Introduction

In the last ten years, novel duck reovirus (NDRV) has been identified in various duck species in China1. Unlike classical DRV, NDRV can cause severe liver and spleen hemorrhage in ducklings, resulting in a high mortality rate of 50%2, which can cause incalculable economic losses to the waterfowl industry in China. Unfortunately, there is currently no commercial vaccine available to prevent NDRV infection and transmission. As a result, understanding the pathophysiology of NDRV in ducklings will aid in the development of new treatments and vaccines against NDRV.

NDRV has been shown to attack the immunological system (bursa of Fabricius) of Muscovy ducklings. Most studies on NDRV have focused on the pathological phenotype of the bursa of Fabricius. On this basis, our team has previously provided a large amount of high-throughput transcriptome and proteome data3,4. From these studies, we found that the nucleic acids carried by the virus can not only influence the viral structure and function but also react with the endogenous miRNAs of the duckling.

In the present study, we used a virulent NDRV strain named HN10 to infect the bursa of Fabricius in Muscovy ducklings. Then, we focused on translation efficiency via combined RNA-seq, Ribo-seq, and m6A MeRIP-seq analyses, and we further investigated the molecular mechanism underlying the influence of NDRV on protein translation regulation in the bursa of Fabricius cells. Our findings could advance our understanding of the molecular biology involved in NDRV infection in ducklings, and this cross-species response may be generalized to other virus-mammal interactions, potentially providing novel ideas and clues for the study of virus evolution, prevention, and treatment.

Results

The novel duck reovirus weakens gene translation in the bursae of Fabricius

The bursa of Fabricius of Muscovy ducklings was infected with the novel duck reovirus (NDRV) HN10, and RNA-seq data were used in our previous research4. Ribo-seq and m6A MeRIP-seq were subsequently conducted to investigate the ribosome-enriched and m6A-modified transcriptomes. The global abundance (transcripts per million, TPM) of ribosome-enriched mRNAs without NDRV-derived transcripts was significantly lower than that in the NC group (p = 1.84 × 10−12) after normalization (Fig. 1a). By assessing the translational efficiency index (TEI, defined as the relative level of translation to transcription) combined with previous RNA-seq data, we observed that the proportion of RNA showing the same TEI value after HN10 infection was much lower than that of uninfected RNA (p = 3.7 × 10−15), which indicated that NDRV HN10 drastically reduced translation more than transcription after eliminating the disturbance of viral RNA (Fig. 1b). Moreover, we further noted that the significantly upregulated m6A-modified mRNAs strongly overlapped (64.8%) with the downregulated ribosome-enriched mRNAs in the NDRV HN10 infection group compared to the negative control group (Fig. 1c).

Fig. 1: NDRV affects the gene translation of the bursa of Fabricius in Muscovy ducklings.
figure 1

a Normalized TPM of RNA-seq and Ribo-seq of the bursa of Fabricius of Muscovy ducklings after NDRV HN10 infection. The significant difference in abundance between transcripts and ribosome-enriched transcripts of NDRV HN10 infection is indicated (p = 1.84 × 10−12) by one-way ANOVA. b Translational efficiency of the Muscovy duckling genes in the control group as well as the Muscovy duckling genes and NDRV transcripts in the HN10 infection group. The TEI was determined as the log2 ratio of Ribo-seq to RNA-seq data. A significant difference in the TEI between the infection and control groups is indicated (p = 3.7 × 10−15) by a two-tailed Wilcoxon rank-sum test. c Overview of genes with differential m6A modifications and differential ribosome enrichment (log2 FC > 1 or <−1, P < 0.05). d, e Firefly luciferase reporter gene assay showing the effect of σB on the luciferase activity of duck embryo fibroblast CCL-141 cells by CHX (d) or BTZ (e). BTZ + NDRV HN10, 2-h BTZ treatment followed by NDRV HN10 infection; NDRV HN10 + BTZ, 10 h NDRV HN10 infection followed by BTZ treatment. Luciferase activity was normalized to the BSA-measured total protein concentration of each sample. f, g qPCR indicated the transcripts of exogenous gene were normalized by β-actin (f). WB assay showed the β-lactamase expression from pSecTag plasmid (g). h Firefly luciferase reporter gene assay showing the effect of 12 NDRV HN10 transcripts on the luciferase activity of DEFs. Luciferase activity was normalized by the 2−ΔΔCt value of each transcript (Flag for EV) by qPCR. i Translational efficiency of the Muscovy duckling genes in DEFs transfected by EV, λA, σA and σB respectively. Significant differences in the TEI between the infection and EV groups are indicated by a two-tailed Wilcoxon rank-sum test. *indicates statistical significance compared to the NC group, with a P value less than 0.05 according to the student’s t-test. NC untreated negative control, EV empty vector, TPM transcripts per million, RNA-seq RNA sequencing, Ribo-seq ribosome footprint profiling sequencing, TEI translational efficiency index, FC fold change, CHX cycloheximide, BTZ bortezomib, DEFs duck embryo fibroblast CCL-141 cells.

To further clarify whether HN10 inhibited transcriptional efficiency or promoted protein degradation, we infected DEFs stably transfected with the CMV-driven firefly luciferase reporter with NDRV HN10 and treated them with the protein synthesis inhibitor cycloheximide (CHX) or the proteasome inhibitor bortezomib (BTZ). Compared with those of the NC, the effects of NDRV HN10 and CHX on reducing luciferase activity were indeed observed (Fig. 1d). On the other hand, the BTZ-induced fluorescence signal could be compromised by subsequent NDRV HN10 infection, whereas BTZ failed to rescue the prior effect of NDRV HN10 (Fig. 1e), indicating that the decrease in luciferase activity observed following NDRV HN10 infection is not connected to proteasomal activity or protein degradation. We divided the twelve coding proteins of NDRV HN105 into individual plasmids and transfected them into DEFs to examine the effect on gene translation (Fig. 1f, g). We observed that only σB had a remarkable toxic effect compared with the other components (Fig. 1h). Ribo-seq also indicated that σB could obviously unload mRNA from ribosomes compared to empty vector or other NDRV HN10 proteins (Fig. 1i). Overall, we concluded that NDRV HN10 might terminate the gene translation process in the bursa of Fabricius.

Characterization of σB-interacting proteins in Muscovy ducklings

To further investigate the mechanism by which NDRV HN10 affects gene translation, transient Flag-σB (with Flag-λA as a control) transfection was conducted in subsequent studies. Flag IP was conducted to capture the interacting proteome, which was further characterized by mass spectrometry. A series of RNA binding proteins showing higher homology with those of Homo sapiens were identified in the list of peptides with the 40 highest confidence levels compared to λA (Fig. 2a, Supplementary Data 1). GO analysis revealed that these proteins were largely related to RNA metabolism and translation (Fig. 2b). We unexpectedly noticed that the m6A methylation readers YTHDF1 and YTHDF3 could interact with σB. Moreover, the stress granule components CAPRIN1 and G3BP1 could also be pulled down by σB. WB analysis of YTHDF1, YTHDF3, CAPRIN1, and G3BP1 to validate the interaction with σB was performed with commercial antibodies against these targets of Muscovy ducklings against the same immunogen across Homo sapiens (Fig. 2c).

Fig. 2: σB interacts with YTHDF1, YTHDF3, CAPRIN1 and G3BP1.
figure 2

a Binding protein profiles of σB compared to λA. Confident candidate proteins were adjusted by the following conditions: log2 FC > 1 or <−1 and FDR < 0.05. b Metascape was used to analyze the function of these candidate proteins (Supplementary Data 1) with p values less than 0.05. c IP-WB shows the substantial interaction of YTHDF1, YTHDF3, CAPRIN1, and G3BP1 pulled down by Flag (σB) compared to that pulled down by Flag (λA). IP immunoprecipitation, WB Western blot.

σB facilitates the assembly of the m6A reader complex and stress granules

Immunocytochemistry showed a remarkable colocalization of G3BP1 and YTHDF1 with σB compared to λA and the empty vector (EV) (Fig. 3a, b) in DEFs treated with sodium arsenite (transfection of a single component cannot mimic NDRV HN10 infection to create stress conditions). Gel filtration showed that the protein fractions containing the highest abundances of YTHDF1/3, G3BP1, CAPRIN1, and σB were identical after σB transfection (Fig. 3d) but staggered after λA transfection (Fig. 3e) compared to EV control (Fig. 3c), suggesting that the m6A methyltransferase complex YTHDF1/3 was assembled with G3BP1/CAPRIN1 by σB. Similarly, G3BP1/CAPRIN1 were also colocalized in the bursa of Fabricius after NDRV HN10 infection compared to that in the NC group in vivo (Fig. 3f, g). Transmission immunoelectron microscopy verified that the G3BP1 (large dots) and YTHDF1 (small dots) proteins were more closely related to each other in response to HN10 infection (Fig. 3h) than in response to the negative control (Fig. 3i). Previous studies have indicated that both G3BP1 and CAPRIN1 are the core members involved in the organization of SGs6,7,8. Therefore, σB of NDRV HN10 might affect the gene translation of Muscovy ducklings by promoting stress granule formation.

Fig. 3: σB is required for the formation of YTHDF1/3 and stress granules.
figure 3

ICC shows the colocalization of σB and G3BP1 (a) or YTHDF1 (b) compared to that of EV and λA in duck embryo fibroblast CCL-141 cells. Images are shown at 80 × 10 magnification. Scale bars, 20 μm. ce GF-WB image showing the coexpression (highlighted by the red frame) of YTHDF1, YTHDF3, CAPRIN1, G3BP1, and σB within the same protein fraction (d) compared to that of EVs (c) and λA (e). The numbers marked are the samples collected in sequence. f, g ICC showed increased expression of CAPRIN1 and G3BP1 in the bursa of Fabricius in Muscovy ducklings infected with NDRV HN10 (f) compared to NC (e) in vivo. Images are shown at 4 × 10 (left) and 40 × 10 (right) magnification. Scale bars, 20 μm. h, i Immunogold-labeled TEM image showing the colocalization of G3BP1 (large dots) and YTHDF1 (small dots) in the bursa of Fabricius in Muscovy ducklings infected with NDRV HN10 (h) compared to those infected with NC (g) in vivo. Scale bars, 200 nm (left) and 10 nm (right). ICC immunocytochemistry, GF gel filtration, WB Western blot, TEM transmission immunoelectron microscopy, EV empty vector, NC negative control.

Stress granule packages of RNA with m6A modification at the 5′ UTR

To further investigate the role of stress granules in the response to NDRV HN10 infection, we conducted RIP-seq to examine the G3BP1-binding transcripts included in stress granules in vivo. Among the four populations with differential G3BP1- or m6A-enriched genes, 73.5% of the highly G3BP1-enriched transcripts overlapped with upregulated m6A-modified RNA (Fig. 4a, Supplementary Data 2). GO analysis indicated distinct functions and pathways of these four populations of genes (Fig. 4b). Furthermore, the m6A signal was primarily distributed at the 5′ UTR of these RNA molecules (Fig. 4c). Consistently, σB and λA RIP-seq of DEFs treated with sodium arsenite also suggested that 94.7% of the G3BP1-binding RNAs in vivo were σB-specific RNA molecules in vitro (Fig. 4d, Supplementary Data 3). Compared to λA-binding RNAs, σB-binding RNAs also exhibited robust m6A modifications at the 5′ untranslated region (UTR) according to in vivo mRIP-seq data (Fig. 4e). These results indicated that robust RNA with m6A modification was packaged into SGs by NDRV HN10 infection.

Fig. 4: G3BP1 is largely occupied by RNA with m6A modification by σB of NDRV HN10.
figure 4

a Overview of four populations of genes with differential G3BP1 occupancy and m6A modification (log2 FC > 1 or <−1, P < 0.05) represented by different colors. The “73.5%“ is highlighted for the transcripts with high G3BP1 enrichment and m6A modification by NDRV HN10 compared to NC. b Heatmap showing the top 10 activity scores of GO analysis in these four populations ranked by t values. K-S test-like rank statistics of the gene set in each pathway were calculated, and the expression matrix was converted into a pathway enrichment score matrix to obtain the activity score. The difference in each population’s activity score compared to all other populations was analyzed using the “limma” package, and t values of the top 10 terms with P values less than 0.05 were used to construct the heatmap. The four colors indicate the same population as in (a). c m6A distribution of the RNAs indicated by the same four colors in (a). RNAs with upregulated enrichment of G3BP1 and m6A in NDRV HN10 compared with NC had high m6A modification at the 5′ UTR. d Venn diagram showing the intersection between G3BP1-enriched RNAs in vivo and Flag (σB)-enriched RNAs in vitro. e m6A distribution of σB- and λA-enriched RNAs. RNAs with σB occupancy have high m6A modification at the 5′ UTR. NC negative control, FC fold change, UTR untranslated region, CDS coding sequence.

σB/YTHDF1/3 links stress granules with m6A-enriched RNA

Finally, we investigated the role of σB in bridging stress granules and m6A-modified RNA. YTHDF1/3, two σB-binding proteins (Supplementary Data 1), play a role as m6A readers in eukaryotic cells9,10. YTHDF1 or YTHDF3 were silenced by siRNA in advance (Fig. 5a), and σB was subsequently transfected into DEFs treated with sodium arsenite. The results of the RIP-qPCR analysis of the two genes with the greatest increases in m6A modification in NDRV HN10 compared to those in the negative controls Reg4 (log2 = 10.65, P < 0.05) and LOC113843712 (log2 = 8.38, P < 0.05) are shown in Fig. 4b. We observed that the abilities of σB (Fig. 5b, c) and G3BP1 (Fig. 5d, e) to capture the transcripts of these two genes were not affected by YTHDF1 or YTHDF3 knockdown alone but were weakened when YTHDF1/3 was simultaneously knocked down. Similarly, simultaneous YTHDF1/3 knockdown impaired the interaction between σB and G3BP1/CAPRIN1 (Fig. 5f). Finally, both YTHDF1/3 knockdowns reduced the translation of endogenous genes in DEFs to some extent via HN10 infection (P = 2.8 × 10−5) (Fig. 5g).

Fig. 5: YTHDF1/3 determine σB-mediated translational shutoff.
figure 5

a WB was used to verify the efficiency of siRNA-mediated knockdown. b, c RIP-qPCR shows that the occupancy of Flag (σB) on the transcripts of Reg4 (b) and LOC113843712 (c) was reduced by YTHDF1/3 knockdown. d, e RIP-qPCR showed that the occupancy of G3BP1 on the transcripts of Reg4 (d) and LOC113843712 (e) was reduced by YTHDF1/3 knockdown. * indicates statistical significance compared to the NC group, with a P value less than 0.05 according to the student’s t-test. f IP-WB indicates that the substantial interaction between G3BP1 and CAPRIN1 was affected by both YTHDF1 and YTHDF3 knockdown. g The translational efficiency of the Muscovy duckling genes was reduced by YTHDF1/3 knockdown. A significant difference in the TEI between YTHDF1 knockdown (green) or YTHDF3 knockdown (purple) and NDRV HN10 knockdown (yellow) cells is indicated (YTHDF1, p = 2.8 × 10−5; YTHDF3, p = 1.9 × 10−6) by a two-tailed Wilcoxon rank-sum test. NC negative control, KD knockdown by siRNA, 1 KD YTHDF1 knockdown by siRNA, 3 KD YTHDF3 knockdown by siRNA, 1 and 3 KD both YTHDF1/3 knockdown by siRNA, TEI translational efficiency index.

Our study revealed that σB of NDRV HN10 could bind with YTHDF1/3 and G3BP1 and then recruit m6A-modified RNA to stress granules for translation shutoff of the endogenous gene of Muscovy ducklings (Fig. 6).

Fig. 6: The working model in this study.
figure 6

When HN10 NDRV infects cells, σB, a viral particle protein, interacts with the m6A methylation recognizer YTHDF1/3 and recruits stress granules to encapsulate 5′-end m6A-methylated RNAs, abolishing their translation. On the other hand, HN10 NDRV RNAs will make full use of intracellular ribosomes to translate viral proteins.

Discussion

Viruses have evolved a variety of ways to manipulate the cellular translation machinery, emphasizing viral translation over normal protein synthesis. These mechanisms include the use of specific molecular markers on viral mRNA, such as internal ribosome entry sites (IRESs) and genome-linked viral proteins (VPgs). Furthermore, virus-encoded factors can quickly transform the global landscape of posttranscriptional processes by coopting and subverting cellular pathways that control mRNA degradation, modification, and translation. These processes facilitate viral translation through unconventional interactions with translation machinery components and by promoting RNA circularization11,12. In contrast, in response to viral infections, host cells typically adopt concurrent cell-intrinsic countermeasures that use posttranscriptional mechanisms to activate key innate immune responses13.

In addition to the overall pathological changes caused by viral infection, the structural and functional protein components of NDRV have many other functions that are gradually being deciphered during the infection process in waterfowl. For example, σNS can promote autophagy14, and p10.8 induces ER stress and apoptosis15. The outer capsid protein of NDRV is predominantly composed of the S3-encoded protein σB, which is functionally identical to the σ3/σB proteins of mammalian reovirus (MRV), avian reovirus (ARV), and  classical duck reovirus (CDRV). The NDRV σB protein has approximately 60% and 70% amino acid homology with ARV and CDRV, respectively. It possesses group-specific neutralizing epitopes and induces a robust antibody response against the virus. The NDRV σB protein has conserved sequence properties and is closely related to virus infection, pathogenicity, and immunogenicity, making it a promising option for NDRV serodiagnosis5.

In response to external stressors such as heat, nutrient deficiency, oxidative stress, UV radiation, and viral infection, eukaryotic cells usually activate stress response mechanisms16, resulting in changes in gene transcription, epigenetics, and mRNA translation. Under such harsh conditions, the cytoplasm forms stress granules, which are mRNA‒protein complexes17. The translation of most mRNAs is temporarily halted, and mRNAs, translation regulatory factors, and other RNA-binding proteins are transported from ribosomes to stress granules18. Limited energy resources are available for the translation of stress proteins (e.g., heat shock proteins) that aid the cell in overcoming stress. The translation of nonessential mRNA is temporarily shut down but not degraded, allowing for the resumption of translation when conditions return to normal. The stress granules function as a “safe haven” for these RNA molecules19,20.

Stress granules are non-membrane-bound aggregates in the cytoplasm comprising a dense and stable “core” structure with a dynamic “shell” area, including an intrinsically disordered region (IDR)21. Shell formation involves liquid-liquid phase separation. Stress-induced stress granule formation occurs through protein kinase-induced phosphorylation of eIF2α22,23 and inhibition of eIF4A/E activity24,25, both of which lead to the blockade of translation initiation. Upon translation initiation blockade, RNA-binding proteins containing IDRs, such as G3BP1 and G3BP2, interact with each other and undergo self-aggregation to form the core of stress granules, followed by further fusion, growth, and shell formation through protein-protein interactions26,27. Stress granule formation is a cellular response mechanism by which eukaryotic cells adapt and survive under stress conditions. It helps cells survive by selectively attracting mRNAs to stress granules for translation or repression. Stress granules recruit signaling molecules or factors that govern cell apoptosis, thereby preventing stress-induced cell death.

Viral infections trigger the formation of stress granules, but the impact and mechanisms of different viruses on stress granules vary. The well acknowledged is that stress granules inhibit host protein synthesis by pausing mRNA translation28. This process helps cells conserve resources to cope with viral infection and may also limit the synthesis of viral proteins29. However, stress granules can also inhibit viral RNA for translation. Many virus takes advantage of the characteristics of the stress granules to develop a series of response mechanisms for their own benefit through various mechanisms30. Multiple viruses have evolved to block or subvert SGs components to promote their replication and spread. For example, West Nile Virus and dengue viral proteins interact with TIA-1/TIAR proteins to interfere the formation of stress granules and body assembly31. Poliovirus encodes 2 A protease and 3 C protease, which can cleave key stress granule proteins, inhibiting the formation of stress granules, thereby facilitating viral replication32. EBOV VP35 represses the assembly of canonical and non-canonical SGs via interaction with G3BP1, eIF3 and eEF233. While others may utilize the formation of stress granules to evade host immune surveillance34. Some viral proteins can directly target stress granules, modulating their formation and function35. Hepatitis C and COVID-19 can enhance stress granule assembly via targeting to eIF3 and G3BP1/236,37,38. So G3BP1 seems to be a target for a variety of viruses including our NDRV HN10. The development of G3BP1 drugs to combat the viral use of SG pathogenesis is worthy of attention.

The viral RNA itself is regulated by m6A methylation, and this modification further affects RNA stability and translation. Thus viruses have also developed a set of functions that affect the activity of m6A regulators, such as enterovirus 2 A(pro) cutting the YTHDF m6A reader to suppress the activity of interferon-driven JAK/STAT signaling39, as well as COVID-19 weakening the expression of METTL3 to decrease m6A levels in COVID-19 and host genes, then subsequently enhancing the downstream innate immune signaling pathway and inflammatory gene expression40.

This study’s findings focus on the process of cellular protein translation via σB. The core idea of this study is that σB acts as a bridge to bind to YTHDF1/3 to recognize m6A-labeled RNAs effectively and then recruits G3BP1-mediated stress particles and wraps these RNAs from ribosomes to terminate translation. YTHDF1/3 knockdown disrupted the interaction between stress granules and σB, indicating that YTHDF1 and YTHDF3 have complementary effects on each other. Although there is no direct evidence of m6A modification in NDRV RNA (we did not find positive NDRV RNA in the m6A MeRIP sequencing data), the virus appears not to be able to disrupt stress particles but rather to cleave the host cellular RNA into stress particles, leaving enough viral transcripts in the cytoplasm for efficient translation. We speculate that viral RNA is likely not modified by m6A. It is interesting to think about how viruses exploit cellular regulatory machinery to serve themselves rather than simply destroying them. The interaction protein of σB interacts with numerous other RNA-binding proteins (Supplementary Data 1). The molecular involvement of σB in eukaryotic cells warrants additional study.

Overall, our study reveals a novel role for σB of NDRV HN10 in the translation shutoff of Muscovy ducklings.

Materials and methods

Animal study

As previously described4, one-day-old healthy Muscovy ducklings were collected and randomly separated into groups (n = 10 for each group). Ducklings were administered 500 μl of the HN10 strain at a titer of 106.4 TCID50/ml for NDRV infection, whereas sterile DMEM was used as a negative control (NC). The bursa of Fabricius specimens was harvested at 72 hours postinfection (hpi) for subsequent experiments. All experiments were performed at the Institute of Animal Husbandry and Veterinary Sciences (IAHV), Zhejiang Academy of Agricultural Sciences (ZAAS). All animal samples were treated in accordance with the Regulations of the Administration of Affairs Concerning Experimental Animals approved by the State Council of China. The bird protocol used in this study was approved by the Research Ethics Committee of ZAAS (permit number 2021ZAASLA09). We have complied with all relevant ethical regulations for animal use.

Cell culture

DEF CCL-141 cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco), 100 IU/ml penicillin and 100 μg/ml streptomycin at 37 °C in a 5% CO2 incubator. CCL-141 cells were transfected with the pGL6-TA-luc2-G418 vector (Beyotime, Shanghai, China), and the cell line with stable luciferase expression was screened with G418. The following drug treatment conditions were used: 0.5 mM sodium arsenite for 1 h; 20 nM working concentration of cycloheximide (CHX) (HY-12320, MCE, Shanghai, China) for 12 h; 50 nM bortezomib (BTZ) (HY-10227, MCE) for 2 h; twelve coding fragments (L1, λA; L2, λB; L3, λC; M1, μA; M2, μB; M3, μNS; S1, σC, p10, p18; S2, σA; S3, σB; and S4, σNS) of NDRV HN10 with a fused 3× Flag tag at the 5′ terminus inserted into the plasmid (pSecTag) were transiently transfected into CCL-141 cells using Lipofectamine 3000 (Thermo Fisher Scientific, Waltham, MA, USA); the empty vector (EV) was used as a blank control; and siRNA for YTHDF1 (5′-GACTGATCCTTATCTGTCCAGTTAT-3′) or YTHDF3 (5′-GCCTAGTTATTATGCTCCATCCATT-3′) of Muscovy ducklings was designed and synthesized by Tsingke Biotech. A 1 μM working concentration of siRNA was used for transfection with Lipofectamine RNAiMAX (Thermo Fisher Scientific).

Ribosome footprint profiling (Ribo)

Some steps were adjusted as previously described41. A total of 5 × 107 cells were added to 500 μl of ice-cold lysis buffer (20 mM Tris pH 7.4, 150 mM NaCl, 5 mM MgCl2, 1 mM DTT, 100 μg/ml cycloheximide, 1% Triton X-100 and Turbo DNase I 25 U/ml) and pipetted gently on ice for 30 min. Then, the supernatant was recovered after centrifugation at 20,000 × g for 15 min. Then, 300 μl of the lysate was transferred to a 13 × 51 mm2 polycarbonate ultracentrifuge tube, and 0.9 ml of a 1 M sucrose cushion was added by carefully positioning a pipette tip at the very bottom of the tube and slowly dispensing the sucrose solution. Ribosomes were clarified by centrifugation at 70,000 rpm for 4 h and transferred to a new tube. Ribosome-enriched RNA was isolated by TRIzol. The RNA was subjected to sodium dodecyl sulfate-polyacrylamide gel (SDS‒PAGE) electrophoresis at 200 V for 1 h, and RNA in the 30 nt range was extracted and isolated using a Zymoclean Gel RNA Recovery Kit (Zymo Research, Orange, CA, USA). RNA was converted into cDNA using a HiScript III 1st Strand cDNA Synthesis Kit (Roche, Basel, Kanton Basel, Switzerland) to construct a library for sequencing.

The TPM was generated for the normalization of the data via \({{{\rm{TPM}}}}={10}^{6}* \frac{\frac{{{{\rm{reads}}}}\; {{{\rm{mapped}}}}\; {{{\rm{to}}}}\; {{{\rm{transcript}}}}}{{{{\rm{transcript}}}}\; {{{\rm{length}}}}}}{{{{\rm{Sum}}}}\left(\frac{{{{\rm{reads}}}}\; {{{\rm{mapped}}}}\; {{{\rm{to}}}}\; {{{\rm{transcript}}}}}{{{{\rm{transcript}}}}\; {{{\rm{length}}}}}\right)}\). For translational efficiency analysis, TPM values from Ribo-seq and RNA-seq of one gene were compared as translational efficiency index (TEI), then TEI of each gene were arranged in order to fit the cumulative curve. In particular, in the Ribo-seq of the infected group, viral RNAs were treated separately from eukaryotic mRNA. The whole transcripts of viral RNA were removed from the infected group, and normalized with the negative control.

RNA immunoprecipitation (RIP) and m6A-methylated RIP (MeRIP)

This experiment was conducted following the Abcam protocol. Briefly, tissues containing at least 1 × 107 cells were harvested and resuspended in 2 ml of nuclear separation buffer (1.28 M sucrose, 40 mM Tris pH 7.5, 20 mM MgCl2, and 4% Triton X-100) and kept on ice for at least 30 min with frequent mixing. The pelleted nuclei were centrifuged at 14,000 rpm for 15 min and resuspended in wash buffer (150 mM KCl, 25 mM Tris (pH 7.4), 5 mM EDTA, 0.5 mM DTT, 0.5% NP40, 100 U/ml RNase inhibitor (Solarbio, Beijing, China), and 1× protease inhibitor cocktail (Solarbio)). For MeRIP, total RNA was isolated using TRIzol.

Nuclei pullets or RNA were sheared via sonication at high power for 30 cycles of 5 s on and 30 s off. After that, the supernatant was incubated with 1 μg of antibodies against m6A (Cat. No. ab284130, Abcam) or G3BP1 (Cat. No. 13057-2-AP, Proteintech, Wuhan, Hubei, China) overnight and 40 μl of protein A/G beads (Thermo Fisher Scientific) for 2 h with gentle rotation at 4 °C, while the remaining 10% was harvested as input. The pellet beads were centrifuged at 3000 rpm for 3 min and washed three times. Both the input and pellet beads were purified by TRIzol, and reverse transcription was conducted using a HiScript III 1st Strand cDNA Synthesis Kit (Roche). cDNA libraries were used for next-generation sequencing or qPCR. For sequencing, the concentration and quality of the purified RIP RNAs were measured with a Nanodrop 2000 (Thermo Fisher Scientific) and an Agilent Bioanalyzer 2100 (Agilent, Santa Clara, CA, USA). Four micrograms of RNA from each group were used for library preparation via the NEBNext Ultra Directional RNA Library Prep Kit for Illumina (NEB, Ipswich, MA, USA) following the manufacturer’s instructions and sequenced on an Illumina HiSeq platform.

Firefly luciferase reporter gene assay

A total of 5 × 104 CCL-141 cells stably expressing luciferase were subcultured into BeyoGold 96-well black opaque plates (FCP966, Beyotime). After replacing the 100 μl of fresh medium, another 100 μl of One-LumiTM II Firefly Luciferase Reporter Gene Assay Solution (RG056, Beyotime) was added for 5 min. An ELx808 Absorbance Reader (Lonza, Basel, Kanton Basel, Switzerland) was used to detect the signal fluorescein at a wavelength of 570 nm.

Quantitative polymerase chain reaction (qPCR)

Template cDNA was converted from total RNA or purified RIP RNA using a HiScript III 1st Strand cDNA Synthesis Kit. cDNA (1 μg) was subjected to qPCR with ChamQ SYBR Color qPCR Master Mix (Vazyme, Nanjing, Jiangsu, China). The sequences of primers used were as follows: Flag (F: 5′- GACTACAAAGACCAT -3′, R: 5′- TGTCATCGTCATCCTT -3′); λA (F: 5′- ACCAATGATAATGATGCTTCTTC -3′, R: 5′- TCAACAGCACTGGTAGCTTTAG -3′); λB (F: 5′- GTATCCGTACTTCCTCTCCGTC -3′, R: 5′- GAGTTTCGCTAACCATTGACAC -3′); λC (F: 5′- TGTGTATGACCTGCGTTTTATG -3′, R: 5′- ACGTGAGTTCGACAAATTGGAG -3′); μA (F: 5′- TGCTTCAGCATTTGGAGTTGAC -3′, R: 5′- GAAAGACAGCGTTACCACGTCC -3′); μB (F: 5′- TAGCATGATGTTCGAGGTGAAC -3′, R: 5′- ACCTCAGCCACTTGAGCTATAC-3′); μNS (F: 5′- GCTGTGGAATTGTTGGCATCTG -3′, R: 5′- ATTTTCACCTTCAACACGG -3′); and σC (F: 5′- ACCTGCTGATGCTGTCTTC -3′, R: 5′- GCGAGGACATAAGCAACATGGTC-3′), β-actin (F: 5′- TACGCCAACACGGTGCTG -3′, R: 5′- GATTCATCATACTCCTGCTTGCTG-3′).

Mass spectrum

Immunoprecipitated Flag proteins were separated by C18 reversed-phase chromatography with a TSQ FortisTM system (Thermo Fisher Scientific) and analyzed with a Q Exactive Hybrid Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific). Peptide-to-spectrum matches (PSMs) were made using the Sequest algorithm within Proteome Discoverer (Thermo Fisher Scientific) compared to Muscovy ducklings forward and reverse protein databases (downloaded from UniProt on 07-04-2016; 20,154 sequences). The parent and fragment ion tolerances were 10 ppm and 0.02 Da, respectively. Carbamidomethylation of Cys was included as a static modification, and deamidation of Asn/Gln, oxidation of Met, and Gly/Gly on His was included as dynamic modifications in the search, along with up to two missed tryptic cleavages. The results were imported into Scaffold (Proteome Software; ver 4.8), and a subset database was reanalyzed using X! Tandem. Final peptide identification was determined at a ≤ 1% false discovery rate (FDR).

Coimmunoprecipitation (IP) assay

A total of 1 × 107 cells were disintegrated using Pierce IP lysis buffer [25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% NP-40 and 5% glycerol] (Thermo Fisher Scientific) containing 1% proteasome inhibitor cocktail and 1% PMSF on ice for 30 min. The supernatant was subsequently centrifuged, mixed with 1 μg of antibodies against Flag (Cat No. 66008-4-lg, Proteintech) or rabbit IgG (Cat No. 30000-0-AP, Proteintech) and gently rotated overnight at 4 °C, and 40 μl of flurry IgA beads (Thermo Fisher Scientific) was added for an additional 2-h incubation. Immunoprecipitates were washed with IP buffer [20 mM HEPES pH 7.9, 350 mM NaCl, 0.1% NP-40, 1 mM DTT, 0.2 mM PMSF, 2 mg/ml leupeptin, and 2 mg/ml aprotinin] three times and resuspended in 20 μl of lysis buffer. The protein lysate was subjected to SDS‒PAGE or MASS Spectrum.

Western blot

Tissues or cells were homogenized in Pierce IP lysis buffer and then centrifuged at 4 °C at 13,000 rpm for 10 min. The protein concentration in the supernatant was determined using a BCA protein assay kit (Well-bio, Changzhou, Jiangsu, China). Equal amounts of protein samples were separated by SDS‒PAGE and transferred to polyvinylidene fluoride membranes. The membranes were then blocked with 5% nonfat milk TBS for 90 min and incubated with primary antibodies against YTHDF1 (1:2,000, Cat No. 17479-1-AP, Proteintech), YTHDF3 (1:2,000, Cat No. 25537-1-AP, Proteintech), CAPRIN1 (1: 2,000, Cat No. 15112-1-AP, Proteintech), G3BP1 (1: 2,000), Flag (1: 1,500), β-lactamase (encoding ampicillin resistant protein)(1: 2,000, Cat No. ab12251, Abcam), β-Actin (1: 2,500, Cat No. 81115-1-RR, Proteintech), σB and λA (1: 1,500, both antibodies were lab-made from mouse serum)5 overnight at 4 °C. The membranes were washed in TBST and incubated with HRP-conjugated AffiniPure goat anti-rabbit IgG (1:10,000 dilution, Cat No. SA00001-2, Proteintech) at room temperature for 1 h. The membranes were then treated with an enhanced chemiluminescence detection kit (Pierce, Rockfold, IL, USA), and the intensity of each band was quantified by densitometry.

For in-house generation of antibodies againstσB and λA, NDRV mAb 2-C10 was produced previously in our laboratory using the prime-boost approach. Briefly, six-week-old female BALB/c mice were immunized subcutaneously with purified recombinant σB or λA protein (50 μg per mouse) emulsified with complete Freund’s adjuvant (Sigma-Aldrich, St Louis, MO, USA). Three booster immunization of the same dose were given at two-week intervals mixed with incomplete adjuvant. Three days after the final booster immunization, the spleen cells isolated from immunized mice were fused with SP2/0 myeloma cells by using 50% (wt/vol) polyethylene glycol 1500 (PEG1500; Boehringer Mannheim, IN, USA) at a ratio of 10:1. The hybridoma cells were screened by indirect enzyme-linked immunosorbent assay (ELISA) coated with purified σB protein. The positive hybridoma clones were subcloned 3× by limiting dilution method. One of the NDRV σB-specific MAbs, designated 2-C10, was selected and used in this study, and its subtype was classified using the Mouse MAb Isotyping Kit (Thermo Fisher Scientific). The hybridoma was injected into the peritoneal cavities of BALB/c mice, and subsequent ascites fluid was collected and purified by affinity chromatography using the HiTrap™ Protein G column (GE Healthcare Life Sciences, Pittsburgh, PA, USA).

Immunocytochemistry

Tissues or cells were fixed in 4% paraformaldehyde, washed with phosphate-buffered saline (PBS), permeabilized with 0.1% Triton-X-100 (Sigma-Aldrich), and blocked with 0.5% horse serum in PBS. Immunostaining of the samples was performed using antibodies against G3BP1 (1:250), Flag (1:200), YTHDF1 (1:500), and CAPRIN1 overnight at 4 °C. After washing with PBS 4 times (5 min per wash), the sections were incubated with donkey anti-rabbit secondary antibodies (1: 20000; Jackson ImmunoResearch, West Grove, PA, USA) for 30 min at room temperature, followed by further incubation with DAPI for 15 min and washing with PBS 4 times. After drying in air, the slices were removed, coated with mounting medium, and coated with cover glasses. Positive staining was statistically analyzed using ImageJ.

Immunogold labeling and transmission electron microscopy

The bursa of Fabricius specimens were subjected to transmission electron microscopy (TEM) and perfused with 0.9% saline, followed by 4% paraformaldehyde and 2% glutaraldehyde overnight at room temperature. After the samples were washed three times with PBS and placed in 10% sucrose/4% agarose for 2 h, they were fixed with 1% osmium tetroxide in water at 4 °C for another 1 h, dehydrated with ethanol and embedded in 812 epoxy resin (TAAB laboratories, Aldermaston, Berks, UK). Ultrathin 70-nm-thick sections were cut with an Ultracut E ultramicrotome (Leica Biosystems, Nussloch, Baden-Wurttemberg, Germany) and mounted on Formvar carbon-coated Cu/Pd 100-mesh grids; the sections were stained with uranyl acetate and lead citrate. A JEM1400 Flash (JEOL, Beijing, China) electron microscope was used at 80 kV, with a Oneview 4 K x4K CMOS camera (Gatan, Pleasanton, CA, USA).

Tissues were perfused with 2% paraformaldehyde/0.2% glutaraldehyde in PBS for postembedding immunogolding. Two hundred-micron-thick slices were cryoprotected in 30% glycerol and frozen in liquid ethane in a cryofixation unit (KF80, Leica). Frozen sections were immersed in 0.5% uranyl acetate in methanol at -90 °C in a freeze-substitution instrument (Leica AFS), infiltrated with Lowicryl HM 20 resin at -45 °C, and polymerized with ultraviolet light. Thin sections of the bursa of Fabricius (70 nm) were incubated with 0.15% glycine in PBS for 15 min to quench free aldehyde groups, followed by blocking buffer (1% BSA, 0.2% Tween 20 and 5% FBS in TBS [Tris 30 mM, NaCl 150 mM pH 8.2]) for 30 min at room temperature. Primary antibodies (rabbit anti-total G3BP1 and YTHDF1) were added to the incubation buffer for 1 h, followed by incubation three times in PBS. Protein A-gold (PAg) at 25 nm (for G3BP1) and 10 nm (for YTHDF1) (Shanghai Joey Biotech) was incubated for 45 min at room temperature. Finally, the sections were stained with uranyl acetate and lead citrate and observed under a JEM1400 Flash (JEOL) electron microscope.

Gel filtration

A gel filtration assay was performed as previously described42. In brief, hypotonic buffer supplemented with NP-40 was used to separate the cytoplasm and nuclei of 1 × 108 cells. The cytoplasmic proteins were isolated by high salt extraction buffer [20 mM HEPES (pH 7.9), 420 mM NaCl, 25% glycerol, 1.5 mM MgCl2, 0.2 mM EDTA, 0.5 mM dithiothreitol and protease inhibitors] and then directly applied to a Sepharose 6B column (Sigma‒Aldrich) equilibrated with column running buffer containing 20 mM HEPES (pH 7.9), 200 mM NaCl, 1 mM dithiothreitol, 0.1 mM phenylmethylsulfonyl fluoride and 10% glycerol. One milliliter of each fraction was collected to detect YTHDF1, YTHDF3, G3BP1, CAPRIN1, and Flag by Western blot analysis.

Statistics and reproducibility

The data are shown as the mean ± standard deviation. The student’s t-test was used to analyze the differences between the two groups of three replicates. Some special methods used for omics analysis included one-way ANOVA for identifying significant differences in the abundance of transcripts and ribosome-enriched transcripts in NDRV HN10-infected plants and two-tailed Wilcoxon rank-sum tests for identifying significant differences in the translational efficiency index between infected and control plants. A P value less than 0.05 was considered to indicate statistical significance.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.