Main

A common feature of coronaviruses is their propensity to induce apoptosis in infected target cells. We recently demonstrated that inhibition of apoptosis with a pan-caspase inhibitor significantly attenuated MERS-CoV replication11,12, suggesting a previously unrecognized connection between the apoptosis cascade and coronavirus replication. Previous studies have revealed that caspase-6 can mediate the cleavage of nucleocapsid (N) protein from a number of coronaviruses, including SARS-CoV-1, transmissible gastroenteritis coronavirus (TGEV) and porcine epidemic diarrhea virus13,14,15 (PEDV), but the biological significance of caspase-6-mediated N cleavage is unknown. In this study, we identify caspase-6—a cysteine-aspartic protease that serves as an executor caspase of the apoptosis cascade—as a host factor that facilitates efficient coronavirus replication.

Caspase-6 aids coronavirus replication

We recently demonstrated that MERS-CoV infection triggers substantial apoptosis, and inhibition of apoptosis with the pan-caspase inhibitor z-VAD-fmk significantly limits MERS-CoV replication11,12 (Extended Data Fig. 1a,b). z-VAD-fmk similarly limited the replication of other human pathogenic coronaviruses, including SARS-CoV-2, SARS-CoV-1, HCoV-229E and HCoV-OC43 (Extended Data Fig. 1b,c), suggesting that the dependency on apoptosis or caspase activity for efficient virus replication is conserved for coronaviruses. Caspases are cysteine-aspartic proteases that regulate the host apoptosis cascade16. To investigate which caspase is most responsible for modulating coronavirus replication, we used MERS-CoV as a model virus to evaluate virus replication in the presence of specific inhibitors against individual caspases, and demonstrated that caspase-6 inhibition had the greatest effect on limiting MERS-CoV replication (Fig. 1a,b). The inhibitory effect on MERS-CoV replication by caspase-6 inhibition was conserved across different cell types with the exception of Vero E6 cells (Extended Data Fig. 1d). Inhibition of caspase-6 with its specific inhibitor, z-VEID-fmk, attenuated the replication of all tested coronaviruses, including that of SARS-CoV-2 and SARS-CoV-1, but did not affect the replication of influenza virus (H1N1) or enterovirus (enterovirus A71) (Fig. 1c and Extended Data Fig. 1e). According to quantitative PCR with reverse transcription (RT–qPCR) and median tissue culture infectious dose (TCID50) assays, the half-maximal inhibitory concentration (IC50) of z-VEID-fmk versus coronaviruses ranged from 3.3 μM for SARS-CoV-2 to 21.1 μM for MERS-CoV in the cell lysate samples and 1.2 μM for SARS-CoV-2 to 30.6 μM for HCoV-OC43 in the supernatant samples (Fig. 1d and Extended Data Fig. 1f).

Fig. 1: Caspase-6 inhibition limits coronavirus replication.
figure 1

a,b, Flow cytometry of MERS-CoV-infected BEAS2B cells treated with specific caspase inhibitors (75 μM). Cells were fixed at 24 hpi and labelled with MERS-CoV N immune serum and an antibody that recognizes active caspase-3 (n = 3). c, Virus replication of MERS-CoV, SARS-CoV-2, SARS-CoV-1, HCoV-229E, HCoV-OC43, H1N1 and EV-71 with or without 100 μM z-VEID-fmk. Host cell lines are indicated in parentheses. Virus gene copy number was quantified by RT–qPCR of the N (for MERS-CoV, SARS-CoV-1, HCoV-229E and HCoV-OC43), RdRp (for SARS-CoV-2), M (for H1N1) or VP1 (EV-71) gene (n = 4 for HCoV-OC43 and EV-71, n = 3 for other viruses). RD, rhabdomyosarcoma cell line. d, IC50 of z-VEID-fmk for the replication of MERS-CoV, SARS-CoV-2 (in Calu3 cells), SARS-CoV-1 (in Huh7 cells), HCoV-229E (in Huh7 cells) and HCoV-OC43 (in BSC1 cells) was determined in cell lysate and supernatant samples by RT–qPCR and TCID50 assays, respectively (n = 3). Host cell lines are indicated in parentheses. HFL, primary human embryonic lung fibroblasts. Data are mean ± s.d. from the indicated number of biological repeats. One-way (b) or two-way (c) ANOVA. NS, not significant.

Source data

Next, we used MERS-CoV and SARS-CoV-2 as model coronaviruses and evaluated the effect of caspase-6 inhibition on coronavirus replication in infected human ex vivo lung tissues, human intestinal organoids and animal models. In human lung tissues, caspase-6 inhibition with z-VEID-fmk significantly reduced MERS-CoV N protein and N gene expression (Fig. 2a,b). Similarly, z-VEID-fmk inhibited MERS-CoV replication in human intestinal organoids and inhibited the production of infectious virus particles by approximately 80% (P < 0.0001) at 24 hours post-infection (hpi) (Fig. 2c–f). As caspase-6 is largely conserved among mammals and the z-VEID-fmk-binding pocket is conserved among humans, mice and hamsters, we can evaluate the effect of caspase-6 inhibition with z-VEID-fmk in these animals (Extended Data Fig. 2a,b). To evaluate the effect of caspase-6 inhibition on MERS-CoV replication in vivo, we infected human DPP4 knock-in (hDPP4-KI) mice with mouse-adapted MERS-CoV17 (MERS-CoVMA) and treated them with z-VEID-fmk or DMSO (Fig. 2g). z-VEID-fmk effectively reduced MERS-CoVMA replication in the lungs of infected mice at both 2 and 4 days post-infection (dpi) (Fig. 2h–j), and significantly attenuated the expression of genes encoding pro-inflammatory cytokines and chemokines (Fig. 2k and Extended Data Fig. 2c). Of note, the z-VEID-fmk treatment largely inhibited body weight loss and significantly improved the survival of infected hDPP4-KI mice from 33.3% to 80% (3 out of 9 versus 8 out of 10; P = 0.0388) (Fig. 2l,m).

Fig. 2: Caspase-6 inhibition attenuates MERS-CoV replication in human lung tissues and human intestinal organoids, and improves the survival of hDPP4-KI mice.
figure 2

a,b, Ex vivo human lung tissues were infected with MERS-CoV and treated with z-VEID-fmk. Tissues and supernatants were collected at 24 hpi for immunostaining (a) and RT–qPCR (b; n = 8). cf Human intestinal organoids were infected with MERS-CoV and treated with z-VEID-fmk. c, Organoids were fixed at 24 hpi for immunostaining to detect N protein expression. d, Infected cells were quantified by counting the percentage of cells exhibiting N protein expression in each organoid (n = 5). e, N gene expression was quantified by RT–qPCR (n = 3). f, Infectious titre was determined by plaque assay (n = 6). The limit of detection (LOD) was 50 plaque-forming units (PFU) per ml. g, hDPP4-KI mice were inoculated intranasally with 2.5 × 103 PFU MERS-CoVMA followed by intraperitoneal administration of DMSO or 12.5 mg kg−1 day−1 z-VEID-fmk for 6 days or until sample collection. h, A subset of mice were collected at 2 and 4 dpi. Mouse lungs were immunolabelled to detect MERS-CoV N expression. The experiment was repeated three times independently with similar results. i, Viral gene expression in mouse lungs was quantified by RT–qPCR (n = 6). j, Infectious titre was determined by plaque assay (n = 6). The LOD was 50 PFU ml−1. k, The expression of genes encoding pro-inflammatory cytokines and chemokines was quantified by RT–qPCR (n = 6 for the two infected groups, n = 3 for the mock-infected group). l,m, The body weight (l) and survival (m) of infected mice was monitored for 14 days (n = 10 for z-VEID-fmk group, n = 9 for DMSO group). m, The fraction of survivors is shown. Data are mean ± s.d. from the indicated number of biological repeats. One-way ANOVA (k), two-way ANOVA (e,l), two-sided Student’s t-test (b,d,f,i,j) or log-rank (Mantel–Cox) test (m). Scale bars: 50 μm (a), 20 μm (c) and 100 μm (h).

Source data

We next tested whether z-VEID-fmk could similarly inhibit SARS-CoV-2 replication in vivo using intranasal challenge of golden hamsters with SARS-CoV-2 and treatment with z-VEID-fmk or DMSO (Fig. 3a). Caspase-6 inhibition with z-VEID-fmk significantly reduced SARS-CoV-2 replication in the lungs (Fig. 3b,c), including small airways (Extended Data Fig. 2d) and alveoli (Fig. 3d), and ameliorated the expression of virus-induced pro-inflammatory cytokines and chemokines (Fig. 3e). The attenuated virus replication and expression of pro-inflammatory markers resulted in significant improvements of body weight of infected hamsters (Fig. 3f). Furthermore, we collected lung tissues from hamsters at 4 dpi to evaluate histological changes. Lungs from infected hamsters in the mock treatment group showed severe bronchiolar epithelial cell death and desquamation, extensive mononuclear cell infiltration of alveolar space, protein-rich fluid exudation, alveolar haemorrhage and severe destruction of alveolar structure. Pulmonary blood vessel wall inflammation and endothelium infiltration were frequently observed (Fig. 3g, middle). These histopathological changes were consistent with our previous report on SARS-CoV-2-infected hamsters18. By contrast, all categories of tissue damage, including bronchiolitis, alveolitis and vasculitis, were significantly ameliorated in the lungs of z-VEID-fmk-treated hamsters. Although the z-VEID-fmk treatment substantially inhibited immune cell infiltration of alveolar space, some degree of haemorrhage was still visible in the treatment group (Fig. 3g, bottom). Semi-quantitative histopathological examination of bronchioles, alveoli and blood vessels using our previously described methods19 showed that z-VEID-fmk treatment significantly ameliorated lung damage in infected hamsters (Fig. 3h). Together, our results demonstrate that caspase-6 inhibition attenuates coronavirus replication in cell culture, human lung tissue, organoid and animal settings.

Fig. 3: Caspase-6 inhibition ameliorates lung pathology and body weight change of golden Syrian hamsters infected with SARS-CoV-2.
figure 3

a, Golden Syrian hamsters were inoculated with intranasally 3 × 103 PFU SARS-CoV-2 followed by intraperitoneal administration of DMSO or 12.5 mg kg−1 day−1 z-VEID-fmk for 4 days. b,c, Hamsters were euthanized at 4 dpi, and viral gene copies were quantified by RT–qPCR (b) and infectious titre in the lungs was quantified by TCID50 assay (c) (n = 6). d, Viral N protein expression in the alveoli of infected hamster lungs with or without z-VEID-fmk treatment was detected by immunofluorescence using rabbit immune serum raised against SARS-CoV-2 N. The experiment was repeated two times independently with similar results. e, The expression of pro-inflammatory cytokines and chemokines was quantified by RT–qPCR (n = 6). f, The change in body weight of hamsters infected with SARS-CoV-2 and treated with mock or z-VEID-fmk was documented up to 4 dpi. g, Representative images of haematoxylin and eosin (H&E)-stained hamster lungs. Top, lung sections from mock-infected hamsters showed normal histology. Middle, in hamsters infected with SARS-CoV-2, lung tissues showed diffuse inflammatory infiltration and exudation with fewer air-exchange structures. Bottom, hamster lung pathology was markedly improved with z-VEID-fmk treatment. h, Scores for the lung histopathological changes of SARS-CoV-2-infected hamsters with or without z-VEID-fmk treatment. Three categories of characteristic histopathological changes, including bronchiolitis, alveolitis and vasculitis, were examined and scored. n = 6 hamsters per group; 2–3 lung lobes were examined from each hamster. Data are mean ± s.d. from the indicated number of biological repeats. One-way ANOVA (e), two-way ANOVA (f) or two-sided Student’s t-test (b,c,h). Scale bars: 100 μm (d) and 200 μm (g).

Source data

Caspase-6 cleaves nucleocapsid protein

Next, we sought to understand the role of caspase-6 in coronavirus replication. Using MERS-CoV as a model, we first assessed the effect of z-VEID-fmk in a time-of-addition assay. The z-VEID-fmk added during MERS-CoV inoculation did not reduce virus replication (Extended Data Fig. 3a). Consistent with this finding, MERS-CoV entry in caspase-6-stable-knockdown A549 and BEAS2B cells was not compromised (Extended Data Fig. 3b–d). Next, we evaluated MERS-CoV N gene expression in caspase-6 stable-knockdown A549 and BEAS2B cells collected at 24 hpi and found that MERS-CoV replication was significantly reduced in the presence of caspase-6 knockdown (Extended Data Fig. 3e). We then treated the stable scrambled knockdown or caspase-6 knockdown A549 and BEAS2B cells with z-VEID-fmk and showed that the viral inhibitory effect of z-VEID-fmk was largely diminished among the caspase-6 stable-knockdown A549 and BEAS2B cells (Extended Data Fig. 3f). We further investigated the role of caspase-6 in MERS-CoV replication in human primary monocyte-derived macrophages (MDMs). Transient depletion of caspase-6 with siRNA markedly reduced MERS-CoV replication in MDMs (Extended Data Fig. 3g,h). For a more complete depletion of caspase-6, we performed caspase-6 CRISPR knockout (KO) in Huh7 cells and demonstrated that MERS-CoV replication was significantly compromised in caspase-6-KO Huh7 cells compared with caspase-6 intact cell controls (Fig. 4a and Extended Data Fig. 3i). As well as gene-depletion studies, we examined MERS-CoV replication in caspase-6-overexpressing cells and demonstrated that overexpression of caspase-6—but not caspase-3—efficiently promoted MERS-CoV replication (Extended Data Fig. 3j,k). Since the z-VEID-fmk may have cross-reactivity against other caspases20,21, we further validated the in vivo importance of caspase-6 on coronavirus replication by performing caspase-6 knockout in hDPP4-KI mice (Extended Data Fig. 4a,b) followed by MERS-CoVMA challenge. Our results demonstrated that MERS-CoVMA replication and MERS-CoVMA-induced lung damage were significantly reduced in lungs of hDPP4-KI caspase-6-KO mice compared with hDPP4-KI/caspase-6 WT mice (Fig. 4b–e and Extended Data Fig. 4c–e). Collectively, these results indicate that caspase-6 is required for efficient MERS-CoV replication in vitro and in vivo.

Fig. 4: Caspase-6-mediated cleavage of coronavirus N interferes with IFN signalling.
figure 4

a, Huh7 cells treated with caspase-6 or scramble single guide RNA (sgRNA) were infected with MERS-CoV at a multiplicity of infection (MOI) of 1. Cell lysate and supernatant samples were collected at 2, 24 and 48 hpi to quantify virus replication by RT–qPCR of the MERS-CoV N gene or TCID50 assay (n = 4). be, hDPP4-KI/caspase-6-KO and hDPP4-KI/caspase-6 wild-type (WT) mice were inoculated intranasally with 2.5 × 103 PFU MERS-CoVMA. Mouse lung samples were collected at 2 and 4 dpi. c,d, MERS-CoV N gene expression in mouse lungs was quantified by RT–qPCR (c; n = 7) and the infectious titre was determined by plaque assay (d; n = 7). The LOD was 40 PFU ml−1 and is indicated by a dashed line in d. e, MERS-CoV N antigen expression at 4 dpi was determined by immunofluorescence staining. f, N cleavage in MERS-CoV-infected caspase-6 (CASP6)-expressing cells with or without z-VEID-fmk. Cl-CASP6, cleaved caspase-6. g, Huh7 cells were pretreated with DMSO, 5 μM filgotinib, 5 μM ruxolitinib or 5 μM IFNα-IFNAR-IN-1 hydrochloride for 1 h and infected with MERS-CoV at a MOI of 1. After infection, the cells were incubated in media supplemented with DMSO, 5 μM filgotinib, 5 μM ruxolitinib or 5 μM IFNα-IFNAR-IN-1 hydrochloride in the presence of z-VEID-fmk as indicated. Cell lysates were collected at 24 hpi. MERS-CoV N gene copy number was quantified by RT–qPCR (n = 3). h,i, 293T cells were transfected with an IFNβ-luciferase (Luc) reporter plasmid, expression constructs for MERS-CoV N or E proteins and caspase-6 or caspase-3, with or without poly(I:C). The cells were incubated for 24 h before analysis by dual-luciferase reporter assays (n = 4). Experiments in e,f were repeated three times independently with similar results. Data are mean ± s.d. from the indicated number of biological repeats. One-way ANOVA with Tukey’s post hoc test (h,i), two-way ANOVA (a,g) or two-sided Student’s t-test (c,d). Scale bar, 100 μm.

Source data

N fragments modulate IFN response

Together with caspase-3 and caspase-7, caspase-6 is one of the three executor caspases that execute apoptosis by proteolytic cleavage of host substrates22. Caspase-6 is activated by cleavage when apoptosis is induced, but can also undergo autoactivation23 (Extended Data Fig. 5a,b). Previous studies have revealed that caspase-6 can cleave the N protein of SARS-CoV-1, TGEV and PEDV13,14,15. We co-expressed caspase-6 with various MERS-CoV components and evaluated viral protein cleavage by caspase-6. We used staurosporine (STS) to trigger apoptosis to mimic the apoptotic environment in MERS-CoV-infected cells. Our results demonstrated that caspase-6 mediated the cleavage of MERS-CoV N but not other viral components (Extended Data Fig. 5c,d). Cleavage of N was completely abolished in the presence of the specific caspase-6 inhibitor z-VEID-fmk (Extended Data Fig. 5e). Of note, N cleavage was readily detected in cells infected by MERS-CoV (Fig. 4f and Extended Data Fig. 5f), and was similarly inhibited by caspase-6 inhibition in infected cells (Fig. 4f). In addition, N was cleaved by capsase-6, but not by the key executor caspase, caspase-3 (Extended Data Fig. 5g). Together, these results demonstrate that caspase-6 specifically cleaves MERS-CoV N. Next, we investigated how caspase-6-mediated N cleavage modulates coronavirus replication. We previously demonstrated that caspase-6 inhibition attenuated MERS-CoV replication in all evaluated cell types with the exception of Vero E6 cells (Extended Data Fig. 1d), which are deficient in interferon (IFN) signalling owing to a homozygous deletion in the type-I IFN gene cluster24,25. We then treated MERS-CoV-infected Huh7 cells with z-VEID-fmk in the presence of DMSO, filgotinib (JAK inhibitor), ruxolitinib (JAK inhibitor) or IFNα-IFNAR-IN-1 hydrochloride (IFNAR inhibitor). Our results demonstrated that the antiviral effect of z-VEID-fmk diminished when the IFN pathway was inactivated by JAK or IFNAR inhibition (Fig. 4g). These results hinted that caspase-6-mediated N cleavage might modulate coronavirus replication by regulating IFN signalling. To test this hypothesis, we evaluated the role of caspase-6 and MERS-CoV N in regulating IFN response with IFNβ reporter assays. Notably, we demonstrated that MERS-CoV N co-expressed with caspase-6 suppressed IFNβ reporter activation in a dose-dependent manner (Fig. 4h and Extended Data Fig. 6a). Co-expression of caspase-3 and MERS-CoV N or caspase-6 and MERS-CoV envelope (E) protein did not significantly impact IFNβ reporter activation (Fig. 4i). In addition to IFNβ reporter activity, co-expression of caspase-6 and MERS-CoV N similarly reduced the expression of IFNβ and representative IFN-stimulated genes, including IFIT1, IFIT2, IFIT3, IFITM3, TRIM22 and OAS1 (Extended Data Fig. 6b). Recent studies have identified MERS-CoV ORF4a, ORF4b and membrane (M) protein as potent IFN antagonists26,27,28. Our data showed that caspase-6 did not modulate the IFN antagonism of these known IFN antagonists (Extended Data Fig. 6c). Moreover, caspase-6-cleaved N modulated IFN response at least as potently as these known IFN antagonists (Extended Data Fig. 6d). Notably, we further demonstrated that caspase-6 similarly mediated N cleavage of other human pathogenic coronaviruses, including that of SARS-CoV-2 and SARS-CoV-1 (Extended Data Fig. 7a,b), which is in agreement with our findings that caspase-6 inhibition attenuated the replication of these coronaviruses (Fig. 1c,d). We next expressed the N genes of these coronaviruses together with caspase-6, which revealed that the co-expression of coronavirus N with caspase-6 antagonized IFNβ reporter activation and reduced the expression of representative IFN-stimulated genes, including IFIT3 and OAS1 (Extended Data Fig. 7c,d). Collectively, these results suggest that caspase-6-mediated N cleavage modulates coronavirus replication by regulating IFN signalling.

To further explain how caspase-6-mediated N cleavage regulates IFN response, we analysed the potential caspase-6 cleavage sites on MERS-CoV N on the basis of known caspase-6 substrate specificity29 and the size of the cleavage fragments, and generated the corresponding N mutants that potentially interfered with caspase-6 cleavage (Fig. 5a). Western blot-based cleavage assays demonstrated that caspase-6-mediated cleavage was abolished for the T239KKA242 mutant, suggesting that caspase-6 cleaves MERS-CoV N at the T239KKD242 motif (Fig. 5b). The T239KKD242 motif is located within the intrinsically disordered region (IDR) of MERS-CoV N that bridges the N-terminal and C-terminal domain of N, which are structurally conserved among coronaviruses30. Similar putative caspase-6 cleavage motifs are also present in the IDR of N of other human pathogenic coronaviruses (Supplementary Table 1). In IFNβ reporter assays, caspase-6 and N-mediated IFN antagonism was attenuated when T239KKA242 was expressed in place of the wild-type N (Fig. 5c). These findings suggested that caspase-6-mediated MERS-CoV N cleavage is essential for its IFN antagonism. Next, we generated N fragments containing residues 1–241 and 242–413, mimicking the N cleavage products (Fig. 5a). We showed that these two fragments were no longer cleaved by caspase-6 (Extended Data Fig. 8a). Consistent with this result, in IFNβ reporter assays and IFNβ ELISA, the two N cleavage products individually limited IFNβ reporter activity but were no longer modulated by caspase-6 (Extended Data Fig. 8b–d). Similarly, we also identified the caspase-6 cleavage site on SARS-CoV-2 N and showed that the fragments of SARS-CoV-2 N also served as potent IFN antagonists (Extended Data Fig. 9a–e). Next, to dissect how the MERS-CoV N fragments modulate IFN signalling, we evaluated the point of action of the MERS-CoV N fragments with IFNβ luciferase reporter assays using RIG-IN (a constitutively active form of RIG-I), MAVS or TBK1 as activators. We demonstrated that MERS-CoV N fragments efficiently reduced the IFNβ luciferase reporter activity when activated by RIG-IN, MAVS or TBK1, suggesting that the N fragments act downstream of RIG-I, MAVS and TBK1 (Extended Data Fig. 8e). Next, we evaluated the capacity of N fragments to interact with different components of the IFN signalling pathway using co-immunoprecipitation assays. We demonstrated that N1–241 and N242–413 interacted with IRF3 but not with other components of the IFN signalling pathway (Extended Data Fig. 8f,g). In Huh7 and 293T cells treated with the TLR3 agonist poly(I:C), IRF3 translocated to the cell nuclei, regardless of MERS-CoV N expression. N1–241 and N242–413 fragments co-localized with IRF3 and abolished its nuclear translocation (Extended Data Fig. 10a–c).

Fig. 5: Recombinant MERS-CoV with a single amino change at the caspase-6 cleavage motif on N protein is attenuated in vitro and in vivo.
figure 5

a, Schematic of N mutant proteins. b, Caspase-6-mediated cleavage of N mutants was evaluated by western blot. The experiment was repeated three times independently with similar results. c, 293T cells were transfected with an IFNβ-Luc reporter plasmid, expression constructs of caspase-6 and MERS-CoV N or N mutants, with or without poly(I:C). The cells were incubated for 24 h before analysis by dual-luciferase reporter assay (n = 3). d, A549, Huh7 and Vero E6 cells were infected with rMERS-CoV/TKKA or rMERS-CoV/TKKD at a MOI of 1 (A549 and Huh7) or 0.5 (Vero E6). Infectious titres were quantified by TCID50 assay (n = 3). eg, hDPP4-KI mice were inoculated intranasally with 3.5 × 104 PFU rMERS-CoV/TKKA or rMERS-CoV/TKKD. Mouse lung samples were collected at 2 and 4 dpi. MERS-CoV N gene expression in mouse lungs was quantified by RT–qPCR (e; n = 6) and infectious titre was determined by plaque assay (f; n = 6). The LOD was 40 PFU ml−1 and is indicated by a dashed line in f. g, MERS-CoV N antigen expression was determined with immunofluorescence staining. The experiment was repeated two times independently with similar results. Data are mean ± s.d. from the indicated number of biological repeats. One-way ANOVA with Tukey’s post hoc test (c), two-way ANOVA (d) or two-sided Student’s t-test (e,f). Scale bar: 100 μm.

Source data

To further investigate the in vitro and in vivo roles of caspase-6-mediated N cleavage, we engineered MERS-CoV with a point mutation at the caspase-6 cleavage site on the N protein that abolished caspase-6 cleavage (rMERS-CoV/TKKA) (Extended Data Fig. 11a,b). We then infected A549, Huh7 and Vero E6 cells with rMERS-CoV/TKKA or the wild-type recombinant MERS-CoV (rMERS-CoV/TKKD). We demonstrated that rMERS-CoV/TKKA replication was significantly reduced compared with rMERS-CoV/TKKD in A549 and Huh7 cells, but not in IFN-deficient Vero E6 cells (Fig. 5d and Extended Data Fig. 11c). Next, we infected Huh7 and A549 cells with rMERS-CoV/TKKA or rMERS-CoV/TKKD, followed by z-VEID-fmk treatment. Our results demonstrated that although z-VEID-fmk remained effective against the replication of rMERS-CoV/TKKD, the replication of rMERS-CoV/TKKA was not limited by z-VEID-fmk (Extended Data Fig. 11d). Finally, we infected hDPP4-KI mice with rMERS-CoV/TKKA or rMERS-CoV/TKKD to evaluate virus replication in vivo. This showed that in hDPP4-KI mice, the single amino acid substitution at the caspase-6 cleavage site on N significantly reduced rMERS-CoV/TKKA replication in the lungs as well as lung damage compared with rMERS-CoV/TKKD (Fig. 5e–g and Extended Data Fig. 11e).

Discussion

Our study showed that human pathogenic coronaviruses exploit the host apoptotic pathway through caspase-6-mediated N cleavage to dampen the host IFN response for efficient replication. Inhibition of caspase-6 markedly attenuates coronavirus replication and ameliorates coronavirus-induced lung pathology in vivo, suggesting that caspase-6 inhibition should be further explored as an option for the treatment of highly pathogenic coronaviruses (Extended Data Fig. 12).

The replication of coronaviruses, including MERS-CoV and SARS-CoV-2, is known to depend on the host serine protease transmembrane protease serine 2 (TMPRSS2), which cleaves and activates the spike (S) protein of coronaviruses for efficient cell entry and replication31,32. By contrast, the role of host cysteine-aspartic protease in coronavirus replication has not been explored. Previous studies have suggested that influenza viruses require caspase-3 for efficient translocation of viral ribonucleoprotein (RNP) complexes across the nuclear membrane, which is essential for efficient virus replication33. Here we show that caspase-6—but not caspase-3—facilitates coronavirus replication. Caspase-6 is known for its role as an executor caspase and its catalytic role in neurodegeneration in Huntington’s and Alzheimer’s disease34. Of note, caspase-6 has been shown to facilitate inflammasome activation, cell death and host defence during influenza virus infection35. In contrast to coronaviruses, influenza virus replicated more efficiently in the lungs of caspase-6-KO mice compared with wild-type mice35. Here we show that caspase-6 knockout or inhibition attenuates virus replication and disease severity in mice and hamsters infected with highly pathogenic coronavirus. These findings suggest that the inhibition of cysteine-aspartic protease caspase-6 should be explored as a therapeutic option against highly pathogenic coronaviruses, including SARS-CoV-2.

The IFN response is a first-line host defence against coronavirus infection. Genomes of coronaviruses encode multiple viral components26,27,28,36,37,38,39,40, including N protein41,42,43,44 that can serve as IFN antagonists by targeting different steps of the IFN signalling pathway. Extending these findings, our study further reveals host proteases as factors that can modulate viral antagonism in conjunction with viral components. This suggests that our current understanding of viral components that antagonize IFN signalling may be incomplete, since viral components may be processed by host proteases, thus modulating their capacity to interact with host IFN signalling pathways.

Coronavirus N is an abundantly expressed multi-functional structural protein30 that is required for forming complexes with viral RNA and assembly of virus particles. A complex with uncleaved N is required to carry out these functions. After virus uncoating, increasing intracellular viral RNA is detected by host innate immune sensors, which activates interferon responses and apoptotic pathways. Concomitantly, viral replication increases the amount of N that can serve another important purpose during which the complex structure is cleaved by caspase-6 to antagonize the IFN signalling pathway so that viral replication can continue. This mechanism exemplifies how coronaviruses evolve to maximize the use of their most abundantly expressed protein in a time-dependent manner.

Overall, our study reveals how coronaviruses exploit a component of the apoptosis cascade to facilitate virus replication in order to maximize virus production. This is an elegant example of virus–host interaction that exemplifies the longstanding arms race between humans and coronaviruses.

Methods

Cell lines

A549, BSC1, Caco2, Huh7, Vero E6 and 293T cells were maintained in Dulbecco’s Modified Eagle medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum (FBS), 100 U ml−1 penicillin, and 100 μg ml−1 streptomycin. BEAS2B and Calu3 cells were maintained in DMEM/F12 supplemented with 10% heat-inactivated FBS, 100 U ml−1 penicillin and 100 μg ml−1 streptomycin. HFL were maintained in minimum essential medium (MEM) supplemented with 10% heat-inactivated FBS, 100 U ml−1 penicillin and 100 μg ml−1 streptomycin. Human primary monocytes were obtained from human peripheral blood mononuclear cells (PBMCs) taken from healthy donors, collected from Hong Kong Red Cross Blood Transfusion Service according to a protocol approved by the Institutional Review Board of the University of Hong Kong. MDMs were differentiated from monocytes in RPMI-1640 medium supplemented with 10% heat-inactivated FBS, 100 U ml−1 penicillin, 100 μg ml−1 streptomycin, 2 mM glutamine, 1% sodium pyruvate, 1% non-essential amino acids, and 10 ng ml−1 recombinant human granulocyte macrophage colony-stimulating factor (GM-CSF) (R&D Systems) as previously described45.

Viruses

The MERS-CoV (EMC/2012) strain of MERS-CoV was provided by R. Fouchier. MERS-CoVMA was a gift from P. McCray. SARS-CoV-2 wild-type HKU-001a46, Omicron BA.1 (GenBank: OM212472) and Omicron BA.2 (GISAID: EPI_ISL_9845731) were isolated from a laboratory-confirmed patient with COVID-19 in Hong Kong. SARS-CoV-1 GZ50, HCoV-229E, HCoV-OC43, enterovirus A71 and influenza A virus strain A/Hong Kong/415742/2009(H1N1)pdm09 were archived clinical isolates at Department of Microbiology, Hong Kong University. All infectious experiments involving MERS-CoV, SARS-CoV-2 and SARS-CoV-1 followed the approved standard operating procedures of the Biosafety Level 3 facility at the Department of Microbiology, Hong Kong University47.

Chemical modulators

The pan-caspase inhibitor, z-VAD-fmk, was obtained from Invivogen (tlrl-vad). The caspase-1-to-caspase-10 inhibitor sampler kit was purchased from R&D Systems. The caspase-6 inhibitors, z-VEID-fmk, used for in vitro and in vivo experiments, were obtained from R&D Systems (FMK006) and APExBIO (A1923). The apoptosis enhancer, STS (S5921), was obtained from Sigma. Filgotinib (JAK1 inhibitor) (HY-18300), ruxolitinib (JAK1/2 inhibitor) (HY-50856), and IFNα-IFNAR-IN-1 hydrochloride (HY-12836A) were obtained from MedChemExpress. Poly(I:C) was from R&D Systems (4287/10).

Antibodies for western blots

MERS-CoV N was detected by an in-house guinea pig immune serum (1:100,000). MERS-CoV Spike was detected by an in-house mouse immune serum (1:10,000). SARS-CoV-2 N was detected by an in-house rabbit immune serum (1:10,000). SARS-CoV-1 N was detected by an in-house rabbit immune serum (1:10,000). Primary antibodies, including rabbit anti-caspase-3 (ab32351) (1:5,000), rabbit anti-caspase-6 (ab185645) (1:5,000), rabbit anti-HA (ab9110) (1:5,000), mouse anti-Flag (ab49763) (1:5,000), mouse anti-His (ab18184) (1:3,000) and mouse anti-β-actin (ab8226) (1:10,000), were from Abcam. Secondary antibodies, including goat anti-mouse horseradish peroxidase (HRP) (62-6520) (1:5,000), goat anti-rabbit HRP (65-6120) (1:5,000) and goat anti-guinea pig HRP (A18769) (1:10,000), were from Thermo Fisher Scientific.

Antibodies for immunofluorescence staining

MERS-CoV N was detected by an in-house guinea pig immune serum (1:2,000). SARS-CoV-2 N was detected by an in-house rabbit immune serum (1:1,000). IRF3 was detected by rabbit anti-HA (ab9110) (1:500). Secondary antibodies, including Alexa Fluor 488 goat anti-guinea pig (A-11073) (1:500), Alexa Fluor 488 goat anti-rabbit (A-11034) (1:500) and Alexa Fluor 568 goat anti-rabbit (A-11036) (1:500), were from Thermo Fisher Scientific.

Ex vivo human lung tissues

Human lung tissues for ex vivo studies were retrieved from patients undergoing surgical operations at the Queen Mary Hospital, Hong Kong. All donors gave written consent as approved by the Institutional Review Board of the University of Hong Kong/Hospital Authority Hong Kong West Cluster. Normal nonmalignant lung tissue fragments in excess for clinical diagnosis were used. The freshly obtained lung tissues were processed into small rectangular pieces and were rinsed with the primary tissue culture medium, which contained the advanced DMEM/F12 medium supplemented with 2 mM HEPES (Gibco), 1× GlutaMAX (Gibco), 100 U ml−1 penicillin, 100 μg ml−1 streptomycin, 20 μg ml−1 vancomysin, 20 μg ml−1 ciprofloxacin, 50 μg ml−1 amikacin, and 50 μg ml−1 nystatin. The specimens were infected with MERS-CoV at a titre of 1 × 108 PFU ml−1. After 2 h, the inoculum was removed and the specimens were washed thoroughly with the primary tissue culture medium. The infected tissues were then incubated with primary tissue culture medium supplemented with 100 μM caspase-6 inhibitor z-VEID-fmk or DMSO. Tissues were collected at 24 hpi with either 10% neutral-buffered formalin for immunofluorescence staining or with RL buffer (TaKaRa) for RT–qPCR analysis.

Human intestinal organoids

Human intestinal organoids were established using biopsied human intestinal tissues from patients who underwent surgical operations at the Queen Mary Hospital, Hong Kong. All donors had written consent as approved by the Institutional Review Board of the University of Hong Kong/Hospital Authority Hong Kong West Cluster. Human intestinal organoids were maintained in expansion medium and induced differentiation by incubating with differentiation medium for 5 days. Differentiated intestinal organoids were sheared mechanically and inoculated with MERS-CoV at 1 MOI for 2 h. After the inoculum was removed, the intestinal organoids were rinsed with PBS and embedded in Matrigel and maintained in differentiation medium containing 100 μM z-VEID-fmk or DMSO. At the indicated time points after inoculation, intestinal organoids were collected for the quantification of intracellular viral load and immunofluorescence staining, whereas the cell-free Matrigel and culture medium were combined for viral titration of extracellular virions using standard plaque assays.

Human DPP4 mouse model

The hDPP4-K mice were provided by P. McCray17. The use of animals complied with all relevant ethical regulations and was approved by the Committee on the Use of Live Animals in Teaching and Research of The University of Hong Kong. On the day of infection, hDPP4-KI mice were inoculated intranasally with 2.5 × 103 PFU mouse-adapted MERS-CoV (MERS-CoVMA) pre-diluted in 20 μl DMEM, followed by intraperitoneal injection with 12.5 mg kg−1 day−1 z-VEID-fmk or DMSO diluted in 200 μl 0.3% methylcellulose/0.1% tween-80/PBS once per day for 6 days or until sample collection. The health status and body weight of the mice were monitored for 14 days on a daily basis or until the mouse was euthanized because it reached the humane endpoint of the experiment. Mice were euthanized at the designated time points and lung tissues from mice of both treatment and control groups were collected for immunofluorescence staining, RT–qPCR and plaque assay analysis.

Golden Syrian hamster model

Infection of golden Syrian hamsters was performed as described previously18. Golden Syrian hamsters aged 6–8 weeks old were obtained from the Chinese University of Hong Kong Laboratory Animal Service Centre through the HKU Centre for Comparative Medicine Research (CCMR). The use of animals has complied with all relevant ethical regulations and was approved by the Committee on the Use of Live Animals in Teaching and Research of The University of Hong Kong. On the day of infection, each hamster was inoculated intranasally with 3 × 103 PFU SARS-CoV-2 pre-diluted in 50 μl DMEM under intraperitoneal ketamine (100 mg kg−1) and xylazine (10mg kg−1) anaesthesia. Infected hamsters were treated with 12.5 mg kg−1 day−1 z-VEID-fmk or DMSO diluted in 600 μl 0.3% methylcellulose/0.1% tween-80/PBS once per day for 4 days. The health status and body weight of the hamsters were monitored on a daily basis or until the animal was euthanized because it reached the humane endpoint of the experiment. Hamsters were euthanized at 4 dpi and lung tissues were collected for immunofluorescence staining, histopathology examination, RT–qPCR and TCID50 assay analysis.

Generation and infection of caspase-6 knockout mice

Generation of caspase-6-KO mice on the hDPP4-KI mouse background performed with help from the Centre for Comparative Medicine Research at the University of Hong Kong. In brief, three sgRNA purchased from Synthego (5′-CCCTCATCTTCAATCACGAGAGG-3′, 5′-CTCCTGCTCAAAATTCACGAGGG-3′, and 5′-TGGCGTCGTATGCGTAAACGTGG-3′) were designed to target caspase-6 gene exon 3, exon 4, and exon 5, respectively (Extended Data Fig. 4a). The Cas9 protein was purchased from Invitrogen (TrueCut Cas9 Protein v2). First, the sgRNAs (0.5 μg μl−1) and Cas9 protein (0.4 μg μl−1) were diluted in Opti-MEM (Gibco). The ribonucleoprotein (RNP) was formed by vortexing the reagent mixture, followed by a 20 min incubation at room temperature. Second, in vitro fertilization was performed to generate homozygous hDPP4 embryos required for generating double mutant by electroporation. Five-week-old homozygous hDPP4 females were super-ovulated with intraperitoneal injection of 7.5 IU pregnant mare serum gonadotropin (PMSG), followed by intraperitoneal injection of 7.5 IU human chorionic gonadotropin (hCG) 48 h later. Twelve hours after hCG injection, three-month-old homozygous hDPP4 stud males were euthanized. Sperm was released from cauda epididymis into one well of a 4-well plate. The sperm was allowed to capacitate in modified human tubal fluid medium (mHTF) for 45 min in an incubator (5% CO2, 37 °C). Oocytes cumulus were collected from the oviducts of the super-ovulated females and were added into the sperm well. Four hours after the in vitro fertilization, embryos were washed twice with mHTF medium and twice with potassium-supplemented simplex optimized medium (KSOM). Embryos were cultured in KSOM in an incubator until the electroporation. Third, the RNP was delivered into pronuclear stage embryos using a square wave electroporator (CUY21 EDIT II, BEX). Five microlitres of the RNP were pipetted into the chamber of the platinum electrode with a 1 mm gap (LF501PT1-5, BEX). The parameters were set as followed, five poring pulses: 200 V, 2 ms length, 50 ms interval, 4 times, 10% voltage decay, + polarity, followed by five transfer pulses: 20 V, 50 ms length, 50 ms interval, 40% voltage decay, alternating + and − polarity. The impedance was adjusted below 0.17 ohm by adding small volume of Opti-mem into the gap. The electroporated embryos were immediately transferred back to KSOM and were incubated for 30 min. The embryos were washed twice with fresh KSOM and were incubated overnight. 2-cell embryos were transferred into the oviduct of 0.5 days post-conception pseudo-pregnant ICR (CD-1) females in the next morning. Twenty days after the embryo transfer, pups carrying the knockout allele were born. The pups were weaned after 3 weeks of age. Ear punch samples were collected for caspase-6 genotyping. On the day of infection, hDPP4-KI caspase-6 KO mice and hDPP4-KI caspase-6 wild-type littermate controls were concurrently infected by intranasal inoculation with 2.5 × 103 PFU MERS-CoVMA pre-diluted in 20 μl DMEM. Mice were euthanized at 2 dpi and 4 dpi and lung tissues from mice of both groups were collected for RT–qPCR, plaque assay analysis, H&E staining and immunofluorescence staining.

Generation and infection of MERS-CoV recombinant virus

The cDNA from the MERS-CoV strain EMC/2012 (GenBank accession number JX869059), assembled into the pBAC (pBAC-SA-FL), was used as the background to generate the TKKA mutant (rMERS-CoV/TKKA) with a D to A change at the TKKD motif of the MERS-CoV N gene48. The mutation was introduced into the pBAC by red recombineering49. The mutation was confirmed by Sanger sequencing. The recombinant clones with mutant site were transformed into DH10B electrocompetent cells, followed by plasmids extraction to acquire ultrapure and high quality of full-length cDNA clone. Infectious virus is recovered by transfection of BHK21 cells with 5 μg of the full-length cDNA clone using Lipofectamine 3000 as transfection reagent. After 6 h post-transfection, the transfected BHK21 cells were re-seeded and co-cultured with Huh7 cells. After 72 h, the supernatant was used to inoculate Huh7 cells for viral passage. The recombinant virus was sent to next generation sequencing to confirm the desired mutation and the absence of undesired mutations in the viral genome. For in vitro infection, Huh7, A549, and Vero E6 cells were infected with rMERS-CoV/TKKA and rMERS-CoV/TKKD. Cell lysate and supernatant samples were collected from the infected cells at 2, 24 and 48 hpi for RT–qPCR and TCID50 assays, respectively. Additionally, rMERS-CoV/TKKA- and rMERS-CoV/TKKD-infected Huh7 cells were collected at 1, 24, and 48 hpi for western blot analysis of N protein expression. For in vivo infection, hDPP4-KI mice were intranasally inoculated with 3.5 × 104 PFU rMERS-CoV/TKKA or rMERS-CoV/TKKD pre-diluted in 20 μl DMEM. Mice were euthanized at 2 dpi and 4 dpi and lung tissues from mice of both groups were collected for RT–qPCR, plaque assay analysis, H&E staining and immunofluorescence staining.

Caspase-6 CRISPR–Cas9 knockout in vitro

Caspase-6 CRISPR–Cas9 KO plasmids were purchased from Santa Cruz Biotechnology. Knockout of caspase-6 in Huh7 cells was operated by co-transfection with HDR plasmid according to the manufacturer’s protocol (https://datasheets.scbt.com/protocols/CRISPR_protocol). In brief, Huh7 cells were seeded in a 6-well plate, followed by transfection with 3 μg caspase-6 CRISPR–Cas9 KO plasmids and 3 μg HDR plasmid, or non-targeting control plasmids using Lipofectamine 3000. At 72 h post-transfection, caspase-6 knockout targeting cells were selected by medium containing 1.2 μg ml−1 puromycin. Caspase-6 knockout was verified with western blots. On the day of infection, Huh7 cells were infected by MERS-CoV at 1 MOI. At 2, 24 and 48 hpi, the cell lysates and supernatants were collected for RT–qPCR quantification and TCID50 assays.

RNA extraction and RT–qPCR

Cells were lysed in RL buffer and extracted with the MiniBEST Universal RNA Extraction Kit (TaKaRa). Viral RNA in the supernatant was extracted with the MiniBEST Viral RNA/DNA Extraction Kit (TaKaRa). RT–qPCR was performed with Transcriptor First Strand cDNA Synthesis Kit and LightCycler 480 master mix from Roche. Data was collected through LightCycler96 (version SW1.1). All primer and probe sequences were provided in Supplementary Table 2.

Plaque assays and TCID50 assays

Infectious titres of MERS-CoV and SARS-CoV-2 were determined with standard plaque assays46. In brief, Vero E6 cells were seeded in 24-well plates 1 day before the experiment. The collected supernatant samples were serially diluted and inoculated to the cells for 2 h at 37 °C. After inoculation, the cells were washed with PBS 3 times, and covered with 2% agarose/PBS mixed with 2× DMEM/2%FBS at 1:1 ratio. The cells were fixed after incubation at 37 °C for 72 h. Fixed samples were stained with 0.5% crystal violet in 25% ethanol/distilled water for 10 min for plaque visualization. In some experiments, infectious titres of coronaviruses were determined with standard TCID50 assays. In brief, Vero E6 cells were seeded in 96-well plates 1 day before the experiment. The collected supernatant samples were serially diluted and inoculated to the cells for 2 h at 37 °C. After inoculation, the cells were washed with PBS 3 times and incubated at 37 °C. After 72 hpi, virus titre was calculated using the Muench and Reed method.

siRNA and shRNA knockdown

On-Targetplus caspase-6 siRNA was obtained from Dharmacon (L-004406-00-0005). Transfection of siRNA on MDMs was performed using Lipofectamine RNAiMAX (Thermo Fisher Scientific). In brief, the cells were transfected with 50 nM caspase-6 siRNA for two consecutive days. At 24 h after the second siRNA transfection, the cells were collected in RIPA buffer for western blot analysis. In parallel, siRNA-transfected cells were challenged with MERS-CoV at 1 MOI for 1 h at 37 °C. Following the inoculation, the cells were washed with PBS and incubated for 24 h. The virus copy number at 24 hpi was determined with RT–qPCR. pLKO.1 lentiviral caspase-6 shRNA plasmid was obtained from Dharmacon. Transfection of caspase-6 shRNA plasmid, psPAX2 packaging plasmid and pMD2.G envelope plasmid on 293T cells was performed using Lipofectamine 3000 (Thermo Fisher Scientific) following manufacturer’s manual. In brief, 293T cells in 10 cm dishes were transfected with 6 μg caspase-6 shRNA plasmid, 4.5 μg packaging plasmid, and 1.5 μg envelope plasmid in FBS-supplemented DMEM medium, followed by aspirating supernatant at 6 h post-transfection and replacing with FBS-free medium. On the next day, the supernatant containing caspase-6 shRNA lentivirus particles was collected and was used to transduce A549 and BEAS2B cells. A549 and BEAS2B caspase-6 stable knockdowns cells were selected by 0.5 μg ml−1 and 0.7 μg ml−1 puromycin, respectively. The selected cells were challenged with MERS-CoV at 0.1 MOI for 1h at 37 °C. The virus copy number at 1 and 24 hpi was determined with RT–qPCR.

Caspase-6 activity assay

Huh7 cells were infected with MERS-CoV at 1 MOI for 12 h. In parallel, Huh7 cells were stimulated with STS at 1 μM for 6 h. Caspase-6 activity in the cell lysate was determined with the caspase-Glo-6 assay kit (Promega). The luminescence signal of caspase-6 activity was measured following manufacturer’s manual with a multilabel plate reader Victor X3 (Perkin-Elmer). Data was analysed with Kaleido (version 1.2) software.

Immunofluorescence and histology

Infected human and animal lung tissues were fixed overnight in 10% formalin50. The fixed samples were then embedded in paraffin with a TP1020 Leica semi-enclosed benchtop tissue processor and sectioned at 5 μm. Tissue sections were fished and dried to fix on Thermo Fisher Scientific Superfrost Plus slides at 37 °C overnight. Antigen retrieval was performed by heating the slides in antigen unmasking solution (Vector Laboratories) for 90 s. MERS-CoV and SARS-CoV-2 were detected with an in-house guinea pig anti-MERS-CoV N immune serum and an in-house rabbit anti-SARS-CoV-2-N immune serum, respectively. Cell nuclei were labelled with DAPI nucleic acid stain (Thermo Fisher Scientific). Alexa Fluor secondary antibodies were obtained from Thermo Fisher Scientific. Mounting was performed with the Diamond Prolong Antifade Mountant from Thermo Fisher Scientific. Images were captured with an Olympus BX53 fluorescence microscope (Olympus Life Science) or a Carl Zeiss LSM 780 confocal microscope in the faculty core facility of HKU using Olympus cellSens Dimension (version 1.17) and ZEISS (ZEN 2) microscope software, respectively. For H&E staining, mice and hamster lung tissue sections were stained with Gill’s H&E Y (Thermo Fisher Scientific). Images were captured with an Olympus BX53 fluorescence microscope. H&E-stained lung tissue sections were blinded for the identities of experimental settings and examined by a trained histopathologist. Lung pathology was graded on a scale of 0 (normal) to 4 (most severe) according to a grading system as previously described19.

Western blot

Cells were lysed with RIPA buffer (Thermo Fisher Scientific) with protease inhibitor (Roche, Basel, Switzerland). Proteins were separated by SDS–PAGE and transferred to PVDF membranes (Thermo Fisher Scientific). Specific primary antibodies were incubated with the blocked membranes at 4 °C overnight, followed by horseradish peroxidase (HRP)-conjugated secondary antibodies (Thermo Fisher Scientific) for 1 h at room temperature. The signal was developed with Immobilon Crescendo Western HRP Substrate (Merck Millipore) and detected using an automatic X-ray film processor (Advansta) or an Alliance Q9 Advanced imager (Uvitec). Raw western blot images were collected using Alliance Q9 software (v17-02) and provided as Supplementary Figs. 14.

Flow cytometry

BEAS2B cells were infected with MERS-CoV at 1 MOI. At 24 hpi, the cells were detached with 10 mM EDTA in PBS, fixed in 4% paraformaldehyde, followed by immunolabelling with an in-house guinea pig anti-MERS-CoV N immune serum and a rabbit anti-active caspase-3 antibody (560626, BD Biosciences) (1:500). Flow cytometry was performed using a BD FACSCanto II flow cytometer (BD) and data were analysed using FlowJo (version vX 0.7) (BD). The gating strategy was demonstrated in Supplementary Fig. 5.

IFNβ luciferase reporter assays

IFNβ luciferase reporter assays were performed as previously described26,27. In brief, 500 ng IFNβ luciferase reporter plasmid, 10 ng transfection efficiency control plasmid (pNL1.1.TK, Promega), 1 μg coronavirus N plasmids, 3 μg caspase-6 expression plasmid, together with or without 5 μg Poly(I:C) were co-transfected into 293T cells for 24 h. On the next day, the cells were collected for luciferase measurement with the dual-luciferase reporter assay system kit (Promega) according to the manufacturer’s protocol using a multilabel plate reader Victor X3 (Perkin-Elmer). Data were analysed with Kaleido (version 1.2) software. To investigate the point of action of the N fragments, IFNβ luciferase reporter plasmid pNL1.1.TK and MERS-CoV N(1–241) or MERS-CoV N(242–413) were co-transfected into 293T cells together with expression plasmids for RIG-IN, MAVS or TBK1. At 24 h post-transfection, the cells were collected for luciferase measurement.

Alignment of human, mouse and hamster caspase-6

Caspase-6 protein sequences of Homo sapiens (Uniprot ID: P55212), Mus musculus (Uniprot ID: O08738) and Mesocricetus auratus (Uniprot ID: A0A1U7QNN7) were downloaded from UniProt. Multiple sequence alignment was performed with MUSCLE (v3.8.31). The crystal structure of caspase-6 and VEID complex was retrieved from the Protein Data Bank (PDB code: 3OD5). Caspase-6 residues within 4 Å of VEID were defined as the binding sites and visualized with PyMOL (version 2.5).

Prediction of potential caspase-6 cleavage sites

The protein sequence of MERS-CoV N (NC_019843) was used for caspase-6 cleavage site analysis. Potential caspase-6 cleavage motifs on MERS-CoV N was determined based on published substrate specificity of caspase-629. The amino acid pattern [TVILENYF]..D was searched against the N sequence, where ‘.’ represented any amino acid.

Statistical analysis

Data in figures are mean ± s.d. Statistical comparison between different groups was performed by one-way ANOVA, two-way ANOVA, Student’s t-test or log-rank (Mantel–Cox) test using GraphPad Prism 9. Differences were considered statistically significant at P < 0.05.

Reporting summary

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