also secreted into the circulation, allowing cGAMP degradation to occur in the plasma (53, 54). Plasma from Enpp1WT mice readily degraded cGAMP, while plasma from Enpp1H362A mice did not (Fig. 4B). Despite the inability to degrade cGAMP, basal tissue cGAMP and Ifnb1 transcript levels were generally low and not significantly different between Enpp1WT, Enpp1H362A, and Enpp1asj mice (SI Appendix, Fig. S5 D and E), suggesting that basal cGAMP signaling is not influenced by ENPP1. Finally, we assessed cGAMP degradation in vivo by subcutaneously injecting cGAMP into each mouse and measuring cGAMP concentration in the plasma by mass spectrometry. Thirty minutes after injection, cGAMP levels in the plasma of Enpp1H362A and Enpp1asj mice were >100-fold higher than in Enpp1WT mice (Fig. 4C), demonstrating that cGAMP hydrolysis is severely impaired in Enpp1H362A mice.
We then assessed ATP activity in these mice using liver lysates. The liver was the only organ suitable for these assays, as it expresses high levels of ENPP1, while ENPP1-independent ATP degradation predominates in other organs, precluding them from being used in this assay. Liver lysates from Enpp1asj mice had a marked deficiency in ATP degradation, but lysates from Enpp1H362A mice degraded ATP as well as those from Enpp1WT mice, confirming that tissue ENPP1H362A retains the ability to degrade ATP (Fig. 4D and SI Appendix, Fig. S5F). Taking these data together, we find that Enpp1H362A mice are unable to degrade cGAMP but retain the ability to degrade ATP.
Enpp1H362A Mice Do Not Exhibit the Severe Systemic Calcification Seen in ENPP1-Null Humans and Mice.
Inactivating mutations in ENPP1 cause progressive calcification of joints, vasculature, and soft tissue, leading to premature death in both mice (36) and humans; the human disease is known as generalized arterial calcification of infancy (37, 38, 55). It is hypothesized that this lethal aberrant calcification is due to the inability to degrade extracellular ATP, leading to a deficiency in PPi, which is known to regulate mineralization (55). However, since cGAMP was only recently identified as a substrate of ENPP1, it is unknown if impaired cGAMP metabolism also contributes to the systemic calcification observed in humans and mice.
As cGAMP degradation is uncoupled from extracellular ATP degradation in Enpp1H362A mice, they are an ideal model for determining the role of extracellular cGAMP in the progressive calcification seen in Enpp1asj mice. Enpp1H362A mice have a normal lifespan and readily breed, unlike the Enpp1asj mice, which have difficulty breeding beyond 2 to 3 mo of age due to worsening arthritis. Furthermore, Enpp1H362A mice do not exhibit the gross joint calcification or tissue calcification seen in Enpp1asj mice (Fig. 4 E and F). Taken together, these results suggest that extracellular cGAMP does not play a significant role in the phenotype seen in Enpp1asj mice and that this phenotype is likely due to the disruption of extracellular ATP metabolism.
Enpp1asj mice are reported to have normal plasma calcium and phosphate levels, while their plasma PPi levels are significantly decreased due to their inability to degrade ATP (36). Like Enpp1WT and Enpp1asj mice, Enpp1H362A mice had normal plasma calcium and phosphate levels (Fig. 4 G and H). However, while Enpp1WT mice had a normal level of plasma PPi (1.5 μM), Enpp1H362A and Enpp1asj mice had 300 nM and 60 nM plasma PPi, respectively (Fig. 4I). This was an unexpected finding, as Enpp1H362A liver lysate did not exhibit a defect in ATP degradation (Fig. 4D), raising the possibility that plasma ENPP1H362A does not exhibit the same behavior as tissue ENPP1H362A. Indeed, Enpp1H362A mouse plasma was defective in ATP degradation, similar to Enpp1asj (Fig. 4J and SI Appendix, Fig. S5G). To test if this discrepancy was due to differences in plasma and tissue ENPP1H362A, we used our in vitro activity assay to compare secreted ENPP1 in cell supernatants with cell-surface ENPP1 present in cell lysates. Unlike the cell-surface ENPP1H362A, the secreted ENPP1H362A was defective in ATP degradation (Fig. 4 K and L and SI Appendix, Fig. S5 H and I). Although we currently lack the ability to measure extracellular PPi levels in tissues, we surmise that tissue PPi is normal in Enpp1H362A mice and that tissue PPi, rather than plasma PPi, is important for preventing tissue calcification. Supporting this hypothesis, a recent study found that osteoblast-specific loss of ENPP1 led to increased bone volume, decreased trabecular spacing, and increased matrix calcification, without any change to plasma PPi (56).
Enhanced Extracellular cGAMP Signaling Confers Resistance to Herpes Simplex Virus-1 Infection.
Extracellular cGAMP has not been implicated in STING-related diseases outside of cancer. Since ENPP1 only regulates the extracellular cGAMP branch of STING signaling, it is unclear if the Enpp1H362A mice would be more resistant to viral infection. In contrast, there is a substantial body of evidence linking extracellular ATP signaling to viral infection (57, 58), including infection by the DNA virus herpes simplex virus (HSV)-1 (59). Therefore, we sought to investigate the relative contributions of extracellular ATP and cGAMP to the antiviral immune response by infecting Enpp1WT, Enpp1H362A, and Enpp1asj mice with a sublethal dose of HSV-1 using an established protocol (4).
Compared to Enpp1WT mice, Enpp1H362A mice were more resistant to HSV-1 infection. Enpp1H362A mice did not experience the weight loss observed in Enpp1WT mice over the course of infection (Fig. 5A). Enpp1H362A mice also had lower levels of the viral transcript HSV-gB in the spleen, liver, and lung at 6 and 12 h postinfection (hpi) (Fig. 5B). This was accompanied by lower levels of replicating virus in Enpp1H362A spleen and kidney lysates (SI Appendix, Fig. S6A). In general, Enpp1H362A mice exhibited decreased expression of Ifnb1 and the downstream cytokines Il6 and Cxcl10 in the liver, lung, and spleen, as well as decreased Tnfa in the liver (Fig. 5 C and D and SI Appendix, Fig. S6 B and C) at 6 and 12 hpi. As an exception, liver Ifnb1 (Fig. 5C) and Il6 (Fig. 5D) were higher in Enpp1H362A mice at 6 hpi. We hypothesize that the much higher levels of HSV in the liver compared to other organs (Fig. 5B) led to a slower decline in liver Ifnb1 expression, allowing us to capture the time point when Enpp1H362A mice mounted a more robust immune response than Enpp1WT mice.
We hypothesize that the lower cytokine expression in Enpp1H362A mice at 12 hpi was due to faster viral clearance as a result of a more robust early immune response. It is also possible that as a plasma membrane protein, ENPP1H362A reduced viral load by preventing or slowing viral entry. To distinguish between these two models, we first measured gene expression of mice at 6 d postinfection and observed no active infection in either genotype, suggesting that the infection was not simply delayed in Enpp1H362A mice (SI Appendix, Fig. S6 D–H). We then infected bone marrow-derived macrophages (BMDMs) isolated from Enpp1WT and Enpp1H362A mice to evaluate for any cell-intrinsic defects in viral entry or replication. Enpp1WT and Enpp1H362A BMDMs were both readily infected by HSV-1 (Fig. 5E) and produced Ifnb1 (Fig. 5F). Resistance to infection and Ifnb1 induction depended on STING in both genotypes (Fig. 5 E and F). These data indicate that the increased resistance to infection and decreased cytokine production observed in Enpp1H362A mice are not due to impaired viral entry or replication in Enpp1H362A cells.
Interestingly, Enpp1asj mice did not significantly differ from Enpp1WT mice in their response to HSV-1 infection (SI Appendix, Fig. S7 A–F), except for a decrease in the expression of Ifnb1 in the lung (SI Appendix, Fig. S7B) and Il6 in the liver (SI Appendix, Fig. S7C). It is possible that impaired ATP metabolism or the overall poor health of Enpp1asj mice compromises the antiviral effect of extracellular cGAMP-STING signaling.
Because Enpp1H362A mice are deficient in plasma ATP hydrolysis, we measured plasma ATP before and after infection to rule out the possibility that increased plasma ATP contributes to controlling viral infection by activating purinergic signaling. We observed generally low (below the Kd toward purinergic receptors) and not significantly different levels of plasma ATP in Enpp1H362A and Enpp1WT mice (SI Appendix, Fig. S7G). These data demonstrate that enhanced extracellular cGAMP alone is protective against HSV-1 infection, and that its hydrolysis by ENPP1 normally dampens this protective signal. Interestingly, we did not detect cGAMP in the plasma of mice infected with HSV-1, with an assay detection limit of 85 pg/mL (125 pM) (SI Appendix, Fig. S7H). The lack of circulating cGAMP suggests that extracellular cGAMP is acting locally as a paracrine immunotransmitter to confer resistance to HSV-1.
Enhanced Extracellular cGAMP Signaling Exacerbates Radiation-Induced Inflammation.
As extracellular cGAMP enhances both the antiviral and anticancer immune responses (22) and ENPP1 negatively regulates extracellular cGAMP, it begs the question: why is the ability of ENPP1 to hydrolyze cGAMP conserved throughout evolution? We hypothesized that paracrine extracellular cGAMP is a pivotal STING activator in extensive or sustained DNA damage and that ENPP1 plays an indispensable role in preventing the subsequent hyperactive and potentially damaging immune response. To test this hypothesis, we used total-body ionizing radiation exposure in our Enpp1H362A mice and Enpp1WT mice to generate systemic DNA damage and inflammation (5, 6, 9, 22). After 8 to 9 Gy of total body irradiation, the Enpp1WT and Enpp1H362A mice were weighed daily and euthanized when they reached the humane endpoint of greater than 20% weight loss for 2 consecutive days. Strikingly, the Enpp1H362A mice exhibited significantly shortened survival compared to Enpp1WT mice (Fig. 6A and SI Appendix, Fig. S8). This was accompanied by induction of cytokines and transcription factors associated with STING activation, including increased plasma IFN-β (Fig. 6B), increased splenic expression of Ifnb1 (Fig. 6C), and increased splenic expression of the downstream transcription factor Irf7 (Fig. 6D). To confirm that the enhanced radiation toxicity in Enpp1H362A mice is STING-dependent, we compared the response to radiation in Sting1−/−/Enpp1H362A and Sting1−/− mice. Without an intact STING pathway, these mice showed no difference in survival or cytokine production in response to radiation (Fig. 6 E–H), suggesting that the enhanced radiation toxicity in these mice is STING-dependent.
Discussion
In this study we identified and characterized ENPP1H362A, a single histidine mutation in ENPP1 that shows remarkable substrate selectivity: it abolishes 2′3′-cGAMP degradation but leaves activity toward ENPP1’s other substrates intact. Remarkably, this histidine is nearly 100% conserved throughout evolutionary history down to bacteria. Previously, the only link between cGAS-STING and NPP was in higher mammals, where ENPP1 degrades extracellular cGAMP to dampen STING activation. We found that Xac NPP not only degrades cyclic dinucleotides but also prefers 2′-5′–linked over 3′-5′–linked cyclic dinucleotides. Furthermore, the conserved histidine is required for degrading the 2′-5′ but not 3′-5′ phosphodiester linkage. Although 3′-5′–linked bacterial cyclic dinucleotides are ubiquitous signaling molecules that are important for cell cycle regulation, biofilm formation, motility, and virulence (60, 61), 2’-5′–linked cyclic dinucleotides were thought to be unique to metazoans until the recent discoveries of bacterial 2′-5′–linked cyclic oligonucleotides (47) and 3′2′-cGAMP (48). We predict that 2′-5′–linked cyclic dinucleotides and oligonucleotides are a general bacterial antiviral defense mechanism and that NPP is a negative regulator of these systems. We propose that mutations of the conserved histidine can be introduced into a wide variety of eukaryotic and prokaryotic organisms to study the pathophysiology of extracellular second messengers with 2′-5′ linkages.
Although previous work has linked cGAMP-STING signaling to defense against HSV-1 infection (62), the role of extracellular cGAMP in anti–HSV-1 defense has only been tested using mice harboring a genetic deletion of the LRRC8 channels (33). The LRRC8 channels transport chloride, aspartate, glutamate, and other organic osmolytes in addition to cGAMP (31, 33). As disrupting the transport of chloride and amino acids can significantly impact the immune response to viral infection, we used the Enpp1H362A mouse strain to specifically investigate the role of extracellular cGAMP in antiviral defense. We found that Enpp1H362A mice are significantly more resistant to HSV-1 infection, definitively demonstrating that extracellular cGAMP plays a key role in viral defense.
Given its negative roles in the antiviral and anticancer immune responses, it is puzzling that the cGAMP hydrolysis activity of the NPP family is so well-conserved. Our discovery of ENPP1’s role in controlling the damage caused by systemic STING overactivation sheds light on this question. The loss of ENPP1’s regulation of extracellular cGAMP significantly increased cytokine production in response to ionizing radiation, resulting in drastically decreased overall survival. These results suggest that extracellular cGAMP exacerbates STING-mediated radiation toxicity and that multicellular organisms rely on ENPP1 to control systemic inflammation. We hypothesize that extracellular cGAMP also contributes to the pathology of many other STING-mediated inflammatory conditions, including autoimmunity (13–16), neurodegeneration (10, 11), myocardial infarction (12), and acute pancreatitis (63). Therefore, ENPP1 may be a vital anti-inflammatory innate immune checkpoint, explaining its remarkable evolutionary conservation (Fig. 6I). Future studies using our Enpp1H362A mice are warranted to assess the involvement of extracellular cGAMP in these conditions and to evaluate the role of ENPP1 as an innate immune checkpoint. Additionally, the regulation of ENPP1 expression and activity in chronic diseases remains an open question.
As we expand our knowledge of the importance of extracellular cGAMP signaling to new diseases and physiological settings, the ability to regulate extracellular cGAMP will provide new therapeutic opportunities to manipulate and treat these diseases. ENPP1 inhibitors, and perhaps even cGAMP-specific ENPP1 inhibitors, may be therapeutically beneficial during viral infection, while administering recombinant ENPP1 or blocking cGAMP import may reduce extracellular cGAMP-associated inflammation.
Methods
See SI Appendix for detailed methods.
Cell Culture.
HEK 293T ENPP1−/− cells were generated in a previous study (22). Vero and Expi293F cell lines were gifts from Peter Kim, Stanford University, Stanford, CA. The 293T ENPP1−/− and Vero cell lines were maintained in a 5% CO2 incubator at 37 °C in DMEM (Corning Cellgro) supplemented with 10% FBS (Atlanta Biologics) and 100 U mL−1 penicillin-streptomycin (Thermo Fisher). The Expi293F cell line was maintained in an 8% CO2 incubator at 37 °C shaking in baffled flasks, in a mixture of 66% FreeStyle Expression Media (Thermo Fisher) and 33% Expi293 Expression Media (Thermo Fisher). All cell lines tested negative for mycoplasma contamination.
Recombinant DNA.
The pcDNA3-mouseENPP1-FLAG plasmid was synthesized by Genscript. The Xac NPP-MBP-pMAL plasmid was a gift from Daniel Herschlag, Stanford University, Stanford, CA (46). The single-point mutations were introduced using QuikChange mutagenesis (Agilent) and verified by sequencing the region of the mutation. Primers used for mutagenesis are shown in SI Appendix, Table S3.
Enzyme Activity Assays.
All enzyme activity assays were performed with protein source indicated, including cell lysate, recombinant protein, organ lysate, or plasma.
Recombinant Mouse ENPP1 and Xac NPP Purification.
His-tagged mouse ENPP1 was expressed in Expi293F cells and purified by cobalt-based immobilized metal ion affinity chromatography (IMAC). His-tagged Xac NPP was expressed in Escherichia coli BL21(DE3) cells and purified by cobalt-based IMAC and anion-exchange chromatography.
NPP Family Sequence Alignment and Signal Peptide Prediction.
Representative species were selected from classic model organisms and the EMBL-EBI protein family (Phosphodiest PF01663). Multiple sequence alignment was performed using MAFFT and visualized using Jalview. Protein accession numbers are listed in SI Appendix, Table S4. To determine the histidine conservation in all known NPP sequences, 998 eukaryotic, 1,000 bacterial, and 584 archaeal NPP protein sequences were downloaded from Uniprot and pairwise-aligned using MUSCLE alignment (64). The histidine corresponding to H362 in mouse ENPP1 was identified and the percent conservation was determined for Eukaryota, Bacteria, and Archaea. Signal peptides were predicted using SignalP-5.0 and transmembrane domains were predicted using TMHMM (DTU Bioinformatics).
Crystallization of Xac NPPT90A with pApG and Xac NPPH214A Apo.
Xac NPPT90A and Xac NPPH214A were expressed, purified, and crystallized as described previously (42).
Mouse Models.
C57BL/6J (Stock #000664), C57BL/6J-Enpp1asj/GrsrJ (Stock #012810), and C57BL/6J-Sting1gt/J (Stock #017537) mice were purchased from the Jackson Laboratory. Enpp1H362A were generated and characterized in house and bred with C57BL/6J-Sting1gt/J mice to generate Sting1−/−Enpp1H362A mice. Male and female mice were included in every experiment, unless otherwise noted. Mice were maintained at Stanford University in compliance with the Stanford University Institutional Animal Care and Use Committee regulations. All procedures were approved by the Stanford University Administrative Panel on Laboratory Animal Care.
In Vivo and In Vitro HSV-1 Infection Models.
For in vivo and in vitro HSV-1 infection models, 2.5 × 107 PFU of HSV-1 was diluted in 100 μL PBS and injected intravenously into the tail vein of 6- to 9-wk-old mice. After 6 h, 12 h, or 6 d, the mice were euthanized in a CO2 chamber and blood and organs were harvested.
Total Body Irradiation Mouse Model.
Male and female 8- to 12-wk-old mice were irradiated with either 8 or 9 Gy using a 225-kVp cabinet X-ray irradiator with a 0.5-mm Cu filter (IC-250, Kimtron Inc.). The mice were weighed daily and were euthanized if they met the humane endpoint of greater than 20% weight loss for 2 consecutive days.
Statistical Analysis.
All statistical tests were performed using GraphPad Prism software and are noted in the figure legends. Data are presented as the mean ± SD, unless otherwise stated.
Supplementary Material
Acknowledgments
We thank S. Wang for signal peptide analysis; F. Sunden for Xac NPP plasmids and expression advice; K. Nguyen for LC-MS/MS analysis; the beamline staff at the Stanford Synchrotron Radiation Lightsource (SSRL) for excellent technical support during data collection; the Stanford Transgenic, Knockout, and Tumor Center for help generating transgenic mice; and all L.L. Laboratory members and J. Long for constructive comments and discussion. J.A.C. was supported by NIH 5F31CA239510 and the Stanford Interdisciplinary Graduate Fellowship affiliated with ChEM-H. A.F.C. was supported by NIH 5T32GM736544 and 1F30CA250145. This work was supported by NIH DP2CA228044 (to L.L.). Use of the SSRL, SLAC National Accelerator Laboratory, is supported by the US Department of Energy, Office of Science, Office of Basic Energy Sciences under Contract No. DE-AC02-76SF00515. The SSRL Structural Molecular Biology Program is supported by the Department of Energy Office of Biological and Environmental Research and by the NIH, National Institute of General Medical Sciences (P30GM133894). The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of National Institute of General Medical Sciences or NIH.
Footnotes
Competing interest statement: L.L. is a scientific cofounder of Angarus Therapeutics, which has exclusive licensing rights to the patent WO2020160333A1. J.A.C., V.B., and L.L. are inventors on the patent WO2020160333A1.
This article is a PNAS Direct Submission.
This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.2119189119/-/DCSupplemental.
Data Availability
All study data are included in the article and SI Appendix.
References
- 1.Wu J., et al. , Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ablasser A., et al. , cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Diner E. J., et al. , The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell Rep. 3, 1355–1361 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Li X.-D., et al. , Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341, 1390–1394 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mackenzie K. J., et al. , cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Harding S. M., et al. , Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548, 466–470 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bakhoum S. F., et al. , Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 553, 467–472 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Woo S. R., et al. , STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Deng L., et al. , STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sliter D. A., et al. , Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yu C. H., et al. , TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell 183, 636–649.e18 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.King K. R., et al. , IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ahn J., Gutman D., Saijo S., Barber G. N., STING manifests self DNA-dependent inflammatory disease. Proc. Natl. Acad. Sci. U.S.A. 109, 19386–19391 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gao D., et al. , Activation of cyclic GMP-AMP synthase by self-DNA causes autoimmune diseases. Proc. Natl. Acad. Sci. U.S.A. 112, E5699–E5705 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gall A., et al. , Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease. Immunity 36, 120–131 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gray E. E., Treuting P. M., Woodward J. J., Stetson D. B., Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi-Goutières syndrome. J. Immunol. 195, 1939–1943 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ablasser A., et al. , Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chen Q., et al. , Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533, 493–498 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Schadt L., et al. , Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity. Cell Rep. 29, 1236–1248.e7 (2019). [DOI] [PubMed] [Google Scholar]
- 20.Gentili M., et al. , Transmission of innate immune signaling by packaging of cGAMP in viral particles. Science 349, 1232–1236 (2015). [DOI] [PubMed] [Google Scholar]
- 21.Bridgeman A., et al. , Viruses transfer the antiviral second messenger cGAMP between cells. Science 349, 1228–1232 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Carozza J. A., et al. , Extracellular cGAMP is a cancer cell-produced immunotransmitter involved in radiation-induced anti-cancer immunity. Nat. Can. 1, 184–196 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Marcus A., et al. , Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response. Immunity 49, 754–763.e4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Li J., et al. , Metastasis and immune evasion from extracellular cGAMP hydrolysis. Cancer Discov. 11, 1212–1227 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cordova A. F., Ritchie C., Böhnert V., Li L., Human SLC46A2 is the dominant cGAMP importer in extracellular cGAMP-sensing macrophages and monocytes. ACS Cent. Sci. 7, 1073–1088 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhou Y., et al. , Blockade of the phagocytic receptor MerTK on tumor-associated macrophages enhances P2X7R-dependent STING activation by tumor-derived cGAMP. Immunity 52, 357–373.e9 (2020). [DOI] [PubMed] [Google Scholar]
- 27.Ergun S. L., Fernandez D., Weiss T. M., Li L., STING polymer structure reveals mechanisms for activation, hyperactivation, and inhibition. Cell 178, 290–301.e10 (2019). [DOI] [PubMed] [Google Scholar]
- 28.Zhang C., et al. , Structural basis of STING binding with and phosphorylation by TBK1. Nature 567, 394–398 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shang G., Zhang C., Chen Z. J., Bai X. C., Zhang X., Cryo-EM structures of STING reveal its mechanism of activation by cyclic GMP-AMP. Nature 567, 389–393 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ritchie C., Cordova A. F., Hess G. T., Bassik M. C., Li L., slc19a1 is an importer of the immunotransmitter cGAMP. Mol. Cell 75, 372–381.e5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lahey L. J., et al. , LRRC8A:C/E heteromeric channels are ubiquitous transporters of cGAMP. Mol. Cell 80, 578–591.e5 (2020). [DOI] [PubMed] [Google Scholar]
- 32.Luteijn R. D., et al. , SLC19A1 transports immunoreactive cyclic dinucleotides. Nature 573, 434–438 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhou C., et al. , Transfer of cGAMP into bystander cells via LRRC8 volume-regulated anion channels augments STING-mediated interferon responses and anti-viral immunity. Immunity 52, 767–781.e6 (2020). [DOI] [PubMed] [Google Scholar]
- 34.Li L., et al. , Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat. Chem. Biol. 10, 1043–1048 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Orriss I. R., Arnett T. R., Russell R. G. G., Pyrophosphate: A key inhibitor of mineralisation. Curr. Opin. Pharmacol. 28, 57–68 (2016). [DOI] [PubMed] [Google Scholar]
- 36.Li Q., et al. , Mutant Enpp1asj mice as a model for generalized arterial calcification of infancy. Dis. Model. Mech. 6, 1227–1235 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rutsch F., et al. , PC-1 nucleoside triphosphate pyrophosphohydrolase deficiency in idiopathic infantile arterial calcification. Am. J. Pathol. 158, 543–554 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rutsch F., et al. , Mutations in ENPP1 are associated with ‘idiopathic’ infantile arterial calcification. Nat. Genet. 34, 379–381 (2003). [DOI] [PubMed] [Google Scholar]
- 39.Kato K., et al. , Crystal structure of Enpp1, an extracellular glycoprotein involved in bone mineralization and insulin signaling. Proc. Natl. Acad. Sci. U.S.A. 109, 16876–16881 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kato K., et al. , Structural insights into cGAMP degradation by Ecto-nucleotide pyrophosphatase phosphodiesterase 1. Nat. Commun. 9, 4424 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Carozza J. A., et al. , Structure-aided development of small-molecule inhibitors of ENPP1, the extracellular phosphodiesterase of the immunotransmitter cGAMP.Cell Chem. Biol. 27, 1347–1358.e5 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zalatan J. G., Fenn T. D., Brunger A. T., Herschlag D., Structural and functional comparisons of nucleotide pyrophosphatase/phosphodiesterase and alkaline phosphatase: Implications for mechanism and evolution. Biochemistry 45, 9788–9803 (2006). [DOI] [PubMed] [Google Scholar]
- 43.Hausmann J., et al. , Structural basis of substrate discrimination and integrin binding by autotaxin. Nat. Struct. Mol. Biol. 18, 198–204 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Döhler C., Zebisch M., Sträter N., Crystal structure and substrate binding mode of ectonucleotide phosphodiesterase/pyrophosphatase-3 (NPP3). Sci. Rep. 8, 10874 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Goding J. W., Grobben B., Slegers H., Physiological and pathophysiological functions of the ecto-nucleotide pyrophosphatase/phosphodiesterase family. Biochim. Biophys. Acta 1638, 1–19 (2003). [DOI] [PubMed] [Google Scholar]
- 46.Sunden F., et al. , Differential catalytic promiscuity of the alkaline phosphatase superfamily bimetallo core reveals mechanistic features underlying enzyme evolution. J. Biol. Chem. 292, 20960–20974 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lowey B., et al. , 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 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fatma S., Chakravarti A., Zeng X., Huang R. H., Molecular mechanisms of the CdnG-Cap5 antiphage defense system employing 3′,2′-cGAMP as the second messenger. Nat. Commun. 12, 6381 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gijsbers R., Ceulemans H., Stalmans W., Bollen M., Structural and catalytic similarities between nucleotide pyrophosphatases/phosphodiesterases and alkaline phosphatases. J. Biol. Chem. 276, 1361–1368 (2001). [DOI] [PubMed] [Google Scholar]
- 50.Wilson J. E., Chin A., Chelation of divalent cations by ATP, studied by titration calorimetry. Anal. Biochem. 193, 16–19 (1991). [DOI] [PubMed] [Google Scholar]
- 51.Storer A. C., Cornish-Bowden A., Concentration of MgATP2- and other ions in solution. Calculation of the true concentrations of species present in mixtures of associating ions. Biochem. J. 159, 1–5 (1976). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Smith R. M., Martell A. E., Chen Y., Critical evaluation of stability constants for nucleotide complexes with protons and metal ions and the accompanying enthalpy changes. Int. Union Pure Appl. Chem. 63, 1015–1080 (1991). [Google Scholar]
- 53.Belli S. I., van Driel I. R., Goding J. W., Identification and characterization of a soluble form of the plasma cell membrane glycoprotein PC-1 (5′-nucleotide phosphodiesterase). Eur. J. Biochem. 217, 421–428 (1993). [DOI] [PubMed] [Google Scholar]
- 54.Jansen S., et al. , Structure of NPP1, an ectonucleotide pyrophosphatase/phosphodiesterase involved in tissue calcification. Structure 20, 1948–1959 (2012). [DOI] [PubMed] [Google Scholar]
- 55.Rutsch F., et al. ; GACI Study Group, Hypophosphatemia, hyperphosphaturia, and bisphosphonate treatment are associated with survival beyond infancy in generalized arterial calcification of infancy. Circ. Cardiovasc. Genet. 1, 133–140 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Roberts F. L., et al. , Osteoblast-specific deficiency of ectonucleotide pyrophosphatase or phosphodiesterase-1 engenders insulin resistance in high-fat diet fed mice. J. Cell. Physiol. 236, 4614–4624 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lee B. H., et al. , Activation of P2X7 receptor by ATP plays an important role in regulating inflammatory responses during acute viral infection. PLoS One 7, e35812 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Corrêa G., et al. , The purinergic receptor P2X7 role in control of Dengue virus-2 infection and cytokine/chemokine production in infected human monocytes. Immunobiology 221, 794–802 (2016). [DOI] [PubMed] [Google Scholar]
- 59.Zhang C., et al. , Virus-triggered ATP release limits viral replication through facilitating IFN-β production in a P2X7-dependent manner. J. Immunol. 199, 1372–1381 (2017). [DOI] [PubMed] [Google Scholar]
- 60.Römling U., Galperin M. Y., Gomelsky M., Cyclic di-GMP: The first 25 years of a universal bacterial second messenger. Microbiol. Mol. Biol. Rev. 77, 1–52 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Davies B. W., Bogard R. W., Young T. S., Mekalanos J. J., Coordinated regulation of accessory genetic elements produces cyclic di-nucleotides for V. cholerae virulence. Cell 149, 358–370 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gao D., et al. , Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903–906 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhao Q., Wei Y., Pandol S. J., Li L., Habtezion A., STING signaling promotes inflammation in experimental acute pancreatitis. Gastroenterology 154, 1822–1835.e2 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Edgar R. C., MUSCLE: Multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004). [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
Data Availability Statement
All study data are included in the article and SI Appendix.