key: cord-0271058-5jswci3m authors: Aouadi, Wahiba; Najburg, Valérie; Legendre, Rachel; Varet, Hugo; Kergoat, Lauriane; Tangy, Frédéric; Larrous, Florence; Komarova, Anastassia V.; Bourhy, Hervé title: Recognition of copy-back defective interfering rabies virus genomes by RIG-I triggers the antiviral response against vaccine strains date: 2022-03-24 journal: bioRxiv DOI: 10.1101/2022.03.24.485606 sha: 3989ed45c0a364e0f5d2694309b390e701f7ddbb doc_id: 271058 cord_uid: 5jswci3m Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year around the world. A typical feature of RABV infection is the suppression of type I and III interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) to initiate IFN signaling remain elusive. Here, we showed that RABV RNAs are recognized by RIG-I (retinoic acid-inducible gene I) sensor resulting in an IFN response of the infected cells but that this global feature was differently modulated according to the type of RABV used. RNAs from pathogenic RABV strain, THA, were poorly detected in the cytosol by RIG-I and therefore mediated a weak antiviral response. On the opposite, we revealed a strong interferon activity triggered by the RNAs of the attenuated RABV vaccine SAD strain mediated by RIG-I. Using next-generation sequencing (NGS) combined with bioinformatics tools, we characterized two major 5’copy-back defective interfering (5’cb DI) genomes generated during SAD replication. Furthermore, we identified a specific interaction of 5’cb DI genomes and RIG-I that correlated with a high stimulation of the type I IFN signaling. This study indicates that RNAs from a wild-type RABV poorly activate the RIG-I pathway, while the presence of 5’cb DIs in vaccine SAD strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection and therefore strongly stimulates the IFN response. Highlights RABV pathogenic strain replication in vitro is characterized by the absence of defective interfering genomes thus induces a weak RLR-mediated innate immunity antiviral response. RABV vaccine attenuated strain shows a high release of 5’ copy-back defective interfering genomes during replication in vitro and therefore enhances a strong antiviral response upon infection. RIG-I is the main sensor for RABV RNA detection within cells. • RABV pathogenic strain replication in vitro is characterized by the absence of defective 17 interfering genomes thus induces a weak RLR-mediated innate immunity antiviral 18 response. 19 • RABV vaccine attenuated strain shows a high release of 5' copy-back defective 20 interfering genomes during replication in vitro and therefore enhances a strong antiviral 21 response upon infection. 22 • RIG-I is the main sensor for RABV RNA detection within cells. To explore in more detail the differential involvement of RLR (RIG-I or MDA5) in RABV 160 RNA sensing, the ISRE reporter cell line was treated with non-targeting siRNAs (si-Neg 161 control), targeting RIG-I (si-RIG-I) or MDA5 (si-MDA5). Transient silencing of RIG-I and 162 MDA5 significantly (p ≤0.001, one-way ANOVA using Tukey method) reduced the level of 163 mRNA for RIG-I and MDA5 by ~61% and ~82%, respectively as compared to si-Negative 164 control cells ( Figure 1C ). When the same cells were transfected with RABV RNAs (THA or 165 SAD), only RIG-I silenced reporter cells showed strongly impaired ISRE promoter activation 166 (p <0.0001, one-way ANOVA using Tukey method), while MDA5 silencing did not affect the 167 signaling ( Figure 1D ). 168 Altogether, these results demonstrate that RIG-I is the main cytosolic PRR that detects RABV 169 RNAs to mediate IFN signaling. Then, RNAs co-purified with RIG-I, MDA5, or CH were extracted and transfected into the 192 ISRE reporter cell line to assess the activation of type-I IFN signaling. First, the ISRE promoter 193 activation was first controlled by transfecting synthetic RNAs. As expected, 5'3P, HMW 194 poly(I:C), and LMW poly(I:C) largely stimulated ISRE expression ( Figure 2B ). These data 195 validated our transfection approach. RIG-I-specific RNAs extracted from THA-infected cells 196 induced a light but significant (p=0.029, single-step method) stimulation of ISRE expression as 197 compared to the negative control CH-specific RNAs ( Figure 2C ). RIG-I-specific SAD RNA 198 induced a strong and significant (p=0.00007, single-step method) ISRE promoter activity in a 199 dose-dependent manner as compared to negative control CH-specific RNAs ( Figure 2D ). For 200 both viral strains, MDA5-specific RNAs showed no significant stimulation of the ISRE 201 promoter activity. These data suggest that RNA extracted from THA and SAD infected cells 202 present molecular patterns that are absent in non-infected cells and that are preferentially 203 recognized by RIG-I. 204 205 To further characterize RLR bound RABV RNAs, ST-RLRs cells were infected with THA or 207 SAD at MOI of 0.5. The cells were harvested after 24h, and RLRs bound RNAs were purified 208 using affinity chromatography. Total RNAs from input cell extract, as well as RLRs-bound 209 RNA output, were extracted. Input and output RNAs were subjected to NGS followed by 210 bioinformatics analysis using the previously described protocol (Chazal et al., 2018). NGS 211 provided high output in the order of 12 to 90 million total reads per sample with around 0.3 and 212 0.29% mapped to THA and SAD genomes, respectively (Table S1 ). The fold change of RLR 213 binding was obtained by normalizing: i) mean abundance of reads (coverage) of either RIG-I 214 or MDA5-ligands by mean read coverage of corresponding input samples, ii) nonspecific RNAs 215 binding were discarded by normalizing RLRs read coverage to reads obtained with cherry 216 negative control. NGS data analysis revealed no visible enrichment of read-abundance in RLR 217 association of negative-and positive-sense viral RNAs upon THA infection in our experimental 218 conditions ( Figure 3A) . Interestingly, the NGS study of the RABV vaccine strain, SAD, showed 219 a significant coverage and much higher enrichment on MDA5 of negative-sense RNA along 220 the whole viral genome than with positive-sense viral RNA. Thus, MDA5 may be engaged in 221 recognition of the SAD negative-sense genome. Conversely, studies analyzing RNA bound to 222 MDA5 in measles virus-infected cells (also negative-sense single-stranded RNA virus) 223 reporting that MDA5 interacts with only positive-strand viral RNA (Runge et al., 2014; 224 Sanchez David et al., 2016) . However, despite viral RNA/MDA5 interaction, we failed to 225 observe any statistically significant stimulation of ISRE promoter in reporter cells in our 226 experimental conditions ( Figure 2D ). 227 Conversely, the analysis of RIG-I-specific RNA ligands purified from SAD-infected cells 228 revealed a significant enrichment of viral negative-and positive-sense RNAs (Figure 3B To further explore the RNA primary sequence motifs of RLR ligands that could explain SAD 237 RNA recognition by RIG-I and or MDA5, we analyzed the AU content of RIG-I and MDA5 238 specific reads as described previously (Sanchez David et al., 2016). We observed that MDA5 239 binding to SAD negative-strand genomic RNAs correlated with high AU content (> AU content 240 of SAD genome 0.55) (p<0.001, Cohen's d=0.53) ( Figure S4D ). However, RIG-I SAD 241 negative-strand ligands were characterized by a lower AU-rich content sequence than that 242 observed for MDA5 ligands suggesting that RIG-I binding did not correlate with RNA primary 243 sequence ( Figure S4B ). For the positive-strand SAD RNA species, we did not detect any 244 preference of binding of AU-rich regions by RIG-I or MDA5 ( Figure S4A and C). In 245 conclusion, our results suggest preferential binding of MDA5 to AU-rich regions of viral RNA 246 but RIG-I ligand interaction to AU-rich sequence was not observed suggesting that RIG-I more 247 presumably recognizes specific RNA secondary or tertiary structures. Table 1 ). 5'cb DI-1668 exhibited statistically significant (p =0.04, one-way ANOVA) read 258 abundance in the RIG-I output samples as compared to the negative control ST-CH. We failed 259 to detect any statistically significant read enrichment for 5'cb DI-2170 in ST-RIG-I output 260 samples comparing to the negative control ST-CH cells (Table 1) . 261 We further studied molecular organization of the detected 5'cb DI-2170 and 5'cb DI-1668 DI 262 viral genomes and juxtaposed them on the Figure 3B Total RNA was extracted using a RNeasy minikit (Qiagen). cDNA was generated from 2 g of 403 total RNA by using Superscript IV vilo (#11766050, Invitrogen) and primer 2 (Table S2) The experiments were performed three times and represented as median with 95% confidence 664 interval. P values were calculated using one way ANOVA with Tukey's multiple comparisons. 665 Non-significant (n.s.) were indicated. *p<0.05, **p< 0.01, ***p<0.001, ****P<0.0001 666 667 curves. Black and blue arrows depicted the breakpoints, BP, and reinitiation sites, RI, for 5' 690 cb DI-2170 and 5'cb DI-1668, respectively. 691 The fold change of RLR binding is obtained by i) normalizing the read coverage of output 692 RNAs co-purified with RIG-I, MDA5 or CH by their input extract, ii) normalized read 693 coverage of output samples were divided by the mean of the coverage of negative control CH, 694 at each genomic position. 695 TGATATCCTCTAGGGTGATCCACCTGTTGTACCTATGAAGGTGTCTTTCCACTTCTCT GGATAGATCCTTGATGCTGCCAACGAGTCTGGTAGTCTTCACTATCTTGTAAATCAA CCTGATCCAGTGAGATGACAGACTCAGGCTAGAATTACAGGCTACCCTTTTGAACAC TGAGGTGTCGTTGGACCAACTCCAAGATAGATAAACGTTCCCTCGAATGACTTGCTT TCTGAAGTAATAAGTTATAGGTCTGTTATACAGGTCGTGAAGTCTTGCGAAGCTAGG GACGTCACCTAAAGCAGTAGATAGATACATCATAGTACTGCAACATATGTTGAAGT GCCTCAGGATTTTGGGATCAGACGGTGGATAGAACGAGGGGTCAGTTAGGGGTTTG GAAACGTTGAAGACTCTGTTGGAGAAAACTATCATGATGGCTATAATGATAGCCAC TTTAGACAGAGTTCCCCTCTGTAACTCAAGATCATCAACCATCTTGTGGACTAGAAA AGATTCTACATCACTGTCAATCAGAGTTATGATCATCTCATTGTAAGGATTTGATAT GATTTCTTCAGGAAATCCTCTCACAAGATCCTGATAGTTCAAGGAACGAGCTCTCTG CATCTCACTCTTGGGGCTGCTACAATTGAATAACACAAGGCTCATTTCTCGAAGGGT GGAAGAGGTCAAGTACTCAGCATCTCTGAAAAACTTCCCTCGTTTGGAGAATCGGA GGTAGAGCTCAGATGAAAAAGACGAAGTTACTTGGGTGATAAACCCTGTGACCGAG GGGAACGCTCTTGACAGGTGTTGAATAGCCTTGTAGTTTGGATTTACTAGCATAGTC CCATAAGTTTTGAAGACCAAATAGAGTGGTCCATCTATAGACAATGCAAAATCGGA CATTAACAGGGTGATCCGGTTGATAGATGCAATGTCAGTAACTTCTGCATCGCAAAT AATGAGGTCATAGGACATGTTGACCTGCTTTTGGACTGACTGGAAGTATTTCCAGGT TGCCAAGTTTCTCAAGTCGGACGGTTTTTCCCAGATTGAGTCAAGATCTATCACTCT GGAGACGATATCATTTCCTCCCCTCATGATTGCTGAAGGAGGCAGTGGATGTGTTCC GGAAGCCATCAGGTCATTCACCTCTAAAAGACTGTTGAACACAAGCTTGGCATCTG GAAACATGTTGAGGACTGCCCTTGATATCCCCCCTGACCCGTCCCCAACTACAAGGC AGAGAGATGGGAAAACATTGAGATCATCTAGAATAGGCTTAAGCTTATAATGAGCA CCGGTTGCCCACTGAACCACTCTCAAGCCCGAGATCAAAGGGTTCTGGAACCTCTTA GAGAGGGCCCTTATGTCAAGCTCCGAGACAGGGGCCGGGTTTGCTGAGGTAGAGAC TGCAACCTGTTGAGCAGAGCAGACCCATTCTGAACATCCTACCTTACGGGACACCTT CTTGTTGGGGCTGTAATCTCCAGTCATGGTTCTAGCTGCATGGCGCACCTCTTGATCC ACCCATCTTGTCCTTCGTAAAGAGTCTTTTAGCAGTCGTTGAATGTTGTCGTCTGACT CTAAGGTATCTTCTCCGTGCCCGCCCAGCACCTGCCTCATCAAAGAACTCAATTGTC GCAGGTTATCTCTCATACTCTTAGATAGGTTTCTCTCAACCCTCTGGAGTAGAAGAT GAGACTGGTAAGTAATGAGGGATAGGTACGTCATTTTGGCACTTCTAAAGTCTGAA AAGATCCATAGCCAGTCATTCTCTGGAGACGCCGTGATTATCTCTCGCTCATAGCGT AGCACATGTTGGAGATAACACAAGATTGATCTGTTGCCTTCTTTCATAGTGGTTGGA TAAGCGGCGGGGATTTTCTGAGGGATAGAAAATATCTCTCCTCTAAGAGACGGTTCT Primers Sequence 1 GAGGGGAACGCTCTTG 2 CGCTTAACAAATAAACAACAA 3 AGA AAG CAA GTC ATT CGA GGG 4 GAGGGGAACGCTCTCG 5 CGCTTAACAAAAAAACAATAA ACGCTTAACAAATAAACAACAAAAATGAGAAAAACAATCAAACAACCAAAGGTTCAGATTT AGGATCTTGTTTTTTTCAAGATACATCACACAAGAGTCTTAGCATGGGCAGGCTTCCAGGAGT ATCCGGTTCACAGGCAACTGTAGTCTAGTAGGGATGATCTAGATCTGATATCCTCTAGGGTGA TCCACCTGTTGTACCTATGAAGGTGTCTTTCCACTTCTCTGGATAGATCCTTGATGCTGCCAAC GAGTCTGGTAGTCTTCACTATCTTGTAAATCAACCTGATCCAGTGAGATGACAGACTCAGGCT AGAATTACAGGCTACCCTTTTGAACACTGAGGTGTCGTTGGACCAACTCCAAGATAGATAAA CGTTCCCTCGAATGACTTGCTTTCTGAAGTAATAAGTTATAGGTCTGTTATACAGGTCGTGAA GTCTTGCGAAGCTAGGGACGTCACCTAAAGCAGTAGATAGATACATCATAGTACTGCAACAT ATGTTGAAGTGCCTCAGGATTTTGGGATCAGACGGTGGATAGAACGAGGGGTCAGTTAGGGG TTTGGAAACGTTGAAGACTCTGTTGGAGAAAACTATCATGATGGCTATAATGATAGCCACTTT AGACAGAGTTCCCCTCTGTAACTCAAGATCATCAACCATCTTGTGGACTAGAAAAGATTCTAC ATCACTGTCAATCAGAGTTATGATCATCTCATTGTAAGGATTTGATATGATTTCTTCAGGAAA TCCTCTCACAAGATCCTGATAGTTCAAGGAACGAGCTCTCTGCATCTCACTCTTGGGGCTGCT ACAATTGAATAACACAAGGCTCATTTCTCGAAGGGTGGAAGAGGTCAAGTACTCAGCATCTC TGAAAAACTTCCCTCGTTTGGAGAATCGGAGGTAGAGCTCAGATGAAAAAGACGAAGTTACT TGGGTGATAAACCCTGTGACCGAGGGGAACGCTCTTGACAGGTGTTGAATAGCCTTGTAGTT TGGATTTACTAGCATAGTCCCATAAGTTTTGAACTCTGTCTAAAGTGGCTATCATTATAGCCA TCATGATAGTTTTCTCCAACAGAGTCTTCAACGTTTCCAAACCCCTAACTGACCCCTCGTTCTA TCCACCGTCTGATCCCAAAATCCTGAGGCACTTCAACATATGTTGCAGTACTATGATGTATCT ATCTACTGCTTTAGGTGACGTCCCTAGCTTCGCAAGACTTCACGACCTGTATAACAGACCTAT AACTTATTACTTCAGAAAGCAAGTCATTCGAGGGAACGTTTATCTATCTTGGAGTTGGTCCAA CGACACCTCAGTGTTCAAAAGGGTAGCCTGTAATTCTAGCCTGAGTCTGTCATCTCACTGGAT CAGGTTGATTTACAAGATAGTGAAGACTACCAGACTCGTTGGCAGCATCAAGGATCTATCCA GAGAAGTGGAAAGACACCTTCATAGGTACAACAGGTGGATCACCCTAGAGGATATCAGATCT AGATCATCCCTACTAGACTACAGTTGCCTGTGAACCGGATACTCCTGGAAGCCTGCCCATGCT Differential recognition of viral RNA by RIG-I Antiviral Response Importance of rabies virus nucleoprotein in viral evasion of interferon response in 575 the brain MDA-5 recognition of a murine norovirus Nonencapsidated 5'Copy-Back Defective Interfering Genomes Recombinant Measles Viruses Are Recognized by RIG-I and LGP2 but Not MDA5 Measles Virus C Protein Impairs Production of Defective Copyback Double-Stranded Viral RNA and Activation of Activation of MDA5 Requires Higher-Order RNA Structures Generated during Virus Infection BEDTools: A flexible suite of utilities for comparing 588 genomic features RIG-I-like receptors: their regulation and roles in RNA 590 sensing RIG-I detects viral genomic RNA during negative-strand 593 RNA virus infection Dissection of Interferon-Antagonistic Functions of Rabies Virus Phosphoprotein : Inhibition of 596 Interferon Regulatory Factor 3 Activation Is Important for Pathogenicity In Vivo Ligands of MDA5 and RIG-I in Measles 599 Comparative analysis of viral RNA 602 signatures on different RIG-I-like receptors LGP2 binds to PACT to 605 regulate RIG-I-and MDA5-mediated antiviral responses Recognition of 5′ Triphosphate by RIG-I Helicase Requires Short Blunt Double-Stranded RNA as Contained in Panhandle of Negative-609 A conserved histidine in 612 the RNA sensor RIG-I controls immune tolerance to N1-2'O-methylated self RNA Differential Type I IFN-Inducing Abilities of Wild Type versus Vaccine Strains of Measles Virus Lyssavirus matrix protein cooperates with phosphoprotein to modulate the Jak-Stat 619 pathway Sendai virus defective-interfering genomes 621 and the activation of interferon-beta Immunostimulatory Defective Viral Genomes from 624 Respiratory Syncytial Virus Promote a Strong Innate Antiviral Response during Infection in 625 Mice and Humans Reduced accumulation of 628 defective viral genomes contributes to severe outcome in influenza virus Attenuated Rabies Virus Activates, while Pathogenic Rabies Virus Evades, the Host Innate Immune Responses in the Central Nervous System Conservation of a Unique Mechanism of Immune 635 Evasion across the Structural basis for dsRNA recognition, filament formation, and antiviral signal 638 activation by MDA5 MDA5 Governs the Innate Immune Response to Defective Interfering Particles of Negative-Strand RNA Ribose 2′-O-methylation provides 646 a molecular signature for the distinction of self and non-self mRNA dependent on the RNA 647 sensor Mda5 Detection of 5'cb DI-1668, 5'cb DI-2170 (left panel), and SAD genome ST-RIG-I, and ST-699 MDA5 cells (MOI of 0.5, 24h post-infection). RT-PCR was performed using universal 700 5' cb primers (1 and 2) and full-length genome primers SAD and THA genomes (right 702 panel) by RT-PCR on total RNAs extracted from infected neuroblastoma cells 703 (SK.N.SH) at MOI of 0.5 for 24h using universal 5' cb primers (1 and 2) and full-length 704 genome primers on 2 µg of total RNAs extracted from SAD or THA infected neuroblastoma 706 cells (SK.N.SH) at MOI of 0.5 for 24h using THA-optimized 5' cb DI primers (4 and 707 5) (left panel) and THA-optimized full-length genome primers In silico analysis of RLR-specific SAD ligands: 767 AU content of specific RLR RNAs partners (A, B) for RIG-I and (C, D) for MDA5. Relation 768 between AU content and mean count for each position We acknowledge all the members of the Lyssavirus, Epidemiology and Neuropathology unit 489 and the Vaccines-innovation laboratory at Pasteur Institute for their help and useful discussions.