INTRODUCTION
Human adenoviruses (Ads) are a family of nonenveloped double-stranded DNA viruses that contribute to upper respiratory infections, epidemic conjunctivitis, and severe acute respiratory diseases in military recruits (
1). Among the 57 serotypes of human adenoviruses, the group C type 2 and 5 adenoviruses are the most studied, providing a basic foundation for adenoviral mechanisms of transformation, viral gene expression, viral DNA replication, and virus entry. Replication-defective versions of adenovirus type 5 (Ad5) have been widely used as vectors for gene delivery, gene therapy, anticancer, and DNA vaccine applications. Infection by wild-type (WT) adenoviruses or replication-defective recombinant Ad5 vectors (rAd5Vs) stimulates an antiviral innate response that is followed by cellular and humoral antiviral immune responses.
Virus uptake by immune sentinel macrophages and dendritic cells (DCs) plays a pivotal role in determining the outcome of a viral infection. Macrophages contribute directly to viral clearance (
2–4). Virus uptake by these cells triggers an antiviral response through the activation of pattern recognition receptors (PRRs) (
5–9). Induction of beta interferon (IFN-β), a type I interferon, was established nearly 50 years ago as a dominant feature of the antiviral recognition response to adenovirus (
10,
11). Adenovirus DNA (vDNA) was subsequently identified as a pathogen-associated molecular pattern (PAMP) that led to IFN-β induction (
12). Using a variety of model systems, several PRRs, including Toll-like receptor 9 (TLR9), DAI, DDX41, p204 (IFI16), RNA polymerase III (Pol III), and cyclic adenine guanine synthase (cGAS), have been identified as DNA sensors that can lead to type I IFN expression (reviewed in references
13–15). Two general rAdV-responsive, IFN-β-inducing, DNA-sensing cascades have been identified: one is MyD88 dependent and the other is MyD88 independent (
7). The TLR9/MyD88 cascade is activated by vDNA in plasmacytoid dendritic cells (pDCs) (
7). This cascade signals through TRAF6 stimulation of interferon response factor 7 (IRF7), a transcription factor that binds a consensus IRF binding site, AANNGAAA, present in the IFN-β promoter (
16–18). In conventional DCs or macrophages, adenovirus induction of IFN-β occurs through the activation of IRF3 by a MyD88-independent mechanism (
6,
7,
19). Virus entry, endosomal escape, and exposure of vDNA to the cytosol are required for the activation of IRF3 (
6). Presumably, vDNA is exposed to PRRs during the process of transport to the nucleus. In murine and human cell lines, cGAS (
20,
21) has been identified as a vDNA PRR required for IFN-β induction following adenovirus infection of responsive cell lines (
22,
23).
The mechanism for cGAS induction of IRF3 has been established through
in vitro studies. Binding of DNA by cGAS induces protein conformational changes and homodimerization (
24,
25). Dimerized cGAS synthesizes 2′-3′ cyclic guanine adenine monophosphate (cGAMP) (
20,
26,
27), and cGAMP binds to the STING adaptor protein. Activation of STING results in migration from the endoplasmic reticulum to membrane vesicles associated with autophagosome proteins (
28,
29). STING binds tank-binding kinase 1 (TBK1), and TBK1 undergoes phosphorylation (
pSer172TBK1) (
30–32). The STING/TBK1 scaffold complex binds IRF3, presenting the C-terminal domain of IRF3 to TBK1 for phosphorylation (
pSer396IRF3) (
31).
pSer396IRF3 undergoes dimerization and translocation to the nucleus, where it engages the beta interferon promoter consensus IRF-binding site and contributes to the upregulation of beta interferon gene expression (
33–36). Not all cells are equally equipped to carry out an efficient antiviral recognition response. Differences in virus entry and variable levels of either cGAS or STING impact the potency of the primary antiviral recognition response (
22,
37).
Through the activation of IRF3 and the induction of IFN-β, the cGAS-induced antiviral cascade is amplified by autocrine and paracrine IFN-β activation of secondary signaling cascades (reviewed in reference
38). IFN-β binds to interferon receptor I (IFNRI) present on infected and uninfected cells. IFNRI activation mediates Tyk/Jak phosphorylation of STAT1 and STAT2, the formation of interferon-stimulated gene factor 3 (ISGF3), and the transcriptional activation of interferon-stimulated genes (ISGs) (
39). Although IFN-β is a dominant feature of the antiviral secondary signaling response to rAd5V, other cytokines and chemokines (tumor necrosis factor alpha [TNF-α], interleukin-6 [IL-6], and IL-1) are also expressed following virus infection and make significant contributions to the antiviral response to rAdV. In cell line models, the antiviral response to rAdV is minimally a combination of two cell populations, naive uninfected cells that undergo simple paracrine cytokine stimulation and infected cells that have undergone both primary antiviral response signaling and secondary cytokine-induced autocrine signaling.
In vivo, several additional factors contribute to the initiation of both the innate and adaptive antiviral immune responses to rAdV (
40). Following systemic administration of virus, rAd5 virions are exposed to diverse serum factors. Serum proteins such as factor X (
41–43), native antibodies (
3,
4,
44), or preexisting serotype-specific antibody (
45) can bind virus and influence mechanisms contributing to virus uptake, transduction, localization, and intracellular signaling. How these interactions impact antiadenovirus responses has not been fully established. A second layer of complexity occurs by the varied population of cells that take up virus
in vivo (i.e., neutrophils, macrophages, dendritic cells, endothelial cells, and hepatocytes). Depending on delivery and the availability of PRR/adaptor complexes, each cell type may initiate a unique antiviral response program. The
in vivo antiviral response to adenovirus reflects the combined aggregate of cell-specific primary activation responses overlaid with cytokine/chemokine-mediated secondary signaling.
The antiviral response to rAdV matures with time. The early PRR innate response occurs rapidly at 0 to 6 h postinfection (p.i.) in the murine model, and by 24 h, the ISG transcript induction phase is in decline (
46). At this point, the majority of systemically administered virus is cleared through innate mechanisms (
47). Coincident with vector clearance, infected dendritic cells undergo maturation and migration to regional lymph nodes, where, through the presentation of viral antigens, they stimulate CD4 and CD8 T-cell activation against viral epitopes (
48). In immunocompetent murine models, the presentation of virus-associated gene products by infected cells contributes to efficient elimination through T-cell-mediated cytolysis (
49–51). Since DC maturation contributes to both T- and B-cell antigen-dependent selection, defects in antigen-presenting cell (APC) maturation may impact the antiviral adaptive immune response.
In this study, we have determined how the antiviral recognition response in APCs derived from cGAS and STING knockout (KO) mice compares to that in wild-type cells derived from C57BL/6 mice or from a negative-control IRF3−/− knockout strain. We extend our characterization of these knockout strains to assess how early innate antiviral signaling is altered in response to systemic administration of rAd5V and how these mutant mouse strains impact the hepatic clearance of rAdV and the production of antiadenovirus neutralizing antibody (NAb).
MATERIALS AND METHODS
Viruses.
Ad5CiG and Ad5βGal were previously described (
52,
53) and were grown on a large scale in HEK-293 cells according to standard protocols, purified through two rounds of CsCl gradient ultracentrifugation, and stored at −80°C in storage buffer (10 mM Tris, 2 mM MgCl
2, 4.0% sucrose [pH 7.5]). Virus particle numbers were quantified by spectrophotometric detection of intact virions at an optical density at 260 nm (OD
260) (10
12 particles/OD
260 unit).
Mice.
Eight- to twelve-week-old male and female C57BL/6 and C57BL/6J-Tmem173gt/J (STING KO) mice were obtained from Jackson Laboratories, cGAS−/− C57BL/6 mice were generously provided by H. Virgin, and IRF3−/− mice were generously provided by T. Taniguchi through Riken BRC.
Bone marrow-derived macrophages and dendritic cells.
Bone marrow cells were extracted from the femurs and tibiae of mice. For bone marrow-derived macrophages (BMMOs), red cells were removed with a lysis solution (0.15 M NH
4Cl, 1 mM KHCO
3, 0.1 mM EDTA). Bone marrow cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 20% fetal bovine serum (FBS) and 25% supernatant derived from confluent L929 cells. On day 7, immature macrophages were collected. Bone marrow-derived dendritic cells (BMDCs) were differentiated in RPMI medium containing 10% FBS, 1%
l-glutamine, and beta-mercaptoethanol (BME) in the presence of 20 ng/ml granulocyte-macrophage colony-stimulating factor (GM-CSF), as previously described (
54). Briefly, bone marrow was harvested and plated into petri dishes at a density of 4 × 10
5 cells/ml, 10 ml/dish. At day 3, 10 ml of medium was added, and at days 6 and 8, 10 ml of medium was replenished. Immature, nonadherent BMDCs were gently harvested at day 10. The typical purity of each population was >90%, as determined by fluorescence-activated cell sorter (FACS) analysis of CD11b and CD11c surface markers, respectively.
Culture infections.
BMMOs were harvested at day 7, BMDCs were harvested at day 10, and cells were plated at densities of 5 × 105 cells/ml in 12-well plates (Western blot and flow cytometry analyses) (5 × 105 cells/well) and 106 cells/ml in 6-well plates (for RNA extraction) (2.5 × 106 cells/well). Twenty-four hours after plating, cells were infected with Ad5CiG at 20,000 virus particles (vp)/cell and diluted in Opti-MEM. For Western blot analysis, cells were harvested at 2, 4, and 6 h p.i. Flow cytometry was performed at 24 h p.i., and RNA extraction was performed at 6 h p.i.
Animal infections.
Mice were infected by intraorbital intravenous (i.v.) injections with 1011 vp/mouse of Ad5βGal diluted in DMEM. Mice were euthanized at 5 h, 24 h, and 28 days postinfection for liver and blood serum harvest. All experiments were performed in accordance with Weill Cornell Medical College IACUC protocols.
Antibodies, surface staining, and fixation.
Antibodies were purchased from BioLegend (San Diego, CA). BMMOs and BMDCs were prepared from bone marrow as described above. For phenotypic analysis, cells were surface stained with the following monoclonal antibodies (MAbs): anti-CD11b allophycocyanin (APC) (catalog number 101212), anti-CD11c Pac Blue (catalog number 117322), anti-CD11c APC (catalog number 117310), anti-CD86 phycoerythrin (PE) (catalog number 105008), and anti-major histocompatibility complex class II (MHC-II) peridinin chlorophyll protein (PerCP)-Cy5.5 (catalog number 116416). After staining, washing, and fixation in 1% paraformaldehyde, cells were analyzed by using a FACSCanto flow cytometer (BD Biosciences). Data analysis was performed by using FlowJo software, and 12,000 events per sample were analyzed.
Western blot analysis.
Whole-cell extracts were prepared by washing cells twice with ice-cold phosphate-buffered saline (PBS) and incubating them in lysis buffer (50 mM Tris [pH 7.5], 150 mM NaCl, 1 mM EDTA, 1% NP-40) with the addition of Phosphatase Inhibitor Cocktails 1 and 2 (catalog numbers P2850 and P5726; Sigma) and protease inhibitors (30 mM sodium fluoride, 1 mM phenylmethylsulfonyl fluoride [PMSF], 10 μg/ml aprotinin, 10 μg/ml leupeptin, 1 μg/ml pepstatin, 1 mM benzamidine) for 30 min at 4°C on a rocking platform before scraping and transferring cells to tubes. The lysates were cleared by centrifugation at 13,000 × g for 20 min at 4°C, and protein quantification was performed with the DC protein assay kit (Bio-Rad Laboratories).
For Western blot analysis, 20 μg total protein was separated by using standard 10% SDS-polyacrylamide gels and transferred onto polyvinylidene difluoride (PVDF) membranes (Immobilon P; Millipore). All blots were blocked in 5% skim milk in Tris-buffered saline (TBS) plus Tween (0.1%) at room temperature for 1 h. Phospho-IRF3 (Ser396) (catalog number 4947), phospho-STAT1 (58D6) (Tyr701) (catalog number 9167), beta-actin (catalog number 4967), total IRF3 (catalog number 4302), STING (catalog number 3337), TBK1 (catalog number 3503), phospho-NF-κB p65 (pNF-κB p65) (Ser536) (catalog number 3033), pTBK1 (Ser172) (catalog number 5483), green fluorescent protein (GFP) (catalog number 2956), and horseradish peroxidase (HRP)-linked anti-rabbit IgG (catalog number 7074) antibodies were obtained from Cell Signaling. Primary antibodies were used at a dilution of 1:2,000 to 1:3,000 in 5% bovine serum albumin (BSA)–TBS. The HRP-linked secondary antibody was diluted 1:4,000 in 5% milk–Tween–TBS. Detection was done with the Luminator Crescendo Western HRP substrate (Millipore).
SYBR green I RT-qPCR.
Total cellular mRNA was isolated by using RNAzol reverse transcriptase (RT) (Molecular Research Center) according to the manufacturer's instructions. For RT quantitative PCR (RT-qPCR), a two-step protocol was employed: first, cDNA was synthesized from 2 μg total RNA in a volume of 20 μl by using random hexamer primers with the Maxima first-strand cDNA synthesis kit (Fermentas), and second, amplifications were carried out with a total volume of 15 μl by using the Maxima SYBR green/ROX qPCR master mix (Fermentas) in an Applied Biosystems Prism 7900H sequence detection system with SDS 2.1 software. Cycles consisted of an initial incubation step at 95°C for 10 min; 40 cycles at 95°C for 15 s, 60°C for 30 s, and 72°C for 30 s; and a melting-curve analysis cycle. Data acquisition was performed during the extension step. All determinations were performed in technical triplicate. Nontemplate and no-RT controls were run with every assay and had cycle threshold (
CT) values that were significantly higher than those of experimental samples or were not detected. The relative abundance of each mRNA was calculated by the ΔΔ
CT method (
55,
56), normalizing to Tata-binding protein (TBP) expression with standardization to one reference sample, as indicated. For comparisons between cell lines, or in late-stage virus infections, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a normalization standard. Sequences of primers are available upon request.
SYBR green I qPCR of viral DNA.
Liver fragments were Dounce homogenized in lysis buffer containing 10 mM NaCl, 10 mM Tris (pH 8.0), 10 mM EDTA (pH 8.0), and 0.5% SDS, followed by proteinase K digestion. Crude homogenates were cleared by double centrifugation. Total DNA was purified by using NaCl salting-out and ethanol precipitation protocols (
57). Following DNA purification and resuspension in 100 μl H
2O, OD
260 DNA concentrations were determined by using a NanoDrop instrument. Five nanograms of sample DNA in a final reaction mixture volume of 15 μl (Maxima SYBR green/ROX qPCR master mix [Fermentas] system) was used for each assay. Ad5 hexon and cellular control primers (IFN-β) were used to characterize each DNA sample. Normalization to the cellular genomic standard was used to derive the relative viral DNA yield/host genome. Assays were carried out with an Applied Biosystems Prism 7900H sequence detection system with SDS 2.1 software. Cycles consisted of an initial incubation step at 95°C for 10 min; 40 cycles at 95°C for 15 s, 60°C for 30 s, and 72°C for 30 s; and a melting-curve analysis cycle. Data acquisition was performed during the extension step. Data points represent the averages of results from biological and technical triplicates.
β-Gal assays.
The Tropix Galacto-Star chemiluminescent reporter gene assay (catalog number T1012; Applied Biosystems) was used for β-galactosidase (β-Gal) detection in the livers of infected mice at 24 h and 28 days p.i., according to the manufacturer's protocol. Liver fragments were sonicated on ice for 20 s in 1 ml lysis buffer with PMSF. Homogenates were spun down twice at 13,000 rpm for 15 min. The supernatant was heat inactivated at 48°C for 1 h to neutralize endogenous β-galactosidase activity and spun down at 13,000 rpm for 15 min. The protein concentration was determined by a Bradford assay (
58). Ten microliters of the supernatant per well was loaded into 96-well Immulon assay plates and incubated with 100 μl reaction buffer for 30 min at room temperature. The luminescence in each well was measured for 1 s. Each sample was loaded in triplicates, and data were averaged. The background given by the lysis buffer was subtracted, and results were expressed per microgram of liver protein.
Antibody neutralization assays.
Mouse blood samples isolated at 28 days postinfection were left at room temperature for 15 min to allow clot formation and then centrifuged at 13,000 rpm for 10 min for serum separation. Mouse serum was immediately flash frozen and thawed when used for biological assays. Briefly, serum neutralization assays were carried out as follows. HeLa cells were plated out at 2.5 × 104 cells/well in 96-well plates 24 h prior to infection. On the following day, serum was thawed, and 1:4 serial dilutions in DMEM were generated for each serum sample. Ad5βGal, diluted in DMEM for a final delivery of 103 particles/cell, was incubated with a serum dilution (technical triplicates for each sample) for 1 h at 37°C. Following virus/serum incubation, the mixture was incubated with HeLa cells for 30 min at 37°C. Medium was removed, and cells were washed with fresh medium and incubated in 100 μl fresh medium for 24 h. At 24 h p.i., plates were spun down at 2,000 rpm for 5 min, and medium was removed. Cell lysates were harvested in a volume of 100 μl lysis buffer. Ten microliters of the supernatant was applied for a Tropix Galacto-Star activity assay as described above. The background generated by the lysis buffer was subtracted from the result for each assay point, and results were expressed as luminescence units.
Cytokine and chemokine multiplex assays.
Mouse serum derived from blood samples as described above was harvested at the indicated times and stored at −80°C until it was evaluated in cytokine assays. Serum was centrifuged for 15 min at 13,000 rpm to remove any residual cells or debris and then evaluated for cytokine secretion with Meso Scale Discovery (MSD) (Rockville, MD) technology using a mouse proinflammatory 7-plex tissue culture kit (catalog number K15012B-1). Kits were run according to the manufacturer's instructions. Samples were run in duplicate, at the Weill Cornell Medical School CTSC facility, by using Meso Scale Discovery Sector Imager 2400 (SI2400).
ELISA.
Mouse IFN-β was measured by using a mouse IFN-β (PBL Interferon Source) enzyme-linked immunosorbent assay (ELISA) kit (catalog number 42400; R&D Systems). Serum from mice was obtained as described above and stored at −80°C until it was evaluated for cytokine secretion. Kits were run according to the manufacturer's instructions. The absorbance at 450 nm was read within 5 min after the addition of the stop solution. Each sample was assayed as a technical duplicate.
Statistical analysis.
Data were expressed as means ± standard errors of the means. Statistical analysis was performed with Student's t test. A P value of <0.05 was considered significant.
DISCUSSION
Induction of the type I interferon antiviral response is a well-established mechanism for controlling virus infections. For both RNA and DNA viruses, viral nucleic acids are key pathogen-associated molecular patterns recognized by host pattern recognition receptors in a cell-specific manner. An array of nucleic acid-sensing PRRs have been identified, which leads to IRF3 activation and the induction of the type I interferon antiviral response. We have asked how the cGAS/STING DNA-sensing cascade contributes to the antiadenovirus recognition response in vitro and in vivo in the nonpermissive mouse model. We have demonstrated that cGAS/STING-dependent signaling is essential for initiating an in vitro antiviral response in murine APCs following exposure to rAdV. Furthermore, the cGAS/STING DNA-sensing cascade has a major role in defining antiviral inflammatory responses in vivo at early times postinfection (5 h). The data also indicate that cGAS/STING-independent antiviral recognition pathways were activated following systemic administration of rAdV. In spite of compromised APC activation in vitro and early antiviral response deficiencies in cGAS/STING knockout mice, adaptive immune endpoint assays (vector clearance and neutralizing antibody at 28 days postinfection) were only marginally affected compared to those for wild-type mice. Based on the latter observations, we conclude that the cGAS/STING DNA-sensing cascade is not essential for clearance of infected cells or the generation of a humoral immune response to rAd vectors.
Criteria used to assess cGAS/STING-dependent antiviral responses in primary BMMOs and BMDCs included the activation of primary and secondary response markers (pSer172TBK1, pSer396IRF3, and pTyr701STAT1), transcriptional induction of established antiviral and proinflammatory genes, and increased surface expression of CD86 costimulatory proteins. In BMMOs from cGAS and STING knockout strains, the lack of a significant antiviral response indicates that the antiviral recognition/signaling response under these in vitro conditions is exclusively dependent on cGAS activation of the STING adaptor pathway. IRF3 knockout mice were used for comparison as a negative control. As the one established target of the cGAS/STING/TBK1 signaling cascade, the IRF3 knockout phenotype might be expected to be phenotypically similar to both cGAS and STING mutations. Data from in vitro assays of BMMO activation and maturation were completely in agreement with this premise, indicating that cGAS/STING/TBK1/IRF3 is the featured antiadenovirus recognition response pathway for BMMOs.
We came to a similar conclusion with BMDCs, but there were noticeable differences. The cGAS/STING cascade is required for primary and secondary signaling, transcript induction, and the upregulation of the maturation marker CD86. However, when rAdV induction of RNA transcripts from cGAS- and STING-deficient BMDCs was assessed, a low-level, cGAS/STING-independent, transcript-specific induction response was detected (
Fig. 3B). This low-level response was not observed for IFN-β mRNA but was detected for select ISGs and proinflammatory transcripts. In BMDCs, we also found several features that distinguished the IRF3 knockout strain from both cGAS and STING knockout BMDCs. As noted above for BMMOs, levels of pTBK1 were induced by rAdV in IRF3
−/− mouse-derived cells. Levels of pSTAT1 were also detectable at 6 h postinfection in IRF3
−/− BMDCs but not in AdV-treated cGAS
−/− or STING
−/− cells. Furthermore, levels of transcript induction in IRF3
−/− BMDC were significantly higher for IFN-β after infection and trending toward higher levels for viperin, proinflammatory transcripts, and ISGs than for cGAS
−/− and STING
−/− BMDC. One possible explanation for the low-level antiviral response in IRF3
−/− BMDCs but not in cGAS
−/− or STING
−/− BMDCs would be TBK1 activation of a second IRF substrate that is not available to the same extent in BMMOs. IRF7 is the most obvious candidate (
19), and a recent study indicated that a cGAS-independent STING/IRF7 pathway plays a significant role in the immune response to DNA vaccines (
62). Other possibilities include secondary signaling targets of cGAMP or STING, independent of TBK1. Further studies will be required to determine the mechanism contributing to residual activity in the knockout BMDCs. These observations indicate that cell-specific phenotypes are detectable when cGAS and STING knockouts are compared to IRF3 knockouts. The virus recognition complexes operating in BMDCs indicate a greater degree of complexity than those functioning in BMMOs.
Specific explanations for differences between BMMOs and BMDCs could involve secondary PRRs functioning in BMDCs but not BMMOs. Such a BMDC pathway necessarily signals through a STING/TBK1/IRF3-independent mechanism. MyD88 signaling pathways that influence the APC response to rAdVs are one possibility; enhanced activities of IFI16, DDX41, or AIM2 are examples of established DNA sensors that may operate in BMDCs as secondary DNA sensors that are able to engage in STING-independent signaling. Alternatively, cell-specific differences in intracellular signaling through penton-integrin stimulation may differentially influence the induction of a subpopulation of antiviral/inflammatory transcripts following infection. Since secondary signaling by IFNs (pSTAT1) was not evident, and upregulation of the CD86 maturation marker was not observed, we believe that the low-level transcription response to rAdV in cGAS and STING knockout BMDCs is biased toward an NF-κB-dependent inflammatory response and not an IFN-inducing mechanism.
The early antiviral response to systemically administered rAdV in cGAS−/− and STING−/− mice compared to that in WT or IRF3−/− mice was largely consistent with the in vitro characterization of APCs. By using an ELISA to quantify serum levels of IFN-β at 5 h postinfection, significant induction was found for WT mice but was absent in STING−/− and cGAS−/− mice and marginally detectable in IRF3−/− mice. This observation was consistent with levels of total liver IFN-β mRNA expressed following virus infection. Inflammatory cytokines corresponding to TNF-α, IL-6, and IL-10 were induced in WT mice, and levels of induction were significantly lower in knockout strains. TNF-α and IL-6 transcript induction occurred in WT mice but was significantly reduced in each knockout strain. ISG transcript induction was predictably diminished in knockout strains. Because total liver RNA assays reflect a population of cell types, we believe that a major contributor to the induction profile arises from Kupffer cell uptake of adenovirus and secondary signaling events through the expression of IFNs. Systemic delivery of rAdV exposes the virus to serum factor binding as well as opsonization, which impacts virus uptake into a variety of cell types. This may expose the virus to PRRs other than cGAS. Our data indicate that following systemic administration of rAdV, signaling through the cGAS/STING cascade was the predominant mechanism for the induction of antiviral response transcripts.
Previous studies established that compromised dendritic cell activation/maturation (
63) influenced both the clearance of infected cells and the induction of an antiviral humoral immune response. Although BMDC activation and maturation were compromised in cells derived from the cGAS
−/−, STING
−/−, or IRF3
−/− strain, using systemically administered virus
in vivo, both vector clearance and antiviral neutralizing antibody responses occurred. The minimal impact of the type I IFN antiviral response on the adaptive immune response to AdV is consistent with data from studies characterizing antiadenovirus responses in the STAT2 knockout Syrian hamster model (
64) and recent observations of rAdV vaccine vectors in IFNRI and STING knockout strains (
65). Our observations that compromised dendritic cell maturation
in vitro can be separated from an adaptive immune response phenotype are consistent with the characterization of the antiviral response to rAdV in type I IFN receptor knockout mice (
66).
The cGAS/STING cascade has been viewed as an adjuvant target for vaccine applications (
20). Clearly, this pathway facilitates APC presentation and upregulation of costimulatory molecules. Our observation that the anti-Ad neutralizing antibody level was modestly reduced in cGAS-deficient mice is consistent with such a role. The distinct parameters necessary for triggering an adaptive immune response through dendritic cell migration/activation and those required for the adjuvant properties associated with stimulation of the cGAS/STING cascade require further investigation.
In vivo, DC maturation and migration following virus activation are complex and dynamic processes leading to CD4 and CD8 T-cell priming. Recent reports have revealed that for different viruses, infection and DC migration to lymph nodes result in distinct spatially and temporally defined interactions between CD4 and CD8 T cells and multiple subsets of DCs (
67,
68). The factors that influence these distinct DC interactions with T-cell subsets have not been fully established for Ad infections. The residual low-level antiadenovirus activation response identified following infection of cGAS/STING-deficient BMDCs may be augmented by unidentified
in vivo interactions leading to eventual T- and B-cell priming in regional lymph nodes. Further studies will be required to identify these interactions. The studies presented here have established the cGAS/STING pathway as the dominant antiadenovirus type I IFN-inducing cascade
in vitro and
in vivo in the murine model, with no observed impact on viral clearance, and a cGAS deficiency resulted in a slightly diminished humoral immune response to viral infection.