Executing tumor cell death
There is increasing interest in developing cancer immunotherapies that target the innate immune pathways regulating cytokine production and cell death, but the interplay between these two closely connected processes is not well understood. In mouse and human cancer cell lines, Boehmer et al. demonstrate that cytokine production and apoptosis induced by retinoic acidтАУinducible gene I (RIG-I) ligands, including 5тА▓-triphosphate RNA (3p-RNA), are two separable events in which RIG-I is required for production of type I interferon but not execution of apoptosis. Mass spectrometry and loss-of-function assays showed that 3p-RNA directly activates OAS1 and RNase L, which promoted translational arrest and depletion of antiapoptotic MCL-1. These results demonstrate that RIG-IтАУmediated apoptosis involves priming and effector stages, reminiscent of inflammasome activation, both of which could serve as potential targets for cancer immunotherapy.
Abstract
Cytoplasmic double-stranded RNA is sensed by RIG-IтАУlike receptors (RLRs), leading to induction of type I interferons (IFN-Is), proinflammatory cytokines, and apoptosis. Here, we elucidate signaling mechanisms that lead to cytokine secretion and cell death induction upon stimulation with the bona fide RIG-I ligand 5тА▓-triphosphate RNA (3p-RNA) in tumor cells. We show that both outcomes are mediated by dsRNA-receptor families with RLR being essential for cytokine production and IFN-IтАУmediated priming of effector pathways but not for apoptosis. Affinity purification followed by mass spectrometry and subsequent functional analysis revealed that 3p-RNA bound and activated oligoadenylate synthetase 1 and RNase L. RNase LтАУdeficient cells were profoundly impaired in their ability to undergo apoptosis. Mechanistically, the concerted action of translational arrest triggered by RNase L and up-regulation of NOXA was needed to deplete the antiapoptotic MCL-1 to cause intrinsic apoptosis. Thus, 3p-RNAтАУinduced apoptosis is a two-step process consisting of RIG-IтАУdependent priming and an RNase LтАУdependent effector phase.
INTRODUCTION
The innate immune system is the first line of defense against invading pathogens. Activation of the innate immune system in infected cells mainly results in two events: (i) induction of antiviral cytokines to alarm neighboring cells and to attract and activate immune cells and (ii) activation of cell-intrinsic defense mechanisms, which may ultimately lead to programmed cell death to limit replication of intracellular pathogens. The pathways of cytokine induction and cell death are highly interconnected and tightly regulated on a molecular level (1). Because erroneous induction of programmed cell death can have fatal consequences (2, 3), multicellular organisms have evolved тАЬsafety switchesтАЭ to prevent inappropriate cell death from being executed. One common principle most prominently described in the context of inflammasomes (4) is a two-step mechanism consisted of a тАЬprimingтАЭ and an тАЬeffectorтАЭ step by which these pathways are activated.
Viral RNAs can bind to and activate innate immune receptors such as the retinoic acidтАУinducible gene I (RIG-I)тАУlike receptors (RLRs). Short double-stranded RNAs (dsRNAs) with a 5тА▓-triphosphate moiety [5тА▓-triphosphate RNA (3p-RNA)] are well established as specific ligands for RIG-I (5тАУ7). Extensive research has characterized the molecular signaling events underlying type I interferon (IFN-I) and cytokine production: Upon binding of 3p-RNA, RIG-I activates downstream IFN regulatory factor 3 (IRF3), mitogen-activated protein kinase, and nuclear factor ╬║B pathways via mitochondrial antiviral-signaling protein (MAVS) (8). Besides its essential role in antiviral cytokine induction, RIG-I has been shown by several studies to activate programmed cell death in response to viral RNA, with some selectivity for tumor cells. Initial reports describing melanoma cellтАУselective apoptosis found IFN-IтАУindependent induction of the proapoptotic Bcl-2 homology (BH)3-only proteins p53-up-regulated modulator of apoptosis (PUMA) and phorbol-12-myristate-13-acetate-induced protein 1 (NOXA) to cause intrinsic apoptosis (9тАУ11). Other reports suggested several other mechanistic models: TNF-related apoptosis-inducing ligand (TRAIL) signaling (12), down-regulation of Bcl-2/baculoviral IAP repeat-containing protein 3 (BIRC3)/protein kinase C epsilon type (PRKCE) (13), activation of caspase-8 via MAVS (14), and RLR-induced IRF3-mediated pathway of apoptosis (15, 16). However, even central aspects of the signaling involved in RIG-IтАУdependent cell death induction such as the requirement for IFN-I signaling are elusive.
We and others (9тАУ11, 17тАУ20) showed that 3p-RNA has strong antitumoral activity through induction of IFN-I and cancer cellтАУselective apoptosis. Recently, we found this cell death to be highly immunogenic, leading to potent adaptive antitumoral immune responses (17, 21). A phase 1 clinical trial of a RIG-I agonist for the treatment of solid tumors is ongoing (NCT03739138), highlighting the need to improve our understanding of 3p-RNAтАУinduced cell death. Specifically in the context of cancers, well-defined genetic evidence discriminating signaling mechanisms that cause apoptosis from those that cause cytokine induction in response to 3p-RNA exposure is still lacking. Here, we provide evidence that 3p-RNA elicits cell death via a two-step mechanism involving RIG-I/IFN-IтАУmediated priming of 3p-RNAтАУsensing executioner pathways and proapoptotic mitochondrial priming of apoptosis. Combining unbiased RNA sequencing (RNA-seq) and proteomic approaches, we characterize the 3p-RNA sensor proteins and analyze in depth the molecular mechanisms of the priming and effector phases.
RESULTS
RIG-I ligands execute tumor cell death by a RIG-IтАУindependent, IFN-IтАУprimed mechanism
To distinguish pathways inducing cytokine production, which might affect cell death in an autocrine or paracrine manner, from cell-intrinsic proapoptotic mechanisms, we first generated human 1205Lu and mouse B16 melanoma cell lines deficient for RIG-I (encoded by DDX58 in the human and Ddx58 in the mouse), melanoma differentiation-associated protein 5 (MDA5) (only in 1205Lu cells, encoded by IFIH1 in the human), and MAVS (encoded by MAVS in the human and Mavs in the mouse) using CRISPR-Cas9тАУmediated gene editing. In line with previous findings (10), we observed that 3p-RNA strongly induced cell death, as measured by annexin A5 assay, and IFN-I signaling, as measured by IFN-IтАУdependent major histocompatibility complex class I (MHC-I) surface expression and IFN-╬▓ expression, compared with a nonstimulatory control RNA (CTRL) in wild-type (WT) cells (Fig. 1, A and B, and fig. S1, A to C). In addition, cells deficient of RIG-I or its adaptor protein MAVS did not show any induction of cell death or cytokines, whereas MDA5 was dispensable for 3p-RNAтАУmediated signaling, confirming 3p-RNA to be a highly specific ligand for RIG-I in both human and murine melanoma cells. To assess how secreted factors may influence cell death induction, we next cocultured WT and RIG-IтАУdeficient cells. Fluorescent labeling of either of the cell lines before seeding allowed their discrimination by flow cytometry (Fig. 1C). Notably, the ability to undergo apoptosis in response to 3p-RNA treatment was restored in RIG-IтАУdeficient 1205Lu and B16 cells under coculture conditions (Fig. 1D and fig. S1D). This finding demonstrates that cell death and cytokine induction are two separable events upon the detection of 3p-RNA. Furthermore, this suggests the existence of one or more factors secreted by WT cells upon RIG-I activation that restore the ability of 3p-RNA to induce cell death in RIG-IтАУdeficient cells.
(A) 1205Lu human melanoma cells of the indicated genotype were stimulated with 80 nM 3p-RNA or nonstimulatory control RNA (CTRL) and analyzed for cell death by annexin A5 assay and for MHC-I expression after 48 hours (n = 2). MFI, median fluorescence intensity. (B) ELISA for human IFN-╬▓ in the cell supernatant after stimulation with 160 nM 3p-RNA or controls for 48 hours (n = 3). (C) Schematic of coculture assay and gating strategy for flow cytometric analysis. (D) WT and RIG-IтАУdeficient 1205Lu cells were cultured alone or cocultured as depicted in (C), treated with 160 nM 3p-RNA or CTRL, and analyzed by flow cytometry (n = 3, StudentтАЩs t test). (E) 1205Lu cells of indicated genotype were prestimulated with IFN-╬▒ (1000 U/ml) 18 hours before transfection with 40, 80, or 160 nM 3p-RNA or CTRL. Viability was assessed by CTB assay 48 hours after RNA transfection (n = 3, StudentтАЩs t test). (F) WT and RIG-IтАУdeficient or WT and RIG-I/IFNAR1тАУdeficient 1205Lu cells were cultured alone or cocultured, treated with 160 nM 3p-RNA or CTRL, and analyzed by flow cytometry (n = 3, one-way analysis of variance (ANOVA) with SidakтАЩs multiple comparisons test; asterisks indicate results of post hoc test.) (G) WT and IRF3тИТ/тИТ 1205Lu cells were prestimulated with IFN-╬▒ (1000 U/ml) or mock for 18 hours before transfection of indicated RNAs as shown (160 nM). Viability was measured by CTB assay (n = 3, one-way ANOVA with DunnettтАЩs multiple comparisons test). (H) Viability of WT and IFNAR1тИТ/тИТ 1205Lu cells 48 hours after transfection with 3p-RNA or CTRL (160 nM) (n = 3, StudentтАЩs t test). (I) CTB assay of WT and DDX58тИТ/тИТ cells transfected with total RNA isolated from WT or M51R VSV-infected cells (1 ╬╝g/ml) for 48 hours. DDX58тИТ/тИТ cells were treated with IFN-╬▒ or mock (├╕) for 18 hours before RNA transfection [n = 2, one-way ANOVA (WT) or two-way ANOVA (DDX58тИТ/тИТ), followed by SidakтАЩs multiple comparisons test]. Asterisks indicate results of post hoc test. Asterisks on bars represent comparison to CTRL-RNA. Means + SEM of independent experiments are shown; *P < 0.05, **P < 0.01, and ***P < 0.001; ns, not significant.
IFN-I are cytokines released upon 3p-RNA stimulation that potently induce an antiviral state by regulating hundreds of genes (22, 23), making them likely candidates for priming cells for RNA-induced death. To address this hypothesis, we stimulated DDX58тИТ/тИТ and MAVSтИТ/тИТ cells with recombinant IFN-╬▒2a (IFN-╬▒) before 3p-RNA transfection. 3p-RNA induced death in the two RLR signalingтАУdeficient human melanoma cell lines 1205Lu (Fig. 1E) and C8161 (fig. S1E) prestimulated with IFN-╬▒. IFN-╬▒ stimulation alone or together with CTRL transfection did not lead to cell death, ruling out a direct cytotoxic effect of IFN-╬▒ or RNA transfection. To determine whether IFN-I was essential for priming RIG-IтАУdeficient cells in the coculture setup, we used cells deficient in RIG-I and the IFN-╬▒/╬▓ receptor 1 (IFNAR1). In contrast to DDX58тИТ/тИТ cells, DDX58тИТ/тИТIFNAR1тИТ/тИТ cells did not undergo cell death upon coculture with WT cells (Fig. 1F). The absence of MHC-I up-regulation confirmed that IFN-I signaling was disrupted (fig. S1F). To further examine the role of IFN-I signaling, we generated knockout (KO) cells for IRF3, encoding the main transcription factor that drives the induction of IFN-I by RLRs (22, 24). Besides its role in IFN-I induction, IRF3 has been implicated in dsRNA-induced apoptosis by directly interacting with Bcl-2-associated X protein (BAX) (25). Cell death induction by 3p-RNA was strongly reduced in IRF3тИТ/тИТ cells but could be rescued by the addition of exogenous IFN-╬▒ (Fig. 1G and fig. S1G). These findings argue against a direct involvement of IRF3 in 3p-RNAтАУinduced apoptosis but suggest IRF3 to be needed for transcriptional priming through IFN-I. In line with these findings, IFNAR1-deficient cells or cells incubated with an IFNAR1-blocking antibody displayed markedly decreased 3p-RNAтАУinduced cell death, despite intact RLR and IRF3 signaling (Fig. 1H and fig. S1H).
To exclude these findings being specific to the synthetic 3p-RNA, we extended our investigation to ligands of RIG-I that are produced during viral infection. We isolated RNA from vesicular stomatitis virus (VSV)тАУinfected cells, transfected it into WT and RIG-IтАУdeficient cells, and measured cellular viability after 48 hours (Fig. 1I). VSV-derived RNAs significantly reduced cell viability compared with control RNA isolated from uninfected cells. Again, RIG-IтАУdeficient cells only showed pronounced induction of cell death when stimulated with IFN-╬▒ before RNA transfection. Analysis of C-X-C motif chemokine 10 (CXCL10) secretion from WT cells confirmed that VSV-derived RNAs were immunostimulatory (fig. S1I).
These findings indicate that IFN-I signaling is critical for an effective induction of cell death by 3p-RNA and natural RIG-I ligands. Furthermore and in contrast to early publications, RIG-I signaling itself seems to be dispensable for cell death when exogenous IFN-I is provided, suggesting the existence of a distinct IFN-IтАУinducible 3p-RNAтАУsensing pathway that executes cell death.
3p-RNA specifically binds oligoadenylate synthetase 1 and activates latent endoribonuclease
We next asked which dsRNA-sensing pathway executes cell death in response to 3p-RNA. To gain further insight into the events downstream of 3p-RNA stimulation in WT and RIG-IтАУdeficient 1205Lu cells, we analyzed the transcriptome after stimulation with 3p-RNA and/or IFN-╬▒ by RNA-seq. Six hours after transfection with 3p-RNA, WT cells showed significant changes of mRNA levels of more than 4000 genes compared with an unstimulated control (Fig. 2A). In contrast, not a single gene was differentially expressed by 3p-RNA treatment alone in DDX58тИТ/тИТ cells. However, IFN-╬▒ priming for 18 hours and subsequent 3p-RNA treatment of DDX58тИТ/тИТ cells significantly changed the expression of more than 6000 genes compared with cells treated with IFN-╬▒ alone. Analysis of Gene Ontology molecular function (GO:MF) terms of those genes revealed an enrichment of genes in transcriptional regulation (fig. S2A). However, because of the lack of known gene signatures associated with RNA recognition, we were not able to infer upstream pathways. Therefore, we analyzed RNA-binding protein genes that were selectively up-regulated in response to IFN-╬▒ by performing RNA-seq of 1205Lu WT cells untreated or treated with IFN-╬▒ for 4 hours. IFN-╬▒ treatment led to a significant up-regulation of 788 genes, among which 71 were annotated with the GO:MF terms тАЬdouble-stranded RNA bindingтАЭ or тАЬRNA bindingтАЭ (fig. S2B). Among the 30 genes with the highest log2 fold changes (FCs) (Fig. 2B), we found a group of RNA sensors known to mediate antiviral defense by directly acting on foreign nucleic acids such as protein kinase R (PKR), oligoadenylate synthetase 1 (OAS1) to OAS3, and the IFN-induced protein with tetratricopeptide repeats (IFIT) family (26). The 5тА▓-triphosphate moiety of viral RNA species is a potent pathogen-associated molecular pattern for their detection by the host and their discrimination from endogenous RNA (5, 27, 28). We further validated the necessity of the 5тА▓-triphosphate moiety for RIG-IтАУindependent cell death by stimulating IFN-╬▒тАУprimed 1205Lu DDX58тИТ/тИТ cells with dephosphorylated 3p-RNA [OH-RNA (FastAP)] and measured cytotoxicity (Fig. 2C and fig. S2C). Cell death induction by 3p-RNA was markedly reduced after FastAP treatment. Besides RIG-I itself, only IFIT1 and, to some extent, PKR (encoded by EIF2AK2) have been described to preferentially bind RNA with a 5тА▓-triphosphate moiety (28тАУ30). However, studying KO cell lines demonstrated that both IFIT1 and PKR are dispensable for cell death induction by 3p-RNA (fig. S3, A and B). IFIT1тИТ/тИТ cells showed a more rapid cell death induction compared with WT and other KO cells (fig. S3B).
(A) Volcano plots of differential gene expression (log2 FC) for indicated conditions upon 3p-RNA treatment. (B) Top 30 induced genes (log2 FC) annotated with the GO:MF term double-stranded RNA binding (blue) or RNA binding (gray) as determined by RNA-seq in 1205Lu DDX58тИТ/тИТ cells stimulated for 4 hours with IFN-╬▒ compared with untreated control. (C) IFN-╬▒тАУprimed 1205Lu DDX58тИТ/тИТ cells were transfected with FastAP- or mock-treated 3p-RNA as indicated. Cell death induction was measured 48 hours after transfection (n = 3, StudentтАЩs t test). (D) Volcano plot of relative LFQ of proteins bound to 3p-RNA-biot compared with FastAP-treated 3p-RNA-biot (OH-RNA-biot) as quantified by LC-MS/MS (n = 5). Proteins up-regulated in (A) with GO:MF term double-stranded RNA binding are highlighted in blue, those significantly enriched are indicated in green. (E) Western blot validation of MS results. 3p-RNA-biot or OH-RNA-biot was bound to streptavidin beads and used for affinity purification (AP) of proteins in lysates from unstimulated or IFN-╬▒тАУstimulated (18 hours) cells. Thirty micrograms of protein lysate was used as input. Representative of three independent experiments. (F) OAS1 activation by chemically synthesized RNAs as measured by pyrophosphate production using a chromogenic assay. CTRL, non-immunostimulatory control RNA; 3p-RNA/OH-RNA, custom-synthesized RNAs with (3p) or without (OH) 5тА▓-triphosphate group; 3тА▓-ssPy, dsRNA with single-stranded 3тА▓-pyrimidine overhang (mean ┬▒ SD of n = 3, one-way ANOVA, followed by DunnettтАЩs multiple comparisons test; asterisks indicate results of post hoc test). (G and H) rRNA integrity analysis of total RNA isolated from 1205Lu DDX58тИТ/тИТ cells using RNA BioAnalyzer. (G) IFN-╬▒тАУprimed cells were transfected with OH-RNA, 3p-RNA (both 80 nM) or poly(I:C) (1 ╬╝g/ml) for 6 hours. Left: Representative image of four independent experiments. Right: Quantification of RNA degradation by RNA-integrity number (RIN) (n = 4, StudentтАЩs t test). (H) Cells untreated or treated with IFN-╬▒ (18 hours), 3p-RNA (6 hours), or IFN-╬▒ and then 3p-RNA. Representative of at least three independent experiments. Unless stated otherwise, means + SEM of independent experiments are shown; *P < 0.05, **P < 0.01, and ****P < 0.0001.
We next used an unbiased proteomic approach by performing affinity purification of proteins from IFN prestimulated 1205Lu lysates using biotinylated 3p-RNA (3p-RNA-biot) or a dephosphorylated control (OH-RNA-biot), followed by mass spectrometry (MS) and label-free quantification (LFQ). Among the top 30 up-regulated proteins with RNA binding function, besides RIG-I that served as the internal positive control, only OAS1 was significantly enriched by 3p-RNA-biot compared with OH-RNA-biot (n = 5) (Fig. 2D). Western blot analysis confirmed the higher affinity of OAS1 to 3p-RNA-biot and revealed that IFN-I induction was required for the detection of OAS1 and OAS3 and for their binding to RNA in these cells, whereas PKR already bound RNA in the steady-state condition and, in our study, bound 3p-RNA-biot less efficiently than OH-RNA-biot (Fig. 2E).
OAS1, OAS2, and OAS3 synthesize the second messenger 2тА▓-5тА▓ oligoadenylate (2-5A) in response to activation by viral dsRNA (31, 32). To validate that OAS1 is directly activated in a 5тА▓-triphosphateтАУdependent manner, we measured pyrophosphate production by recombinant human OAS1 as a reaction by-product of 2-5A synthesis using a chromogenic assay as previously described (Fig. 2F) (33). Addition of 3p-RNA showed potent activation of OAS1, comparable to a dsRNA carrying a 3тА▓тАУsingle-stranded pyrimidine (3тА▓-ssPy) motif, a known potentiator of OAS1 activation (33), whereas the nonphosphorylated control RNA (OH-RNA) did not show significant levels of OAS1 activity.
2-5A mediates dimerization, and thereby activation, of the latent endoribonuclease (RNase L, encoded by RNASEL) (34, 35). The hallmark of OAS/RNase L activation is the degradation of viral and cellular RNA, including ribosomal RNA (rRNA) (36, 37). To test whether RNase L gets activated in response to 3p-RNA treatment, we studied rRNA integrity in RIG-IтАУdeficient cells. 3p-RNA caused degradation of cellular RNA in IFN-╬▒тАУprimed cells, whereas OH-RNA did not (Fig. 2G). Furthermore, only 3p-RNA transfection in combination with IFN-╬▒ priming led to degradation of rRNA (Fig. 2H), indicating that RNase L can get activated only after IFN-IтАУdependent priming, likely involving recognition of 3p-RNA by OAS.
These results together exclude known 3p-RNA sensors to be responsible for cell death induction. Instead, we reveal a 3p-RNAтАУspecific binding and activation characteristic of OAS1 and show the activation of RNase L after 3p-RNA transfection (Fig. 2, D to H).
The OAS1/RNase L pathway mediates cell death induced by 3p-RNA
To investigate whether the activation of OAS/RNase L is essential for cell death induced by 3p-RNA, we generated RNASELтИТ/тИТ and DDX58тИТ/тИТRNASELтИТ/тИТ cells and assessed RNA degradation after 3p-RNA stimulation, with or without IFN-╬▒ pretreatment (Fig. 3A and fig. S4A). The RNA degradation in WT or DDX58тИТ/тИТ cells upon stimulation with 3p-RNA entirely depended on the presence of RNase L. In addition, RNASELтИТ/тИТ cells were highly resistant to 3p-RNAтАУinduced cell death, which was particularly pronounced at higher RNA concentrations (Fig. 3B and fig. S4B). Cell death assay showed complete resistance of IFN-╬▒тАУprimed DDX58тИТ/тИТRNASELтИТ/тИТ cells over the course of 48 hours of 3p-RNA treatment (Fig. 3C). RNase L has been described to strengthen the IFN-I response through sensing RNA cleavage products by RLRs (38). To rule out a defect in IFN-IтАУmediated priming in RNase LтАУdeficient cells, we investigated a coculture setup of WT, MavsтИТ/тИТ, RnaselтИТ/тИТ, or MavsтИТ/тИТRnaselтИТ/тИТ B16 cells, ensuring equal IFN-I signaling in cells with all genotypes. As expected, MavsтИТ/тИТ cells did not show a significant difference compared with WT cells upon coculture, indicating effective priming (fig. S4C). In contrast, RnaselтИТ/тИТ and MavsтИТ/тИТRnaselтИТ/тИТ cells showed significantly reduced cell death.
(A) rRNA integrity analysis of total RNA isolated from 1205Lu cells with indicated genotypes untreated or treated with IFN-╬▒ (1000 U/ml, 18 hours), transfected with 3p-RNA (12 hours, 160 nM), or treated with IFN-╬▒ (18 hours), and then with 3p-RNA (12 hours). Representative of two independent experiments. (B) Cell death induction in 1205Lu WT and RNASELтИТ/тИТ cells treated with increasing doses of 3p-RNA (40, 80, and 160 nM) for 48 hours (n = 4, StudentтАЩs t test). (C) Kinetic analysis of cell death induction using xCELLigence real-time cell analyzer. WT and indicated KOs in 1205Lu cells were plated on an E-Plate 96 PET with or without IFN-╬▒ (1000 U/ml) for 16 hours and then transfected with 3p-RNA (160 nM). Impedance was analyzed for 48 hours after 3p-RNA transfection. Mean of n = 3. (D) Western blot analysis of 1205Lu DDX58тИТ/тИТ cells overexpressing OAS1, OAS2, or OAS3 or empty vector control with or without 18-hour stimulation with Dox (1 ╬╝g/ml) or IFN-╬▒ (1000 U/ml). Representative of two independent experiments. Relative transgene expression to IFN-╬▒тАУstimulated sample after normalization to loading control is shown below. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (E) CTB assay of cells as in (D) with or without stimulation with either IFN-╬▒ (1000 U/ml) or Dox (0.01, 0.1, and 1 ╬╝g/ml) and treatment with 160 nM 3p-RNA for 48 hours (n = 5, StudentтАЩs t test). (F) Viability of 1205Lu WT and OAS1тИТ/тИТ cells 48 hours after RNA transfection with 160 nM, normalized to untreated (n = 3, StudentтАЩs t test). (G) CTB assay of 1205Lu WT and RNASELтИТ/тИТ cells treated with total RNA (1 ╬╝g/ml) from cells infected with indicated VSV or SeV for 48 hours [n = 7 (CTRL-RNA), 5 (VSV-M51R), and 4 (VSV-WT and SeV), StudentтАЩs t test]. (H) Flow cytometric analysis of viability of WT or RNase LтАУdeficient B16F10 cells 48 hours after transfection (n = 3, StudentтАЩs t test). RNA preparations as described in (G) were used. Unless stated otherwise, means + SEM of independent experiments are shown; *P < 0.05, **P < 0.01, and ***P < 0.001.
We further investigated the importance of OAS-specific up-regulation compared with the induction of other IFN-IтАУstimulated genes and analyzed the individual contribution of OAS1, OAS2, and OAS3 in 3p-RNA sensing. We generated cell lines with doxycycline (Dox)тАУinducible overexpression of OAS1, OAS2, or OAS3 (Fig. 3D) to bypass the need for IFN-╬▒ prestimulation for the up-regulation of these proteins. Subsequent transfection with 3p-RNA or control RNA showed that overexpression of OAS1 or, to a lesser extent, OAS2 was sufficient to induce cell death upon 3p-RNA transfection (Fig. 3E). OAS1 induction in combination with 3p-RNA transfection led to a robust activation of RNase L as monitored by specific rRNA degradation patterns (fig. S4D). In addition to the gain-of-function experiments, we used OAS1-deficient 1205Lu cells to examine the importance of OAS1 in cells that harbor intact RIG-I signaling. OAS1 deficiency caused a strong reduction of cell death upon transfection with 3p-RNA (Fig. 3F).
To expand our results to natural ligands of RIG-I, we isolated total RNA from cells infected with different negative-strand RNA viruses and transfected them into WT or RNase LтАУdeficient cells. RNase L deficiency significantly protected cells from death upon transfection of RNA isolated from cells infected with VSV-WT, VSV-M51R, or Sendai virus (SeV) (Fig. 3, G and H). These data indicate that natural RIG-I ligands similarly induce RNase LтАУdependent apoptosis.
Collectively, these data demonstrate that the OAS/RNase L system triggers cell death in response to synthetic and natural RIG-I ligands. Despite apparent redundancies within the OAS protein family, OAS1 seems to be the main sensor for 3p-RNA upstream of RNase L activation.
3p-RNA induces intrinsic apoptosis
To characterize the pathways leading to cell death in the presence or absence of RIG-I signaling, we analyzed the activation of cell death pathways that are reported to be engaged in response to viral dsRNA. We first measured the activation of caspases, one hallmark of apoptosis activation (39), in response to 3p-RNA treatment by a fluorescently labeled caspase probe. As expected, the activation of caspases was highly dependent on intact RIG-I signaling in the absence of exogenous IFN-╬▒ (Fig. 4A). Prestimulation of cells deficient in RIG-I or MAVS with IFN-╬▒, however, rescued their ability to activate caspases. Using the pancaspase inhibitor z-VAD-fmk (z-VAD), we saw a strong dependence of cell death induction by 3p-RNA on caspases in WT and DDX58тИТ/тИТ cells (Fig. 4B).
(A) 1205Lu cells of indicated genotypes were transfected with 3p-RNA with or without prestimulation with IFN-╬▒ (1000 U/ml) for 18 hours. Caspase-positive, fixable viability dye (FVD)тАУnegative cells were measured using Live Cell Caspase Probe via flow cytometry 30 hours after transfection (n = 3, StudentтАЩs t test). (B) 1205Lu cells were incubated with the pancaspase inhibitor z-VAD (20 ╬╝M, + or 80 ╬╝M, ++) or solvent control for 1 hour before RNA transfection, and cell death induction was measured after 48 hours (n = 2). (C) Western Blots of BAX and BAK expression in WT and RIG-IтАУdeficient cells treated with sgRNAs targeting BAX and BAK1 (sgBAX/BAK) or an irrelevant control (sgCTRL). Representative of two independent experiments. (D) Cell death induction by 3p-RNA treatment (160 nM) 48 hours after transfection in IFN-╬▒тАУprimed DDX58тИТ/тИТ or WT 1205Lu cells (n = 2, StudentтАЩs t test). Means + SEM of independent experiments are shown; *P < 0.05, **P < 0.01, and ***P < 0.001.
In the setting of RLR-induced apoptosis, both intrinsic and extrinsic pathways have been shown to be involved (10, 12, 15). Intrinsic apoptosis is induced by an imbalance of pro- and antiapoptotic B-cell lymphoma 2 (BCL2) family members toward proapoptotic, facilitating BAX/Bcl-2 homologous antagonist/killer (BAK)1 pore formation in the outer mitochondrial membrane and subsequent cytochrome c release to the cytosol. To test the involvement of BAX/BAK in apoptosis induction by 3p-RNA, we targeted genes encoding these proteins in WT and DDX58тИТ/тИТ 1205Lu cells using CRISPR-Cas9 and evaluated the efficiency of KO on protein level by Western blotting (Fig. 4C). Both WT and DDX58тИТ/тИТ cells showed profoundly impaired ability to undergo apoptosis upon 3p-RNA transfection after genetically targeting BAX/BAK (Fig. 4D), with the extent of cell death reduction correlating to the KO efficacy observed in Fig. 4C. Thus, intrinsic apoptosis is the main cell death pathway engaged by 3p-RNA treatment in WT and DDX58тИТ/тИТ cells in vitro.