You are on page 1of 24

Articles

https://doi.org/10.1038/s41556-019-0416-0

Mitochondria-localised ZNFX1 functions as a


dsRNA sensor to initiate antiviral responses
through MAVS
Yao Wang1, Shaochun Yuan1, Xin Jia1, Yong Ge1, Tao Ling1, Meng Nie1, Xihong Lan1, Shangwu Chen1 and
Anlong Xu 1,2*

In the past two decades, emerging studies have suggested that DExD/H box helicases belonging to helicase superfamily 2
(SF2) play essential roles in antiviral innate immunity. However, the antiviral functions of helicase SF1, which shares a con-
served helicase core with SF2, are little understood. Here we demonstrate that zinc finger NFX1-type containing 1 (ZNFX1), a
helicase SF1, is an interferon (IFN)-stimulated, mitochondrial-localised dsRNA sensor that specifically restricts the replication
of RNA viruses. Upon virus infection, ZNFX1 immediately recognizes viral RNA through its Armadillo-type fold and P-loop
domain and then interacts with mitochondrial antiviral signalling protein to initiate the type I IFN response without depending
on retinoic acid-inducible gene I-like receptors (RLRs). In short, as is the case with interferon-stimulated genes (ISGs) alone,
ZNFX1 can induce IFN and ISG expression at an early stage of RNA virus infection to form a positively regulated loop of the well-
known RLR signalling. This provides another layer of understanding of the complexity of antiviral immunity.

T
he innate immune system is the first line of host defence against infected cell surfaces28,29. In addition to inhibiting a specific viral life
pathogen invasion. Upon viral infection, virus-derived nucleic cycle stage, some ISGs can enhance innate pathogen-sensing capa-
acids are mainly sensed by two cytosolic nucleic acid sen- bilities or positively regulate IFN signalling24. For example, poly-
sors, the DNA sensor cyclic guanosine monophosphate–adenosine glutamine binding protein 1 binds to reverse-transcribed HIV-1
monophosphate synthase (cGAS)1,2 and the retinoic acid-inducible DNA and interacts with cGAS to activate the type I IFN response30,
gene I (RIG-I)-like receptors (RLRs), including RIG-I and MDA53–6. RNF128 functions as an E3 ligase for K63-linked ubiquitination
In addition to these well-known nucleic acid sensors, many other and activation of TBK131 and human IFN-inducible oligoadenyl-
nucleic acid sensors, including several DExD/H box helicases, have ate synthetase-like protein mediates RIG-I activation by mimicking
been identified in a wide range of cell types to supplement the polyubiquitin32. Thus, ISGs exert diverse functions at multiple levels
functions of RLRs and cGAS7. For example, DDX60 is involved in of antiviral immunity.
RIG-I-dependent and -independent antiviral responses8, DDX1, To determine the antiviral functions of rarely studied helicase
DDX21 and DHX36 helicases form a complex with TRIF to sense SF1 and identify more ISGs, we analysed previously published
dsRNA in dendritic cells9, DHX9 mediates NLRP9b recognition of in vitro transcription-sequencing alternative polyadenylation sites
dsRNA stretches and forms inflammasome complexes to promote (IVT-SAPAS) data of vesicular stomatitis virus (VSV)-infected
the maturation of interleukin (IL)-18 and gasdermin D-induced monocyte-derived macrophages (MDMs) and identified ZNFX1
pyroptosis10 and NLRP6 binds viral RNA via DHX15 and interacts (a member of helicase SF1) as an ISG and mitochondrial-local-
with mitochondrial antiviral signalling protein (MAVS) to induce ised dsRNA sensor33. ZNFX1 can initiate IFN and ISG expression
type I/III interferons (IFNs)11. These emerging studies highlight immediately upon RNA virus infection by sensing viral RNAs and
the crucial roles of DExD/H box helicases, which belong to helicase interacting with MAVS, without depending on RLRs. The identi-
superfamily 2 (SF2), in antiviral immune responses. However, as a fication of ZNFX1 not only sheds light on the functions of RNA
different superfamily of many other helicases, helicase SF1 is rarely helicase SF1 in antiviral immunity, but also provides a target for
characterized in sensing pathogenic nucleic acids, although these antiviral therapy.
proteins share a conserved core composed of two tandem RecA
domains with ATPase activity and RNA binding ability12–14. Results
After binding viral-derived nucleic acids to cytosolic viral sen- Viral infection induces ZNFX1 expression. To identify undis-
sors, two adaptors, MAVS and STING, can be recruited to induce covered molecules involved in antiviral innate immunity, we ana-
the production of type I IFNs and a number of interferon stimulated lysed our previously published IVT-SAPAS data of VSV-infected
genes (ISGs) with antiviral activities15–24. For example, the myxovi- MDMs and found numerous genes with changes in their expres-
rus resistance 1 (Mx1) gene product is the inhibitor of virus entry25, sions (Extended Data Fig. 1a)33. By comparing VSV-infected human
the interferon-induced transmembrane (IFITM) family prevents MDM data and mouse peritoneal macrophage data, we found
infection before virus can traverse the lipid bilayer of the cell26, PKR some common genes, which were upregulated more than three-
impedes viral translation through phosphorylation of eIF2a elon- fold, including FFAR2, ZEB2, TIPARP, PTGS2, CMPK2, ZNFX1
gation factors27, while tetherin causes the retention of virions on and CSRNP1. We then knocked down these genes in A549 cells and

State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, School of Life Science, Sun Yat-sen University,
1

Guangzhou, China. 2School of Life Science, Beijing University of Chinese Medicine, Beijing, China. *e-mail: lssxal@mail.sysu.edu.cn

1346 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
subsequently infected the cells with VSV-eGFP (enhanced green IRF9 binding motifs. Next, these two reporter constructs were
fluorescence protein). Flow cytometry to detect the percentage transfected with increasing amounts of STAT1, STAT2, IRF1 or
of GFP-positive cells showed that knocking down ZNFX1, whose IRF9 expression plasmids into 293T cells. Reporter assays showed
antiviral roles have never been reported, facilitated VSV infection that the expression of reporter gene could be induced by the Znfx1
(Extended Data Fig. 1b). Many previous studies have suggested that promoter but not by a gene lacking the transcriptional factor bind-
DExD/H box RNA helicases, belonging to RNA helicase SF2, play ing sites in the presence of STAT1, STAT2, IRF1 or IRF9 (Fig. 1l).
diverse roles in antiviral immune responses7. ZNFX1, a representa- In all, ZNFX1 is an ISG whose upregulation upon viral infection is
tive of RNA helicase SF1, contains an Armadillo-type fold (ARM) dependent on the JAK1-STAT1 cascade.
domain, a P-loop helicase domain and a zinc finger (ZF) domain.
Because genes from helicases SF1 and SF2 share a conserved ZNFX1 is essential for innate anti-RNA virus in vitro and in vivo.
core structure, characterized by two tandem RecA domains, we To further explore the role of ZNFX1 in antiviral immune responses,
proposed that SF1 members may also play important roles in antivi- we silenced the endogenous expression of ZNFX1 in A549 and L929
ral immunity, as previously suggested13. cells. The cells were then infected with VSV-eGFP and subjected
To test this hypothesis, we analysed the gene expression pro- to flow cytometry analysis (Extended Data Fig. 2a,b). The results
files of SF1 and SF2 using IVT-SAPAS data and found a number show that knockdown of Znfx1 significantly enhanced VSV rep-
of genes in these two subfamilies with changes in expression upon lication both in A549 and L929 cells (Fig. 2a). Meanwhile, higher
VSV infection (Fig. 1a). Flow cytometry results showed that knock- VSV titres and increased VSV-G protein were observed in Znfx1
ing down MOV10, HELZ2, UPF1 and ZNFX1 in A549 cells led to silencing cells (Fig. 2b,c). Consistently, knockdown of Znfx1 expres-
markedly enhanced VSV replication, whereas SEXT knockdown sion significantly decreased the production of IFN-α and IFN-β
caused the opposite result (Extended Data Fig. 1c,d). MOV10, UPF1 protein in A549 cells after VSV infection or poly(I:C) stimulation
and SEXT have been implicated previously in antiviral immunity, (Extended Data Fig. 2c). In contrast, lower VSV mRNA and VSV
validating our screening approach based on RNAi and viral infec- titres in A549 cells were observed when ZNFX1 was overexpressed
tion, and suggesting that ZNFX1 is probably an undiscovered (Extended Data Fig. 2d).
antiviral molecule34–36. To obtain more functional evidence for ZNFX1 in antivi-
To obtain more insight into the role of ZNFX1 in antiviral ral responses, we employed a CRISPR-mediated genome editing
responses, qRT–PCR or immunoblot analyses were conducted to approach to knock out Znfx1 in 293T and A549 cells. Similar to the
obtain the upregulated expression pattern of ZNFX1 in A549 cells, transient silencing of Znfx1 in A549 or L929 cells, we found that
peripheral blood mononuclear cells (PBMCs) or mouse embryonic genome editing of Znfx1 in 293T and A549 cells enhanced VSV
fibroblast (MEF) cells upon VSV infection or poly(I:C) stimula- replication (Fig. 2d and Extended Data Fig. 2e,f). Meanwhile, over-
tion (Fig. 1b–d). The abundance of Znfx1 in mouse spleen and lung expression of ZNFX1 in wild-type or Znfx1−/− A549 cells restricted
and the upregulation in immune-related tissues including thymus, VSV replication (Fig. 2e). Further RNA sequencing analysis com-
lymph node, spleen and lung after intravenous injection of VSV paring the genes differentially expressed between wild-type and
were also demonstrated by qRT–PCR (Fig. 1e). We also conducted ZNFX1-overexpressed A549 cells showed the upregulated enrich-
statistical analyses of previously collected microarray data sets (GD ment of genes involved in negative regulation of viral replication
S4231/4214/1271/3210/6082/6063) and found that RNA levels and signal transduction of type I IFN (Fig. 2f). Interestingly, virus
of Znfx1 were significantly higher in patients with HIV infection mRNA levels were increased in Znfx1 knockdown A549 cells with
and monkeys with encephalitis caused by simian immunodefi- EMCV and influenza A virus (H1N1) infection, but not signifi-
ciency virus (SIV) when compared with healthy controls (Fig. 1f,g). cantly changed upon herpes simplex virus type 1 (HSV-1) infection,
Moreover, the transcription of Znfx1 is significantly increased after suggesting a broad anti-RNA-viral spectrum of ZNFX1 (Extended
different virus infections in different cell types, including macro- Data Fig. 2g).
phages, dendritic cells and epithelial cells (Extended Data Fig. 1e,f). To investigate the physiological role of ZNFX1 in vivo, we gener-
These data all indicate that ZNFX1 may be an antiviral molecule. ated Znfx1−/− C57BL/6J mice using CRISPR/Cas system (Extended
Data Fig. 3a–c). Znfx1−/− mice were viable, without obvious physi-
ZNFX1 is an IFN-stimulated gene. Because viral infection led to ological or behavioural abnormalities. Six- to eight-week-old mice
the enhancement of ZNFX1 expression at both mRNA and protein were used in this study. First, BMDMs from wild-type and Znfx1−/−
levels, we next examined whether ZNFX1 is an ISG by incubating mice were isolated and infected with VSV, EMCV, H1N1 or HSV-1.
A549 cells with human recombinant IFN-α and IFN-β. The results Compared to wild-type counterparts, Znfx1−/− BMDMs exhibited
of qRT–PCR and immunoblot analyses showed that the expression lower IFN-α or IFN-β secretion after RNA virus infection, includ-
of ZNFX1 could be induced by IFN-α and IFN-β (Fig. 1h,i). In con- ing VSV, EMCV and H1N1 but not HSV-1 (Fig. 2g and Extended
trast, the induction of Znfx1 mRNA by VSV could be inhibited by Data Fig. 2h). Consistently, higher virus mRNA levels were detected
the JAK1 inhibitor ruxolitinib and was almost abolished in Ifnar1−/− in VSV, EMCV and H1N1 infected Znfx1−/− BMDMs but not in
A549 cells (Fig. 1j,k). HSV-1 infected cells (Fig. 2h). Compared to wild-type counterparts,
To identify which nucleotides and region are important in deter- Znfx1−/− MEF and A549 cells exhibited lower IFN-β secretion after
mining Znfx1 transcription, bioinformatics analyses using the RNA virus infection of VSV and EMCV (Fig. 2i,j).
promoter 2.0 prediction server (http://www.cbs.dtu.dk/services/ To evaluate the importance of ZNFX1 in host defence against
promoter/), JASPAR (http://jaspar.genereg.net/) and CpG plot viral infection in vivo, wild-type and Znfx1−/− mice were injected
(http://www.ebi.ac.uk/Tools/seqstats/emboss_cpgplot/) were per- with VSV through the caudal vein. Enhancement of VSV replication
formed to identify the region between −1,064 and +63 of Znfx1 in lung and liver of Znfx1−/− mice was observed when compared with
containing TATA box, GC box and CAAT box, which are the char- wild-type mice (Fig. 3a). With reduced expression of Ifnb1 in lung
acteristics of a promoter. Further analyses of transcription factor and liver, lower secretion of IFN-α and IFN-β in serum was detected
binding sites revealed that the promoter of Znfx1 contains STAT1, in Znfx1−/− mice (Fig. 3b,c). Notably, no difference in IFN-α and
STAT2, IRF1 and IRF9 binding motifs in a relatively concentrated IFN-β in serum was observed between Znfx1−/− and wild-type mice
and overlapped region, as shown in Extended Data Fig. 1g. Based on after HSV-1 infection (Fig. 3c). Interestingly, one day post infec-
these analyses, we constructed a Znfx1 promoter reporter construct tion with VSV, 80% of Znfx1−/− mice suffered from conjunctivitis,
containing the region between −1,064 and +63 and a mutated which is the early symptom of VSV infection (Fig. 3d). In addition,
reporter construct with deletions of the STAT1, STAT2, IRF1 and more infiltration of inflammatory cells into the lungs of Znfx1−/−

Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology 1347


Articles NATURE CELL BIoLoGy

a MDM b c
VSV: 0 2 4 8 16 24 h 50 VSV: 0 4 8 16 24 h Poly(I:C) 0 4 8 16 24 h

expression of Znfx1
Relative mRNA
40 Znfx1 Znfx1

A549

A549
30
20 β-actin β-actin

10
0 Znfx1 Znfx1

MEF

MEF
VSV : – + + + + – – – – –
Poly(I:C) – – – – – – + + + + β-actin β-actin
Time (h): 0 4 8 16 24 0 4 8 16 24

d VSV: 0 4 8 16 24 h e
245 kDa Spleen Thymus
IB: ZNFX1 Lung Lymph node
A549

Kideny Spleen
Heart Lung
IB: GAPDH 35 kDa Bowel Bowel
Liver Muscle
Thymus Kidney
Lymph node Brain Virus infection
IB: ZNFX1 245 kDa
PBMC

Brain Heart Uninfected


–2 0 2 Muscle Liver
SF2: DEAD-box IB: GAPDH 35 kDa 0 30 60 90 120 0 5 10 15 20 25
MDM Znfx1 mRNA (relative) Znfx1 mRNA (relative)
VSV: 0 2 4 8 16 24 h
f 2.0 P = 0.0123 1.0 P = 0.0004 Uninfected (UI) g 11 P = 3.77 × 10–15 10 P = 4.50 × 10–12 Uninfected (UI)
transformed count

transformed count

transformed count

transformed count
HIV infection SIV infection
1.5 10 8
0.5
1.0
Znfx1

Znfx1
Rig-I

Rig-I
9 6
0.5
–1.5 0 1.5 0
0 8 4
SF1 RNA helicase
–0.5 –0.5 7 2
UI HIV UI HIV UI SIV UI SIV
h 15
expression of Znfx1

i
Relative mRNA

IFN-α: 0 4 8 16 24 h IFN-β: 0 4 8 16 24 h
10
IB: ZNFX1 245 kDa IB: ZNFX1 245 kDa

5
A549

IB: RIG-I 100 kDa IB: RIG-I 100 kDa


0
IFN-α: – + + + + – – – – – IB: β-actin 45 kDa IB: β-actin 45 kDa
IFN-β: – – – – – – + + + +
Time (h): 0 4 8 16 24 0 4 8 16 24

j UT k WT l
wt Znfx1-Luc
Rux Ifnar –/–
ΔZnfx1 promotor-Luc
P = 0.0065 –6 P = 0.0003 P = 7.44 × 10–6
15 P = 1.69 × 10
15 4 P = 0.0009 4 10 80 P = 4.40 × 10–6
expression of Znfx1

expression of Znfx1

P = 0.0043 P = 0.0003
Relative mRNA

Relative mRNA

Rel. Znfx1-Luc

Rel. Znfx1-Luc

Rel. Znfx1-Luc

Rel. Znfx1-Luc

3 3 8 60
10 10
activity

activity

activity

activity

6
2 2 40
4
5 5
1 1 2 20
0 0 0 0 0 0
VSV: 0 4 8h VSV: 0 4 8h EV + – – – + – – – EV + – – – + – – – EV + – – – + – – – EV + – – – + – – –
STAT1 – – STAT2 – – IRF1 – – IRF9 – –

Fig. 1 | Viral infection induces Znfx1 expression, an IFN-stimulated gene. a, MDMs were infected with VSV, then IVT-SAPAS was performed. The heatmap
shows an abundance change of RNA helicases SF1 and SF2 in virus-infected MDMs. b, A549 cells were challenged with VSV or stimulated with poly(I:C)
for the indicated time. The mRNA levels were analysed by qRT–PCR. c, A549 and MEF cells were challenged with VSV or stimulated with poly(I:C) for the
indicated time. The mRNA levels were analysed by RT–PCR. d, Immunoblot (IB) analysis of ZNFX1 protein in A549 cells and peripheral blood mononuclear
cells (PBMCs) upon VSV infection at the indicated time points. e, Left: Distribution of Znfx1 in various mice tissues was determined using qRT–PCR analysis;
the transcription of Znfx1 in muscle was set to 1, values of other tissues are expressed as the ratio compared with muscle. Right: Znfx1 mRNA expression
change in various mice tissues following intravenous injection of VSV determined by qRT–PCR. f, RNA levels of Znfx1 are significantly higher in patients
with HIV infection compared with uninfected controls (UI) (n = 9 independent samples for uninfected controls, n = 14 independent samples for HIV
infection). g, RNA levels of Znfx1 and Rig-I in encephalitis monkeys infected with simian immunodeficiency virus (SIV) compared with healthy controls
(n = 18 independent samples). h, Znfx1 expression was measured by qRT–PCR in A549 cells after IFN-α or IFN-β treatment for the indicated time points.
i, Immunoblot analysis of ZNFX1 and RIG-I expression in A549 cells treated with human recombinant IFN-α or IFN-β for the indicated time points. j, Znfx1
expression was measured by qRT–PCR in A549 cells after ruxolitinib (Rux) pretreatment for 2 h, then infected with VSV for time points. k, Znfx1 expression
was measured by qRT–PCR in wild-type (wt) and Ifnar1−/− cells after VSV infection. l, Luciferase activities determined from 293T cells transfected with Znfx1
promoter reporter (wt Znfx1-Luc) or its mutation (ΔZnfx1 promotor-Luc) together with increasing amounts of Flag-STAT1, STAT2, IRF1, IRF9 expressing
plasmids. For c,d,i, experiments were repeated at least three times independently, with similar results obtained. For b,e,h,j–l, n = 3 independent experiments.
Means ± s.d. are shown, and P values were calculated using two-tailed unpaired Student’s t-test.

1348 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
a VSV-eGFP (MOI: 2) 6 h VSV-eGFP (MOI: 5) 6 h c VSV-eGFP
d
VSV-eGFP (MOI: 0.5) 12 h VSV-eGFP (MOI: 1) 12 h A549
65 kDa

VSV-GFP+ (% of total)
–5
P = 0.0013 P = 1.23 × 10–6 IB: VSV-G P = 7.34 × 10
80 100 100 WT

A549
A549 VSV-GFP+

P = 0.0028
80 80 Rig i –/–
(% of total)

(% of total)
60

VSV-GFP+
P = 4.60 × 10–6 IB: GAPDH 35 kDa
60 60 Mda5 –/–

L929
40 65 kDa Znfx1–/–
40 IB: VSV-G 40

L929
20 20 20
0 0 IB: GAPDH 35 kDa 0
Uninfect. VSV
N X1

N X1

1
ZN VS

ZN VS

ZN VS

ZN VS
C STM

C STM

C STM
A l

A l

C STM
A l

A l
M tro

M tro

M tro

M tro

1
S
FX

FX

TM

M l
tro

FX
AV
F

F
on

on

on

on

on
-S
-

ZN
N

N
C
VSV-eGFP VSV-eGFP VSV-eGFP VSV-eGFP

b P = 0.0002 P = 3.19 × 10
–6 e P = 0.0003
f
30 40 80
p.f.u. ml−1 (×106)

p.f.u. ml−1 (×106)

P = 0.0023 WT Negative regulation of viral replication 4.60 × 10−8


Znfx1–/–

(% of total)
VSV-GFP+
30 60
20 Type I IFN signalling 1.60 × 10−7
A549

L929

20 40 EGFR signalling 2.50 × 10−7


10 Defence response to virus 2.60 × 10−7
10 20
TRIF-dependent TLR signalling 3.80 × 10−7
0 0 0
EV + + – + + – – 0 2 4 6 8
1
ZN VS

M rol

1
ZN VS
M trol

FX

FX

VSV – + + – + + +
t
A
A

on
on

P value (–log10)
C
C

ZNFX1 – – + – – – +
VSV-eGFP VSV-eGFP RIG-I – – – – – + –

g –6
h P = 0.0042 P = 0.0019 P = 4.24 × 10–5 P = 0.2616
P = 1.36 × 10–6

Rel. mRNA of HSV-1


Rel. mRNA of EMCV
P = 1.09 × 10

Rel. mRNA of H1N1


3,000 2,000 10,000
Rel. mRNA of VSV

2,500 2,500 8,000 WT


WT
IFN-β (pg ml−1)

IFN-β (pg ml−1)

2,000 2,000 1,500 8,000 Znfx1–/–


Znfx1–/– 6,000
2,000
1,500 1,500 6,000
4,000 1,000
1,000 1,000 1,000 4,000
500 500 2,000 500 2,000
0 0 0 0 0 0
VSV EMCV VSV EMCV H1N1 HSV-1
i
500
P = 7.89 × 10–6
800
P = 0.6519
3,000 P = 0.0001 P = 0.0006 j 1,500 P = 0.0003 P = 3.42 × 10 –6

WT

IFN-β (pg ml−1)


IFN-β (pg ml−1)
IFN-β (pg ml−1)

IFN-β (pg ml−1)

400 600 WT
2,000 Rig i –/– 1,000
300 Mda5 –/– Rig i –/–
400
200 Znfx1–/– Mda5 –/–
1,000 500
100 200 Znfx1–/–
0 0 0 0
H1N1 HSV-1 Uninfect. VSV EMCV Uninfect. VSV EMCV

Fig. 2 | ZNFX1 deficiency impairs cellular antiviral RNA response. a, FACS analysis of A549 and L929 cells transfected with control small interfering
(si)RNA or the indicated siRNAs followed by VSV-eGFP infection at different multiplicity of infection (MOI) (n = 4 independent experiments). b, Plaque
assay of VSV titres in cell supernatants after VSV infection for 12 h (n = 4 independent experiments). c, Immunoblot analysis of VSV-G protein expression
in A549 and L929 cells infected with VSV for 12 h. The experiments were repeated three times, independently, with similar results. d, FACS analysis
of wild-type, Znfx1−/−, Rig-I−/− or Mda5−/− cells infected by VSV-eGFP. e, FACS analysis of wild-type and Znfx1−/− A549 cells transfected with RIG-I,
ZNFX1 expressing plasmids or empty vector (EV) following VSV-eGFP infection. f, Gene ontology analysis of the pathways among genes significantly
upregulated between ZNFX1 overexpressed cells and wild-type A549 cells. The Kolmogorov–Smirnov test was used to calculated P values. g, Enzyme-
linked immunosorbent assay (ELISA) of IFN-β in supernatants of bone marrow-derived macrophages (BMDMs) from wild-type and Znfx1−/− mice infected
with VSV, encephalomyocarditis virus (EMCV), influenza A virus (H1N1) or herpes simplex virus type 1 (HSV-1) for 16 h (n = 5 independent experiments).
h, Viral mRNA transcripts were analysed in VSV, EMCV, H1N1 and HSV-1 infected wild-type and Znfx1−/− BMDMs by qRT–PCR analysis. i, ELISA of IFN-β in
supernatants of MEFs from wild-type and Znfx1−/−, Rig-I−/−, Mda5−/− mice infected with VSV and EMCV. j, ELISA of IFN-β in supernatants of wild-type and
Znfx1−/−, Rig-I−/−, Mda5−/− A549 cells infected with VSV and EMCV. For d,e,h–j, n = 3 independent experiments. All data are presented as mean ± s.d.
P values were calculated using two-tailed unpaired Student’s t-test.

mice following VSV infection was observed (Fig. 3e) and Znfx1−/− the activation of Ifnb1 promoter triggered by VSV, two types of
mice were less resistant to VSV infection in overall survival assays poly(I:C) or poly(dA:dT) in a dose-dependent manner (Fig. 4b,c).
(Fig. 3f). These data suggest that Znfx1 deficiency has impaired In contrast, knockout of Znfx1 decreased VSV-induced activation of
innate immune responses against RNA virus infection by producing Ifnb1 reporter and ISRE reporter in 293T cells (Fig. 4d). Meanwhile,
less type I IFNs in vivo. In addition, we suggested that ZNFX1 exerts transcription of Ifnb1 and ISGs was induced in a dose- and time-
its antiviral properties specifically on RNA viruses. dependent manner when ZNFX1 was overexpressed alone in A549
cells but reduced in the absence of ZNFX1 (Fig. 4e and Extended
ZNFX1 initiates IFN-based antiviral responses. To investigate the Data Fig. 3d,e), while protein expression of a number of ISGs was
role of ZNFX1 in the IFN signalling, luciferase reporter assays were reduced in Znfx1−/− A549 and MEF cells when compared with wild-
conducted by transiently transfecting the A549 cells with increas- type cells, respectively (Fig. 4f). Consistently, the phosphorylation
ing amounts of ZNFX1 expression plasmid and Ifnb1-reporter or of TBK1 and IRF3 in Znfx1−/− macrophages upon VSV infection
IFN-stimulated response element (ISRE) reporter plasmids fol- was lower than that in wild-type macrophages (Fig. 4g). The dimer-
lowed with or without VSV challenge. The results showed that ization of IRF3 was also lower in VSV-infected Znfx1−/− 293T cells
overexpression of ZNFX1 activated the Ifnb1 promoter and ISRE than that in wild-type cells (Fig. 4h). Notably, restricted replication
reporter slightly (Fig. 4a). Moreover, ZNFX1 significantly enhanced of VSV was observed in wild-type but not in Ifnar1−/− A549 cells

Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology 1349


Articles NATURE CELL BIoLoGy

a b P = 0.0005
15 15 200 800 WT

replicates (fold ×103)

replicates (fold ×103)


P = 0.0008 WT –/–
P = 0.0009 Znfx1
P = 8.69 × 10–6 Znfx1
–/–

Rel. mRNA of

Rel. mRNA of
150 600
VSV RNA

VSV RNA
10 10

Ifnb1

Ifnb1
100 400
5 5
50 200

0 0 0 0
PBS VSV PBS VSV PBS VSV PBS VSV
Lung Liver Lung Liver

c WT Znfx1–/–
d
P = 3.12 ×10–9 VSV injection 24 h
P = 0.1361
1,800
IFN-α (pg ml−1) 800 –11
P = 7.65 ×10 250 200 P = 0.9430

IFN-α (pg ml−1)


IFN-β (pg ml−1)
IFN-β (pg ml−1)

600 200 150


1,200
150
400 100
100
600
200 50 50

0 0 0 0
PBS VSV PBS VSV PBS HSV-1 PBS HSV-1 WT Znfx1–/–

e Znfx1+/+ Znfx1–/–
f
Znfx1+/+
100 Znfx1–/–

80
Survival (%)
PBS

60
40
P = 0.0283
20
0
0 1 2 3 4 5
Time after injection (days)
VSV

Fig. 3 | ZNFX1 is essential for host defence against RNA virus in mice. a, VSV mRNA transcripts were analysed by qRT–PCR in the lung and liver from
wild-type and Znfx1−/− mice with VSV infection by intravenous injection (2 × 107 p.f.u. per g body weight, n = 4 mice per group). b, qRT–PCR analysis of
Ifnb1 mRNA in the lung and liver from wild-type and Znfx1−/− mice (n = 4 mice per group). c, IFN-α and IFN-β production in sera from wild-type mice and
Znfx1−/− mice after intravenous injection with VSV and HSV-1 (8 or 4 mice per group). d, One day post infection with VSV, Znfx1−/− mice suffered from
conjunctivitis, unlike wild-type mice. e, Haematoxylin & eosin staining of lung sections from wild-type and Znfx1−/− mice infected with VSV for 24 h. Scale
bars, 80 μm. The experiments were repeated three times, independently, with similar results. f, Survival of wild-type and Znfx1−/− mice (n = 10 mice per
group) after intravenous injection of VSV (2 × 107 p.f.u. per g body weight). Data in a–c are presented as mean ± s.d. Statistical differences were detected
with two-tailed unpaired Student’s t-tests in a–c. A Gehan–Breslow–Wilcoxon test was applied in f.

(Fig. 4i). All these data suggest that ZNFX1 is involved in antiviral Pulldown assays were performed to show that ZNFX1 and RIG-I
immune responses by inducing type I IFN production and its anti- proteins bind to poly(I:C) but not poly(C) (Fig. 5g). Additionally,
viral property also depends on IFN signalling. cGAS but not ZNFX1 binds to both single- and double-stranded
DNA (ISD, a 45 bp double-stranded DNA; Fig. 5h). Competition
ZNFX1 directly binds to viral RNA. Many genes belonging to experiments revealed the preferable binding of ZNFX1 proteins to
RNA helicase SF2 have been identified as RNA virus sensors that HMW poly(I:C), but not LMW poly(I:C), poly(dA:dT) or 5′ppp-
bind viral RNA, so we assumed that ZNFX1 belonging to SF1 may dsRNA (Fig. 5i). To map the poly(I:C)-binding sites of ZNFX1, we
also bind viral RNA as an RNA sensor. To verify this hypothesis, we prepared truncated versions of ZNFX1 and performed poly(I:C)
first overexpressed ZNFX1 in 293T cells and then performed pull- pulldown experiments. The results showed that the ARM and
down and qRT–PCR assays to determine the high binding ability of P-loop domains, but not the ZF domain, were required for the
ZNFX1 with VSV-RNA (Fig. 5a,b). A similar assay also detected the binding of ZNFX1 to poly(I:C) (Fig. 5j). Meanwhile, we observed
binding ability of endogenous ZNFX1 to VSV, EMCV and Senda enhanced oligomerization of ZNFX1 after viral infection (Fig. 5k,l).
virus (SeV) RNAs in A549 cells (Fig. 5c). RNA immunoprecipita- Thus, ZNFX1 functions as an RNA virus sensor, which prefers to
tion (RIP)-sequencing analysis by isolating RNA from immunopre- bind the long RNA molecules.
cipitates with antibodies against endogenous ZNFX1 also mapped
ZNFX1-associated RNA to VSV genome (Fig. 5d), preferentially to ZNFX1 localises to mitochondria and interacts with MAVS. To
a segment of VSV strand around 6,000–8,000 nt (Fig. 5e). Notably, determine the subcellular localisation of ZNFX1, we separated
when ZNFX1-associated RNA was transfected into A549 cells, VSV-infected cells into cytoplasmic and nuclear fractions, and
induced transcription of Ifnb1 and induced expression of Ifnb1 pro- detected ZNFX1 protein mainly in the cytoplasm (Fig. 6a, upper
moter-based reporter were observed by qRT–PCR and luciferase panel). Previous studies about ARM-containing proteins, ALEX3
activity analyses (Fig. 5f). and SARM1, have indicated these proteins to be mitochondrially
To further assess the nature of viral RNA bound by ZNFX1, we localised. Thus, we next determined, by immunoblot analyses,
first incubated nucleic acid analogue linked beads with a cells lysis whether ZNFX1, an ARM-containing protein, can also localise to
of 293T overexpressed with Flag-tagged ZNFX1, RIG-I or MAVS. this immune-related organelle37–39. The results show the increased

1350 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
a b VSV (MOI:0.1) d
VSV (MOI:0.5) WT ZNFX1–/–
P = 0.0009 P = 0.0006 P = 0.0006 P = 0.0097
8 6 200 P = 8.29 × 10
–5 8 P = 0.0140 400

Rel. IFN-β-Luc
Rel. IFN-β-Luc

Rel. IFN-β-Luc
Rel. ISRE-Luc
Rel. ISRE-Luc
P = 0.0006
6 150 6 300
4

activity

activity
activity

activity
activity
4 100 4 200
2
2 50 2 100
0 0 0 0 0
EV + – – + – – EV + + – – + + – – + + – – + + – – VSV – –
ZNFX1 – – VSV – + + + – + + + – + + + – + + +
ZNFX1 – – – – – – – – P = 0.0060
245 kDa 245 kDa 150
IB: α-Flag

Rel. ISRE-Luc
100

activity
IB: GAPDH 35 kDa 35 kDa

50
c –5 P = 0.0002
P = 0.0001 P = 0.0038 P = 5.80 × 10 15 0
120
VSV – –
Rel. IFN-β-Luc

90 Rel. ISRE-Luc P = 0.0040 P = 0.0028 Poly(I:C) (HMW)


10
activity
activity

Poly(I:C) (LMW)
60
Poly(dA:dT)
5
30
e
0 0 ZNFX1: ZNFX1: 12 24 36 h
ZNFX1 – – – – – – – – – – – – Ifnb1 Ifnb1
Poly(I:C) (HMW) – + + + – – – – – – – – – + + + – – – – – – – –
Poly(I:C) (LMW) – – – – – + + + – – – – – – – – – + + + – – – – Rig-I Rig-I
Poly(dA:dT) – – – – – – – – – + + + – – – – – – – – – + + +
Ifi44 Ifi44
IB: α-Flag 245 kDa 245 kDa
Isg15 Isg15

IB: GAPDH 35 kDa 35 kDa Ifit1 Ifit1

Ifit2 Ifit2
f –/–
g Peritoneal macrophage
WT ZNFX1 Ifit3 Ifit3
VSV 0 2 4 8 0 2 4 8h WT Znfx1–/– Ifih1 Ifih1
IB: ZNFX1 245 kDa VSV 0 1 2 4 0 1 2 4h
Oas2 Oas2
IB: p-IRF3 45 kDa
IB: MDA5 135 kDa Mx1 Mx1

IB: RIG-I 100 kDa IB: IRF3 45 kDa Irf7 Irf7


A549

Znfx1 Znfx1
IB: Mx1 75 kDa IB: p-TBK1 75 kDa

65 kDa 0 5 10 4 8 12
IB: OAS1 IB: TBK1 75 kDa log2
(fold change)
IB: GAPDH 35 kDa
IB: GAPDH 35 kDa
i
WT Znfx1 –/– WT Ifnar1–/–
h P = 0.9733
VSV-GFP+ (% of total)

VSV 0 2 4 8 0 2 4 8h WT ZNFX1 –/–


80
IB: RIG-I 100 kDa VSV: 0 2 4 8 0 2 4 8h 60 P = 0.0001
MEF

Native-gel

IRF3 dimer 40
IB: GAPDH 35 kDa
20
IRF3 monomer
0
245 kDa EV + – – + – –
IB: ZNFX1
SDS–PAGE

VSV – + + – + +
ZNFX1 – – + – – +
IB: IRF3 45 kDa

IB: GAPDH 35 kDa

Fig. 4 | ZNFX1 initiates IFN-based antiviral responses. a,b, A549 cells were transfected with IFN-β-Luc or ISRE-Luc with increasing amounts of ZNFX1
plasmid for 24 h, and then uninfected (a) or infected with VSV for 16 h before luciferase assays (b). c, Luciferase activity analysis of A549 cells that were
transfected with the indicated luciferase reporters followed by stimulation with low-molecular-weight (LMW) poly(I:C), high-molecular-weight (HMW)
poly(I:C) and poly(dA:dT) for another 16 h. d, Luciferase activity of Ifnb1 or ISRE in wild-type and Znfx1−/− 293T cells that were infected with increasing
amounts of VSV for 24 h. e, qRT–PCR analysis of Ifnb1 and ISGs in A549 cells transfected with ZNFX1-expressing plasmids with increasing amounts
(125, 250 or 500 ng per well) for 24 h (left) or with 500 ng per well for the indicated time points (right). f, Immunoblot analysis of ISGs level in wild-
type and Znfx1−/− A549 cells or RIG-I expression in wild-type and Znfx1−/− MEFs infected by VSV for the indicated time points. g, Immunoblot analysis of
phosphorylated (p-) IRF3 and (p-) TBK1 in peritoneal macrophages from wild-type and Znfx1−/− mice upon VSV infection for the indicated time points.
h, Immunoblot analysis of IRF3 dimerization in wild-type and Znfx1−/− 293T cells upon VSV infection for the indicated time points with native PAGE. i, FACS
analysis of wild-type or Ifnar1−/− A549 cells transfected with ZNFX1-expressing plasmids or empty vector (EV) for 24 h followed by VSV-eGFP infection.
For f–h, the experiments were repeated three times, independently, with similar results obtained. Data in a–e,i are from three independent experiments
(mean ± s.d.). P values were calculated using two-tailed unpaired Student’s t-test.

Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology 1351


Articles NATURE CELL BIoLoGy
a VSV infection b 30 P = 0.0002 c
P = 0.0009 P = 4.18 × 10–6 –5
P = 6.06 × 10
8 6 25

VSV RNA
20 20

EMCV RNA
Bound
6

VSV RNA

SeV RNA
4

Bound

Bound

Bound
15
10 4
2 10
ZNFX1-Flag 2 5
0
RIG-I-Flag Flag or ctrl IP 0 0 0
MDA5-Flag 245 kDa IP:

G
G

1
IB: α-Flag

FX

FX

FX
Ig

Ig
Ig
135 kDa

ZN

ZN

ZN
RNA extraction 100 kDa

ag

ag

g
FX gG

la
Test for IFN stimulatory

Fl

Fl

-F
I
1-

-I-

A5
activity Test for VSV RNA

IG

D
R
ZN

M
d VSV genome (nt) f
60 25 ng 25 ng
(+) strand 40 P = 3.96 × 10
–5 50

Rel. IFN-β-Luc
50 ng P = 0.0005 50 ng

Rel. mRNA of
40
Relative numbers

30 40
P = 0.0026

activity
20 30

Ifnb1
20 P = 0.0001
0 20
10 10
−20
0 0
−40

A
er

er
ZN -RN

ZN -RN

N
(–) strand

N
at

at
R

-R

-R
W

W
−60

1-

-I-

1-

-I-
G

A5

A5
FX

FX
IG

IG
Ig

Ig
D

D
R

R
M

M
0
0

0
0

0
00

00

00

00

,0
2,

4,

6,

8,

10

e g ZNFX1 RIG-I MAVS h


) C)
IgG
I:C

)
I: ZNFX1 cGAS

C
)

ly )
3 ly( ly(
(C

(C

Po (C
(I:
Bound VSV RNA

ZNFX1 P = 0.0009 ly o Po

ly

ly
Pulldown: P Pulldown: – S D – S D
Po

Po

Po
(IP/input)

2
245 kDa
1
IB: α-Flag 100 kDa IB: α-Flag
0 65 kDa
VSV gene: N NS M G G L L L 75 kDa
VSV strand: 0–1 K 1–2 K 2–3 K 3–4 K 4–5 K 5–6 K 6–8 K 8–10 K 245 kDa 245 kDa
WCL
WCL
IB: α-Flag 100 kDa
IB: α-Flag
65 kDa
75 kDa
i Poly(I:C)(HMW) Poly(I:C)(LMW)
Competitor – –
IP: Poly(I:C) 245 kDa j ZNFX1-FL 1 ARM P-loop NH ZF 1918
IB: anti-Flag
ZNFX1-F1 580 P-loop NH ZF 1918
Poly(dA:dT) 5’-ppp dsRNA
Competitor: – – ZNFX1-F2 1261 ZF 1918

IP: Poly(I:C) 245 kDa ZNFX1-F3 1 ARM P-loop NH 1261

IB: anti-Flag ZNFX1-F4 580 P-loop NH 1261

ZNFX1-F5 1 ARM

k l ZNFX1-Flag – + + 245 kDa


ZNFX1-Flag + + + VSV – – + 135 kDa
ZNFX1-HA – + + IP: Poly(I:C) 100 kDa
IB: α-HA 75 kDa
VSV – – +
IP: α-Flag

IB: α-HA 245 kDa IB: α-Flag


ZNFX1
245 kDa
IB: α-Flag 245 kDa aggregates
Input: 135 kDa
SDD–PAGE IB: α-HA 100 kDa
IB: α-HA 245 kDa
75 kDa
WCL

IB: α-Flag 245 kDa 245 kDa SDS–PAGE


FL-HA

F1-HA

F2-HA

F3-HA

F4-HA

F5-HA

Fig. 5 | ZNFX1 binds to viral RNA. a, Schematic of the experimental set-up for ZNFX1, RIG-I and MDA5 immunoprecipitation. b, VSV-infected 293T
cells were transfected with the indicated plasmids, immunoprecipitated and analysed by qRT–PCR to detect bound VSV-RNA. c, Endogenous ZNFX1-
immunoprecipitated RNA was analysed by qRT–PCR using VSV-, EMCV- and SeV-infected A549 cells. d, RIP-sequencing mapped endogenous ZNFX1-
associated sequences to the VSV genome. The data shown are normalized with sequences from the input sample. + and − RNA is shown in red and blue,
respectively. e, qRT–PCR analysis of endogenous ZNFX1-immunoprecipitated RNA in VSV-infected A549 cells, the x axis corresponds to the positions in
the VSV genome, includeing of five coding genes. f, ZNFX1 co-immunoprecipitated RNA was first transfected into A549 cells, then luciferase activity and
mRNA transcripts of Ifnb1 were assessed. g, Flag-tagged ZNFX1, RIG-I or MAVS plasmids were transfected into 293T cells, then whole-cell lysates
(WSL) were incubated with agarose bead-conjugated poly(C) or poly(I:C). The proteins bound to agarose beads were analysed by immunoblotting.
h, Flag-tagged ZNFX1 or cGAS plasmids were transfected into 293T cells and agarose bead-conjugated DNA was incubated with whole-cell lysates,
then the bound proteins were analysed by immunoblotting (S, single-stranded DNA; D, double-stranded DNA). i, Flag-ZNFX1 recombinant protein
was incubated with agarose beads-conjugated poly(I:C) in the absence or presence of increasing amounts (0.5, 5 and 50 μg ml−1) of unlabelled HMW-
poly(I:C), LMW-poly(I:C), poly(dA:dT) or 5′ppp-dsRNA. Bound complexes were pelleted by centrifugation and analysed by immunoblot. j, Schematics
of ZNFX1 and its serial deletion mutants. The truncated versions of HA-tagged ZNFX1 were transfected into 293T cells, then whole-cell lysates were
incubated with agarose bead-conjugated poly(I:C). Bound proteins were analysed by immunoblotting. k, Co-immunoprecipitation and immunoblot
analysis of 293T cells transfected with Flag-ZNFX1 and HA-ZNFX1. l, Immunoblot analysis of ZNFX1 in 293T cells transfected with Flag-ZNFX1 plasmids
by SDD–PAGE or SDS–PAGE assay. Data in b,c,e,f are presented as mean ± s.d. (n = 3 independent experiments). P values were calculated using two-tailed
unpaired Student’s t-test. For g–l, the experiments were repeated three times, independently, with similar results obtained.

1352 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
a Cyt. Nuc. b Mock VSV 8 h VSV 16 h
c
Mito.
VSV – + – + Proteinase K – + +

ZNFX1-Flag
IB: ZNFX1 245 kDa 1% Triton – – +
IB: ZNFX1 245 kDa
IB: Lamin B1 65 kDa
IB: MAVS 75 kDa
IB: β-actin 45 kDa
IB: COX IV 15 kDa
Cyto. Mito.
Mito.

Mito
VSV – + – +
Alkali – + +
IB: ZNFX1 245 kDa M P S
IB: ZNFX1 245 kDa
IB: β-actin 45 kDa

IB: COX IV IB: MAVS 75 kDa

Merge
15 kDa
IB: COX IV 15 kDa
d
M 88

G
ST S

Flag-adaptor
IN
M F

AV
yD
I
EV
TR

ZNFX1-HA + + + + + e IP IgG ZNFX1 IgG ZNFX1 f IP IgG MAVS IgG MAVS


IP: α-Flag 245 kDa WT Znfx1–/– WT Mavs–/–
IB: α-HA
VSV – – + – – + VSV – – + – – +
100 kDa
245 kDa IB: ZNFX1 245 kDa
IP: α-Flag * 75 kDa IB: ZNFX1
IB: α-Flag
35 kDa IB: MAVS 75 kDa IB: MAVS 75 kDa

IB: α-HA 245 kDa IB: ZNFX1 245 kDa IB: ZNFX1 245 kDa
WCL

WCL
WCL

100 kDa IB: MAVS 75 kDa IB: MAVS 75 kDa


* 75 kDa
IB: α-Flag IB: GAPDH 35 kDa IB: GAPDH 35 kDa

35 kDa
h j
g Ifnb1
ZNFX1-Flag

IP IgG MAVS Rig-I


VSV 0 0 4 8 12 h
Ifi44
245 kDa 11
IB: ZNFX1

log2(fold change)
Ifit1
75 kDa
MAVS-GFP

IB: MAVS Ifit2


65 kDa 6.5
IgG (H) Ifit3
Ifih1
2
IB: ZNFX1 245 kDa Oas2
Isg15
Merge
WCL

IB: MAVS 75 kDa


Mx1
IB: GAPDH 35 kDa
245 kDa
Mock VSV 8 h 135 kDa

i k 100 kDa
Δ D
S- AR

75 kDa
TM

HA-ZNFX1 – FL F1 F2 F3 F4 F5
AV C
M -Δ

FL F1 F2 F3 F4 F5
S

Flag-truncation
S

Flag-MAVS + + + + + + +
AV
AV
M

EV + – – – – – –
M

HA-ZNFX1 + + +
IP: α-HA 75 kDa IP: α-Flag 245 kDa
IB: α-Flag
IB: α-HA
245 kDa
IP: α-HA
135 kDa IP: α-Flag
100 kDa 75 kDa
75 kDa IB: α-Flag
IB: α-HA

IB: α-Flag 75 kDa


IB: α-HA 245 kDa
WCL

245 kDa
135 kDa
WCL

IB: α-HA IB: α-Flag 75 kDa


75 kDa

Fig. 6 | ZNFX1 localises to mitochondria and interacts with MAVS. a, Endogenous ZNFX1 was detected in mitochondrial fractions but not nucleic fractions
of A549 cells by immunoblot analysis. b, Flag-ZNFX1 was expressed in A549 cells for 24 h, then the cells were challenged with VSV. Immunofluorescence
with anti-Flag antibody was used to detect the overexpressed protein (green). Mitochondria were identified by mitochondrial marker (red). Scale bar,
10 μm. c, Mitochondrial fraction isolated from A549 cells was treated with proteinase K (100 ng ml−1) in the absence or presence of 1% TritonX-100
for 15 min on ice, and the reactants were developed by immunoblot analysis. Alkali extraction was carried out using Na2CO3 to treat the mitochondrial
fraction isolated from A549 cells. Both the soluble protein fraction (S) and integral membrane protein fraction (P) were isolated and used for immunoblot
analysis. ‘M’ indicates untreated control mitochondrial sample. d, Co-immunoprecipitation of extracts of 293T cells transfected with Flag-tagged TRIF,
MyD88, MAVS and STING, together with HA-ZNFX1. e–g, Co-immunoprecipitation and immunoblot analysis of A549 cells infected with VSV and
harvested at the indicated time points. WCL, whole-cell lysates. h, Confocal microscopy of 293T cells transfected with Flag-ZNFX1 and GFP-MAVS, with
or without VSV infection. Scale bar, 10 μm. i, Co-immunoprecipitation and immunoblot analysis of 293T cells transfected with Flag-MAVS and deletion
mutants of HA-ZNFX1 or empty vector are shown. j, qRT–PCR analysis of Ifnb1 and ISGs in A549 cells transfected with ZNFX1 and its truncated versions.
k, Co-immunoprecipitation and immunoblot analysis of 293T cells transfected with HA-ZNFX1 and truncation of MAVS. Data are representative of three
independent experiments with similar results.

Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology 1353


Articles NATURE CELL BIoLoGy

presence of ZNFX1 protein in the mitochondrial fractions isolated could further enhance the viral loads of VSV-eGFP in Rig-I−/− cells
from VSV-infected cells (Fig. 6a, lower panel). Confocal microscopy (Fig. 7i). Similar results were noted in that the viral loads of VSV-
confirmed the co-localisation of overexpressed ZNFX1 protein with eGFP in DKO cells were much higher than those in Rig-I−/− cells
mitochondria and enlarged ZNFX1 specks were observed after viral (Fig. 7j). All these results suggested that ZNFX1 performs its antivi-
infection (Fig. 6b). To further explore the sub-mitochondrial locali- ral roles without depending on RLRs.
sation of ZNFX1, protease protection assays were performed. As
shown in Fig. 6c, when mitochondria were treated with proteinase K Discussion
without Triton X-100, MAVS and most of the ZNFX1 protein were The innate immune system detects invading viruses mainly by rec-
digested, indicating ZNFX1 as an outer mitochondrial membrane ognizing the pathogenic nucleic acid through pattern recognition
protein. In contrast, cytochrome c oxidase subunit IV (COX IV), a receptors. Overwhelming evidence has shown that RLRs, includ-
mitochondria inner membrane protein, was resistant to proteinase ing RIG-I and MDA5, are perhaps the most important lines of viral
K digestion. To further characterize the mitochondria association RNA sensing3–6,41. However, expression of RIG-I and MDA5 is rare
of ZNFX1, mitochondria was then subjected to alkali extraction. in resting cells and becomes detectable until 7 h in viral infection42,43.
Supernatant with peripheral membrane proteins and pellet frac- Thus, although the antiviral roles of RIG-I and MDA5 have been
tions with integral membrane proteins were recovered. well established, how the type I IFN is primed to initiate the early
We found that the outer membrane integral protein MAVS was expression of a series of ISG genes, including RIG-I and MDA5,
primarily recovered in the pellet following extraction, but observed remains unanswered.
COX IV in both fractions. ZNFX1 was largely recovered in the Here, we have demonstrated that ZNFX1, a member of RNA heli-
supernatant (Fig. 6c), indicating that ZNFX1 localises to mitochon- case SF1, is an IFN-stimulated gene and can restrict the replication
dria as a peripheral membrane protein, like SARM1 and ALEX3 in of RNA viruses. Similar to RIG-I and MDA5, ZNFX1 induces a type
previous reports37,38,40. I IFN response by sensing viral RNAs and interacting with MAVS.
To further characterize how ZNFX1 transduces the antiviral However, ZNFX1 differs from RIG-I and MDA5 in several aspects.
signals when sensing viral RNA, we examined the interaction of First, although RIG-I, MDA5 and ZNFX1 are commonly expressed
ZNFX1 with adaptors involved in IFN production, including TRIF, ISGs regulated by the Jak-STAT pathway, the constitutive mRNA or
MyD88, MAVS and STING. Co-immunoprecipitation experi- protein levels of ZNFX1 are much higher than those of RIG-I and
ments showed that HA-tagged ZNFX1 was associated with Flag- MDA5 in various tissues and cell types (Fig. 1i and Extended Data
tagged MAVS but not with TRIF, MyD88 and STING (Fig. 6d). Figs. 1f, 2f and 5a–c). Meanwhile, the induced expression of ZNFX1
Endogenous co-immunoprecipitation experiments further showed is primed and reaches a peak much earlier than RIG-I upon IFN-α
that ZNFX1 and MAVS formed a complex in A549 cells naturally, and IFN-β stimulation (Fig.1i). These differences may indicate the
and their association was enhanced upon viral infection owing to prime importance of ZNFX1 to monitor viral invasion at an early
the upregulation of ZNFX1 (Fig. 6e–g). Confocal microscopy also stage. Second, unlike RIG-I and MDA5, localised in the cytosol with
indicated that ZNFX1 co-localised with MAVS (Fig. 6h). Further cytosol-to-mitochondrial membrane translocation after sensing
co-immunoprecipitation assays using a series of ZNFX1 trunca- viral RNA, ZNFX1 is localised to the outer membrane of mitochon-
tion mutants showed that full-length ZNFX1 and ARM-containing dria, which should facilitate its natural interaction with mitochon-
mutants, including F3 and F5, could interact with MAVS (Fig. 6i). dria-localised MAVS. Although no classical mitochondria targeting
Consistent with co-immunoprecipitation assays, overexpressed motif can be predicted from ZNFX1, functional studies of the ARM
full-length and F3 truncation mutant of ZNFX1 resulted in induced domain containing proteins ALEX3 and SARM suggest that the
expression of Ifnb1 and ISGs and restricted VSV replication (Fig. 6j ARM in ZNFX1 may be responsible for its mitochondrial localisa-
and Extended Data Fig. 4a). Moreover, ZNFX1 could not interact tion37,38,44–46. Third, although ZNFX1 can interact with MAVS, the
with truncated MAVS without the transmembrane (TM) domain ZNFX1–MAVS complex is not as efficient as the RIG-I–MAVS
(Fig. 6k), and the mitochondrial distribution of ZNFX1 was inde- complex in inducing the transcription of Ifnb1 and ISGs. Such a
pendent of its interaction with MAVS (Extended Data Fig. 4b). functional difference may be due to the lack of the caspase recruit-
ment domain (CARD) in ZNFX1. Although ZNFX1 binds to virus
ZNFX1-mediated signalling is independent of RIG-I and MDA5. RNA and interacts with MAVS in a fashion independent of RIG-I
To validate the role for ZNFX1 in generating antiviral immune and MDA5, the deficiency of ZNFX1 could impair the induced
responses relative to existing antiviral sensors RIG-I and MDA5, expression of a series of ISGs, including RIG-I or MDA5, leading
we first performed endogenous co-immunoprecipitation assays and to antiviral defect in the early stage of virus infection47–49. Thus, we
found that ZNFX1 interacts with neither RIG-I nor MDA5 in rest- infer that ZNFX1 is probably an early sensor for RNA viruses rather
ing or VSV-infected cells. qRT–PCR analyses were performed and than a redundant RNA sensor to trigger IFN signalling upon sens-
indicated that Znfx1 deficiency did not affect RIG-I-, MDA5- and ing dsRNA. However, the efficient antiviral responses based on the
MAVS-mediated induction of Ifnb1 expression (Fig. 7a). Moreover, induction of IFNs are still required for the activation of RLRs, whose
overexpressed ZNFX1 could intensely induce the mRNA expression expression could be primed by ZNFX1 in the early antiviral stage.
of Ifnb1 and Ifit1 in wild-type and Mda5−/− cells, slightly in Rig-I−/−, From this point of view, ZNFX1 might also be regarded as a posi-
but not in Mavs−/− cells (Fig. 7b). The qRT–PCR results also showed tive regulator of RIG-I and MDA5 signalling, leading to an efficient
reduced viral loads of VSV and EMCV in Mda5−/− cells when cells antiviral signalling network later on (Extended Data Fig. 7).
were transfected with ZNFX1, RIG-I and MDA5 expressing plas- In addition to the functional differences that have been men-
mids, respectively (Fig. 7c). Likewise, FACS analyses indicated a tioned above, we found that ZNFX1 is highly conserved from fruit
reduced percentage of GFP+ cells in VSV-eGFP infected wild-type, flies to humans (Extended Data Figs. 5d and 6 and Supplementary
Mda5−/− and Rig-I−/− cells, which were transfected with viral sensors, Table 1), which is strikingly different from RIG-I, which is lacking
including ZNFX1 (shown in Fig. 7d,e). By using RNA sequencing, in fruit fly, fugu fish and chicken lineages. In addition to ZNFX1,
we observed that ZNFX1 overexpression enhanced the induction another representative of RNA helicase SF1, MOV10, is well con-
of IFNs and ISGs in Rig-I−/− cells upon poly(I:C) stimulation (Fig. served during evolution and shown to be a co-factor to enhance the
7f). We also observed the VSV and poly(I:C) induction of Ifnb1 and nuclear export of viral mRNA34. Thus, the evolutionarily conserved
Ifit1 in Znfx1 and Rig-I double knockout cells (DKO) when cells RNA helicase SF1 rather than RLRs may play more important anti-
were transfected with ZNFX1 or RIG-I-overexpressed plasmids, viral roles in lower species. The arms race between viruses and the
respectively (Fig. 7g,h). Moreover, siRNA knockdown of ZNFX1 host may finally result in the recruitment of many RNA or DNA

1354 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
a WT Znfx1–/– b WT Rig-I –/– Mda5 –/– Mavs–/– WT Rig-I –/– Mda5 –/– Mavs–/–

Rel. mRNA of Ifnb1


Rel. mRNA of Ifnb1
P = 0.1758

Rel. mRNA of Ifit1


1,500 200 P = 0.0007 P = 0.0004 400 P = 0.0294
P = 0.5914 P = 0.0304 P = 0.0004
1,000 150 300 P = 0.0002
P = 0.3851
500 100 200
P = 0.0734
100 50 P = 0.1935 100 P = 0.6618
0 0 0
EV + + + + EV + + + +
1

S
M 5
M I
ZN V

-
FX

A
AV
IG
E

ZNFX1 + + + + ZNFX1 + + + +

D
R
RIG-I + + + + RIG-I + + + +
MDA5 + + + + MDA5 + + + +

c WT d
EV+ RIG-I+ ZNFX1+
80,000 P = 0.0015 Mda5 –/– 40.43% 7.85% 3.40% e
WT Rig-I –/– Mda5 –/–

WT
Rel. mRNA

60,000 P = 8.41 × 10 –5
of VSV

80 P = 0.0097 P = 1.03 × 10–5


40,000 –6
P = 2.93 × 10

(% of total)
VSV-GFP+
60
20,000
EV+ RIG-I+ ZNFX1+ 40
0
59.94% 30.42% 51.41%

Rig-I –/–
EV + + 20
ZNFX1 + +
0
RIG-I + +
EV + + ++ + +
MDA5 + +
ZNFX1 + + +
VSV VSV
EV+ MDA5+ ZNFX1+ RIG-I + +
MDA5 +

Mda5–/–
62.34% 18.37% 13.54%
P = 0.0030 VSV-eGFP VSV-eGFP VSV-eGFP
SSC-A

60,000
P = 0.0005
Rel. mRNA
of EMCV

40,000
VSV-eGFP
20,000

0 g DKO DKO
h DKO DKO
EV + + P = 0.0001
Rel. mRNA of Ifnb1

P = 0.0030 Rel. mRNA of Ifnb1

Rel. mRNA of Ifit1


Rel. mRNA of Ifit1

ZNFX1 + + 15 P = 0.0003 6 100,000 P = 0.0004 1,000


+ + 10,000
RIG-I 100
10 4 1,000
MDA5 + + 100 10
EMCV EMCV 5 2 10
1
1
0 0 0.1 0.1
EV + + EV + + EV + + EV + +
ZNFX1 + + ZNFX1 + + ZNFX1 + + ZNFX1 + +
RIG-I + + RIG-I + +
VSV VSV
Poly(I:C) Poly(I:C)

f i j
Apobec3

WT
Tnfsf10

WT
Parp14
Trim38
Trim25
Trim21

Psmb9

Myd88
Usp18

Dhx58

Rig-I –/–
Tdrd7

Isg20
Ifitm3
Ifitm1

Gbp4
Gbp3
Gbp2

50 50
Hla-e
Hla-b

Cd38
Tap1

P = 0.0026 Rig-I –/–


Ccl5
Ccl2
Ifi35
Ifi16
Nmi
Tlr3

Ifit3
Ifit2
Irf1

P = 0.0051
40 40
(% of total)

(% of total)
VSV-GFP+

VSV-GFP+

DKO
30 30
EV
20 20
Rig-I –/–

ZNFX1 10 10
EV+ 0 0
si 1

1
si si C rl
FX l

si si C rl
FX l
ZN tr

ZN tr

N-STM VSV-eGFP
t

t
C

ZNFX1+
si

Row z-score N-STM VSV-eGFP


–1.5 0 1.5

Fig. 7 | ZNFX1 induces IFN signalling in an RLR-independent manner. a, qRT–PCR analysis of Ifnb1 in wild-type and Znfx1−/−A549 cells transfected
with the indicated expressing plasmids. b, qRT–PCR analysis of Ifnb1 and Ifit1 in wild-type, Rig-I−/−, Mda5−/− or Mavs−/− A549 cells transfected with the
indicated expressing plasmids. c, qRT–PCR analysis of cellular VSV and EMCV loads in wild-type or Mda5−/− A549 cells transfected with the indicated
expressing plasmids for 24 h followed by viral infection for another 12 h. d, FACS analysis of WT, Rig-I−/− and Mda5−/− A549 cells transfected with ZNFX1,
RIG-I or MDA5 expressing plasmids or EV for 24 h followed by VSV-eGFP infection. The experiments were repeated three times, independently, with
similar results obtained. e, Statistical analysis of the data from the multiple repeated FACS experiments in d. f, Heatmap of ISGs expression profiles in
ZNFX1 overexpression in Rig-I−/− cells with or without poly(I:C) stimulation. g,h, Ifnb1 and Ifit1 mRNA transcripts were analysed by qRT–PCR in Rig-I−/− and
Znfx1−/− DKO cells transfected with the indicated expressing plasmids followed by VSV or poly(I:C) stimulation. i, FACS analysis of WT and Rig-I−/− A549
cells transfected with siRNAs of control or Znfx1 for 48 h followed by VSV-eGFP infection. j, FACS analysis of WT, Rig-I−/− and DKO A549 cells followed by
VSV-eGFP infection. For a–c,e,g–j, n = 3 independent experiments. Data are presented as mean ± s.d. P values were calculated using two-tailed unpaired
Student’s t-test.

binding proteins, such as the DExD/H box helicases, to detect the In all, our study here has identified ZNFX1 as a dsRNA sensor
invading viruses in the evolution of vertebrates. As well as acting as that induces the production of IFNs and ISGs in the early stage of
a dsRNA sensor to trigger antiviral responses, ZNFX1 is required viral infection, providing further insights into the existing complex-
for epigenetic inheritance in Caenorhabditis elegans, based on ity of the antiviral immunity mediated by RLRs and MAVS. ZFNX1,
two recent reports50,51. Thus, determining further roles of ZNFX1 as the monitor to sense viral RNA in the early stage of viral infec-
in other physiological or pathological processes still requires tion, may serve as a strategy for early therapeutic intervention in
further studies. viral infections in the future.

Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology 1355


Articles NATURE CELL BIoLoGy

Online content 24. Schoggins, J. W. et al. A diverse range of gene products are effectors of the
Any methods, additional references, Nature Research reporting type I interferon antiviral response. Nature 472, 481–485 (2011).
25. Pillai, P. S. et al. Mx1 reveals innate pathways to antiviral resistance and lethal
summaries, source data, extended data, supplementary informa- influenza disease. Science 352, 463–466 (2016).
tion, acknowledgements, peer review information; details of author 26. Bailey, C. C. et al. IFITM-family proteins: the cell’s first line of antiviral
contributions and competing interests; and statements of code defense. Annu. Rev. Virol. 1, 261–283 (2014).
and data availability are available at https://doi.org/10.1038/s41556- 27. Munir, M. & Berg, M. The multiple faces of proteinkinase R in antiviral
defense. Virulence 4, 85–89 (2013).
019-0416-0.
28. Neil, S. J., Zeng, T. & Bieniasz, P. D. Tetherin inhibits retrovirus release and is
antagonized by HIV-1 Vpu. Nature 451, 425–430 (2008).
Received: 16 October 2018; Accepted: 26 September 2019; 29. Yan, N. & Chen, Z. J. Intrinsic antiviral immunity. Nat. Immunol. 13,
Published online: 4 November 2019 214–222 (2012).
30. Yoh, S. M. et al. PQBP1 is a proximal sensor of the cGAS-dependent innate
References response to HIV-1. Cell 161, 1293–1305 (2015).
1. Ablasser, A. et al. cGAS produces a 2′–5′-linked cyclic dinucleotide second 31. Song, G. et al. E3 ubiquitin ligase RNF128 promotes innate antiviral
messenger that activates STING. Nature 498, 380–384 (2013). immunity through K63-linked ubiquitination of TBK1. Nat. Immunol. 17,
2. Wu, J. et al. Cyclic GMP-AMP is an endogenous second messenger in innate 1342–1351 (2016).
immune signaling by cytosolic DNA. Science 339, 826–830 (2013). 32. Zhu, J. et al. Antiviral activity of human OASL protein is mediated by
3. Yoneyama, M. et al. The RNA helicase RIG-I has an essential function in enhancing signaling of the RIG-I RNA sensor. Immunity 40, 936–948 (2014).
double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 5, 33. Jia, X. et al. The role of alternative polyadenylation in the antiviral innate
730–737 (2004). immune response. Nat. Commun. 8, 14605 (2017).
4. Pichlmair, A. et al. RIG-I-mediated antiviral responses to single-stranded 34. Goodier, J. L., Cheung, L. E. & Kazazian, H. H. Jr. MOV10 RNA
RNA bearing 5′-phosphates. Science 314, 997–1001 (2006). helicase is a potent inhibitor of retrotransposition in cells. PLoS Genet. 8,
5. Kato, H. et al. Differential roles of MDA5 and RIG-I helicases in the e1002941 (2012).
recognition of RNA viruses. Nature 441, 101–105 (2006). 35. Serquina, A. K. et al. UPF1 is crucial for the infectivity of human
6. Goubau, D. et al. Antiviral immunity via RIG-I-mediated recognition of RNA immunodeficiency virus type 1 progeny virions. J. Virol. 87,
bearing 5′-diphosphates. Nature 514, 372–375 (2014). 8853–8861 (2013).
7. Ahmad, S. & Hur, S. Helicases in antiviral immunity: dual properties as 36. Miller, M. S. et al. Senataxin suppresses the antiviral transcriptional response
sensors and effectors. Trends Biochem. Sci. 40, 576–585 (2015). and controls viral biogenesis. Nat. Immunol. 16, 485–494 (2015).
8. Oshiumi, H. et al. DDX60 is involved in RIG-I-dependent and independent 37. López, D. G. et al. The Eutherian Armcx genes regulate mitochondrial
antiviral responses, and its function is attenuated by virus-induced EGFR trafficking in neurons and interact with Miro and Trak2. Nat. Commun. 3,
activation. Cell Rep. 11, 1193–1207 (2015). 814 (2012).
9. Zhang, Z. et al. DDX1, DDX21 and DHX36 helicases form a complex with 38. Mukherjee, P. et al. Activation of the innate signaling molecule MAVS by
the adaptor molecule TRIF to sense dsRNA in dendritic cells. Immunity 34, bunyavirus infection upregulates the adaptor protein SARM1, leading to
866–878 (2011). neuronal death. Immunity 38, 705–716 (2013).
10. Zhang, Z., Yuan, B., Lu, N., Facchinetti, V. & Liu, Y. J. DHX9 pairs with IPS-1 39. West, A. P., Shadel, G. S. & Ghosh, S. Mitochondria in innate immune
to sense double-stranded RNA in myeloid dendritic cells. J. Immunol. 187, responses. Nat. Rev. Immunol. 11, 389–402 (2011).
4501–4508 (2011). 40. Yoshizumi, T. et al. Influenza A virus protein PB1-F2 translocates into
11. Wang, P. H. et al. Nlrp6 regulates intestinal antiviral innate immunity. Science mitochondria via Tom40 channels and impairs innate immunity. Nat.
350, 826–830 (2015). Commun. 5, 4713 (2014).
12. Singleton, M. R., Dillingham, M. S. & Wigley, D. B. Structure and 41. Loo, Y. M. et al. Distinct RIG-I and MDA5 signaling by RNA viruses in
mechanism of helicases and nucleic acid translocases. Annu. Rev. Biochem. innate immunity. J. Virol. 82, 335–345 (2008).
76, 23–50 (2007). 42. Lu, H. B. et al. DHX15 senses double-stranded RNA in myeloid dendritic
13. Fairman-Williams, M. E., Guenther, U. P. & Jankowsky, E. SF1 and SF2 cells. J. Immunol. 193, 1364–1372 (2014).
helicases: family matters. Curr. Opin. Struct. Biol. 20, 313–324 (2010). 43. Lian, H. et al. The zinc-finger protein ZCCHC3 binds RNA and facilitates
14. Linder, P. & Jankowsky, E. From unwinding to clamping—the DEAD box viral RNA sensing and activation of the RIG-I-like receptors. Immunity
RNA helicase family. Nat. Rev. Mol. Cell. Bio. 12, 505–516 (2011). 18, 49 (2018).
15. Seth, R. B., Sun, L., Ea, C. K. & Chen, Z. J. Identification and characterization 44. Serrat, R. et al. The non-canonical Wnt/PKC pathway regulates mitochondrial
of MAVS, a mitochondrial antiviral signaling protein that activates NF-κB dynamics through degradation of the arm-like domain-containing protein
and IRF 3. Cell 122, 669–682 (2005). Alex3. PLoS One 8, e67773 (2013).
16. Ishikawa, H. & Barber, G. N. STING is an endoplasmic reticulum adaptor 45. Panneerselvam, P. et al. T-cell death following immune activation is mediated
that facilitates innate immune signalling. Nature 455, 674–678 (2008). by mitochondria-localized SARM. Cell Death Differ. 20, 478–489 (2013).
17. Chen, Q., Sun, L. J. & Chen, Z. J. Regulation and function of the 46. Yuan, S. et al. Amphioxus SARM involved in neural development
cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 17, may function as a suppressor of TLR signaling. J. Immunol. 184,
1142–1149 (2016). 6874–6881 (2010).
18. Munoz-Wolf, N. & Lavelle, E. C. Hippo interferes with antiviral defences. 47. Hou, F. et al. MAVS forms functional prion-like aggregates to activate and
Nat. Cell Biol. 19, 267–269 (2017). propagate antiviral innate immune response. Cell 146, 448–461 (2011).
19. Zhang, Q. et al. Hippo signalling governs cytosolic nucleic acid sensing 48. Gack, M. U. et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for
through YAP/TAZ-mediated TBK1 blockade. Nat. Cell Biol. 19, RIG-I-mediated antiviral activity. Nature 446, 916–920 (2007).
362–374 (2017). 49. Jiang, X. et al. Ubiquitin-induced oligomerization of the RNA sensors RIG-I
20. O’Shea, J. J. & Plenge, R. JAK and STAT signaling molecules in and MDA5 activates antiviral innate immune response. Immunity 36,
immunoregulation and immune-mediated disease. Immunity 36, 959–973 (2012).
542–550 (2012). 50. Ishidate, T. et al. ZNFX-1 functions within perinuclear nuage to balance
21. Schoggins, J. W. et al. Pan-viral specificity of IFN-induced genes reveals new epigenetic signals. Mol. Cell 70, 639–649 (2018).
roles for cGAS in innate immunity. Nature 505, 691–695 (2014). 51. Wan, G. et al. Spatiotemporal regulation of liquid-like condensates in
22. Chan, Y. K. & Gack, M. U. Viral evasion of intracellular DNA and RNA epigenetic inheritance. Nature 557, 679–683 (2018).
sensing. Nat. Rev. Microbiol. 14, 360–373 (2016).
23. Schneider, W. M., Chevillotte, M. D. & Rice, C. M. Interferon-stimulated Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
genes: a complex web of host defenses. Annu. Rev. Immunol. 32, published maps and institutional affiliations.
513–545 (2014). © The Author(s), under exclusive licence to Springer Nature Limited 2019

1356 Nature Cell Biology | VOL 21 | NOVEMBER 2019 | 1346–1356 | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles
Methods were purified using poly(T) oligo-attached magnetic beads. Purified libraries
Mice. C57BL/6 Znfx1−/− mice were generated by a CRISPR/Cas9 system from were quantified using a Qubit 2.0 fluorometer (Life Technologies) and validated
GemParmatech Co. Exons 3 to 11 were selected as the targeted region. Mice were using an Agilent 2100 Bioanalyzer to confirm the insert size and to calculate the
kept and bred in pathogen-free conditions. The Institutional Animal Care and mole concentration. Library preparations were sequenced on an Illumina Hiseq
Use Committee of Sun Yat-sen University, Guangzhou, China approved all the Xten platform and paired-end reads were generated. Library construction and
experimental protocols concerning the handling of mice. The study is compliant sequencing were performed at Biomarker Corporation.
with all relevant ethical regulations regarding animals.
Ultraviolet crosslinked RIP assay. 293T cells were transfected with the indicated
Reagents and antibodies. The poly(I:C) (both HMW and LMW) and poly(dA:dT) Flag-tagged plasmids for 24 h, followed by VSV infection for another 12 h. Cells
were purchased from InvivoGen. Recombinant human IFN-α, recombinant human were crosslinked with ultraviolet light at 254 nm (300 mJ cm−2), then harvested and
IFN-β and all IFN ELISA kits were purchased from R&D Systems. Lipofectamine lysed with immunoprecipitation lysis buffer. Flag M2 antibody (Sigma-Aldrich)
2000, Lipofectamine 3000 and Lipofectamine RNAiMAX were purchased from was added to the cell lysate and incubated overnight at 4 °C. Protein G beads were
Invitrogen. Protein G agarose, used for immunoprecipitation, was purchased from washed and added to antibody–cell lysate mixture and incubated for 2 h at 4 °C.
Pierce. Poly(I) and agarose-bond poly(C) were from Sigma (P4154 and P9827). The bead-bound immunoprecipitates were washed three times with lysis buffer
The following antibodies were used for immunoblot analysis: anti-human ZNFX1 containing RNase inhibitors. The beads were then split into two samples for protein
(1:1,000; ab179452, Abcam), anti-mouse ZNFX1 (1:1,000; Dia-An Biotech), anti- or RNA extraction. Proteins were extracted for western blot analysis and the RNA
MAVS (1:1,000; ab31334, Abcam), anti-IRF3 (1:1,000; sc-33641, Santa Cruz), anti- was extracted using Trizol reagent before real-time PCR analysis for VSV RNA. For
RIG-I (1:1000; D14G6, Cell Signaling), anti-phosphorylated IRF3 (1:1,000; 4D4G, endogenous ZNFX1-RIP analysis, ZNFX1-associated immunoprecipitates were
Cell Signaling), anti-TBK1 (1:1,000; D1B4, Cell Signaling), anti-phosphorylated prepared by incubating IgG or anti-ZNFX1 (2 μg) antibody and protein G beads
TBK1 (1:1,000; D52C2, Cell Signaling), anti-GAPDH (1:10,000; ET1601-4, with cell lysates of VSV-, EMCV- or SeV-infected A549 cells. RNA was extracted
HuaBio), anti-β-actin (1:10,000; 60008-1-Ig, Proteintech), anti-lamin B1 (1:2,000; using Trizol reagent before real-time PCR analysis for viral RNA.
12987-1-AP, Proteintech), anti-COX-IV (1:3,000; 66110-1-Ig, Proteintech), anti-
HA (1:5,000; clone HA-7, H6533, Sigma), anti-Flag (1:5,000; clone M2, F1804, Confocal microscopy. A549 cells were transfected with the expression plasmid
Sigma), goat anti-mouse IgG-HRP (1:10,000; HA1006, HuaBio), goat anti-rabbit for Flag-tagged ZNFX1. After 24 h, cells were mock or VSV-infected for 8 h and
IgG-HRP (1:10,000; HA1001, HuaBio). 16 h. The cells were fixed in 4% paraformaldehyde and permeabilized with 0.1%
saponin, then blocked for 30 min with 10% goat serum, incubated overnight at 4 °C
Cells and pathogens. The A549, MEF, L929 and 293T cell lines were purchased with anti-Flag and mitochondrial marker MitoTracker Red CMXRos (Invitrogen).
from ATCC. All these cells were cultured in endotoxin-free DMEM (Gibco), Imaging of the cells was carried out using Leica TCS SP5 confocal laser microscopy
supplemented with 10% FBS (Gibco) and 1% penicillin–streptomycin (Gibco). under a ×100 oil objective.
PBMC and mouse peritoneal macrophage were isolated and cultured as described
previously33. VSV (Indiana strain), VSV-eGFP and HSV-1 were from our Flow cytometry. For gene knockdown, A549 and L929 cells were transiently
laboratory. Human influenza virus A/Puerto Rico/8/34 (H1N1) (PR8) and SeV transfected with siRNAs for 48 h and then challenged with VSV-eGFP for 6 h
were provided by J. Cui (College of Life Sciences, Sun Yat-sen University). H1N1, at 2 MOI. For gene overexpression, cells were transfected with the indicated
SeV, VSV and HSV-1 were titred on Vero cells. expression plasmids for 24 h following VSV-eGFP infection at the indicated MOI
Analogue stimulation and virus infection. Cells were stimulated with poly(I:C) for 6 h or 12 h. The cells were washed once in cold PBS and resuspended in cold
(3 μg ml−1) and poly(dA:dT) (3 μg ml−1) delivered by polyethylenimine (PEI) for the staining buffer (PBS with 1% fetal bovine serum). For each condition, 10,000
indicated hours. Cells were infected at an indicated MOI in serum-free DMEM for cells were counted and the followed analyses were performed on a flow cytometer
1 h, washed by 1× PBS, and incubated with fresh complete medium for the times (FACSCalibur and CytoFLEX).
shown in the figures. For in vivo virus infection, six- to eight-week-old mice were
infected with VSV for 24 h by intravenous injection (2 × 107 p.f.u. per g body weight). Mitochondrial fractionation and proteolysis. Mitochondrial fractionation
and proteolysis assays were performed as previously described, with slight
RNA interference. Lipofectamine RNAi MAX was used for the transfection modification41. Briefly, virally infected A549 cells were washed once with cold 1×
of siRNAs (30 nM) into A549 or L929 cells according to the manufacturer’s PBS (pH 7.2), scraped off the culture plate, and lysed in 800 μl of homogenization
instructions. The sequences of siRNAs with higher silencing efficiency are as follows: buffer (20 mM HEPES (pH 7.5), 70 mM sucrose and 220 mM mannitol) by 30
human Znfx1-siRNA: 5′-GGAAUAUCGUGGAGUGUAA dTdT-3′; strokes in a Dounce homogenizer. The homogenate was centrifuged at 800g
human Mavs-siRNA: 5′-CCACCUUGAUGCCUGUGAA dTdT-3′. for 5 min to precipitate the nuclei, and the resulting supernatant was further
These human RNA oligos were synthesized at Guangzhou Ribobio. The mouse centrifuged at 10,000g for 10 min (4 °C) to precipitate the crude mitochondrial
siRNAs were purchased from Thermo Scientific. The target for Znfx1 is ID:509273 fraction. The resulting supernatant was further centrifuged at 100,000g for 30 min
and for Mavs is ID:169532. (4 °C) to precipitate light membrane organelles, and the final supernatant was used
as the cytosolic fraction.
RNA isolation, qRT–PCR and ELISA. Total RNA was extracted using TRIzol For the proteinase K resistance assay, the isolated mitochondria pellet was
reagent (Invitrogen) and reversed-transcribed with a PrimeScript RT reagent washed once with homogenization buffer. Then, the pellet was resuspended in
kit with gDNA Eraser (TaKaRa) according to the manufacturer’s instructions. A buffer and treated with proteinase K (100 ng ml−1) in the absence or presence
Light Cycler480 instrument (using the 384-well module) with RealStar Green Fast of 1% Triton X-100 (wt/vol). The proteolytic reaction was performed on ice for
Mixture (GenStar) was used for quantitative real-time PCR analysis. The data were 15 min, and the reactants were subjected to western blot analysis with the indicated
normalized by the level of β-actin expression in each individual sample. The 2−ΔΔCt antibodies.
method was used to calculate relative expression changes. The primer sequences
are listed in Supplementary Table 2. The concentrations of IFN-α and IFN-β in Generation of Znfx1−/−, Rig-I−/−, Mda5−/−, Mavs−/− and Ifnar1−/− cells.
culture supernatants were measured using kits from R&D Systems, according to Znfx1−/−, Rig-I−/−, Mda5−/−, Mavs−/− and Ifnar1−/− cells were constructed using
the manufacturer’s instructions. the CRISPR/Cas9 gene-editing system. Small guide RNAs (sgRNAs) targeting
the genome sequence of target genes were cloned into PX458 vector. The sgRNA
Luciferase reporter gene assay. A549 cells or 293T cells were plated at a density targeting sequences were 5′-TCCTAACCAACGAGTCTGTT-3′ for ZNFX1,
of 105 cells per well in a 48-well plate. A549 cells in each well were transiently 5′-GCAGGTGCAGAGAAATTGG-3′ for RIG-I, 5′-GGGTGAAAATGTACA
transfected with 100 ng IFN-β or ISRE luciferase and 5 ng Renilla luciferase TCCAG-3′ for MDA5, 5′-CCTCTGACCTGGCAGCCCT-3′ for MAVS and
reporter vectors plus ZNFX1 expression vector using Lipofectamine 3000. 293T 5′-TCATTTACACCATTTCGCAA-3′ for IFNAR1. CRISPR plasmids were
cells including wild-type 293T and Znfx1 deficiency 293T cells (Znfx1−/−-293T) transfected into 293T or A549 cells using Lip2000 and Lip3000, and single-cell
were transfected with reporter vectors using Lipofectamine 2000. At 24 h post colonies of GFP+ cells were picked and validated at protein expression level by
transfection, cells were stimulated with 3 μg ml−1 poly(I:C) or poly(dA:dT) or western blot.
infected with the indicated dose of VSV. Cells were harvested 24 h after stimulation
or infection, and luciferase activities were measured with a dual-luciferase reporter In vitro pulldown and competition. For poly(C) and poly(I:C) pulldown assays,
assay system (Promega) according to the manufacturer’s instructions. Data are poly(C)- or poly(I:C)-coated beads were equilibrated in binding buffer and
normalized for transfection efficiency by dividing Firefly luciferase activity by were combined with an equal volume of whole-cell lysates prepared from 293T
Renilla luciferase activity. Each experiment was replicated at least three times. cells transiently transfected with the indicated expression plasmids that were
treated with lysis buffer. The cell extracts were supplemented with protease and
Library construction for RNA sequencing and sequencing procedures. Total phosphatase inhibitors. The mixtures were incubated with gentle agitation for
RNA was isolated using an RNeasy mini kit (Qiagen). RNA integrity was assessed 2 h at 4 °C. Beads were then washed three times with lysis buffer and analysed by
using an RNA Nano 6000 assay kit of the Agilent Bioanalyzer 2100 system. immunoblotting with anti-HA-HRP.
Sequencing libraries were generated using an NEBNext UltraTM directional RNA To generate purified ZNFX1 proteins, 293T cells transiently transfected
library prep kit for Illumina (NEB). The poly(A) containing mRNA molecules with plasmids encoding Flag-ZNFX1 proteins were lysed with lysis buffer. After

Nature Cell Biology | www.nature.com/naturecellbiology


Articles NATURE CELL BIoLoGy
centrifugation, the supernatants were incubated with anti-Flag M2 affinity gel Acknowledgements
for 2 h at 4 °C. The beads were washed three times with wash buffer, and the This work was supported by the National Natural Science Foundation (NNSF) of China
Flag-ZNFX1 proteins were eluted by competition with Flag peptide. For in vitro under grants 31770943, 81430099 and 31900661 and by the Natural Science Foundation
pulldown and competition, purified ZNFX1 proteins were incubated for 4 h with of Guangdong Province of China under grants 2015A030306043, 2018A030313924 and
agarose bead-conjugated poly(I:C) in the absence or presence of poly(I:C) and 2018A030313051.
other competitors. Centrifugation was followed by the incubation and the beads
were analysed by immunoblotting with anti-HA-HRP.
Author contributions
Statistics and reproducibility. Statistical analyses were carried out using Y.W., S.Y. and A.X. conceived the ideas and designed the experiments. Y.W., X.J., Y.G.,
Microsoft Excel software and GraphPad Prism to assess the differences between T.L., M.N. and X.L. performed the experiments. Y.W., S.Y. and X.J. analysed the data. S.Y.,
experimental groups. Statistical significance was determined using a two-tailed, Y.W., X.J. and S.C. contributed to editing the manuscript. S.Y. and A.X. supervised the
unpaired Student’s t-test with a confidence interval of 95%. P ≤ 0.05 was denoted research and wrote the paper. S.Y. and X.J. are joint co first authors.
as statistically significant. All experiments were performed three or more times
independently under identical or similar conditions.
Competing interests
Reporting Summary. Further information on research design is available in the The authors declare no competing interests.
Nature Research Reporting Summary linked to this article.
Additional information
Data availability Extended data is available for this paper at https://doi.org/10.1038/s41556-019-0416-0.
RNA sequencing data that support the findings of this study have been deposited Supplementary information is available for this paper at https://doi.org/10.1038/
in the Gene Expression Omnibus (GEO) under accession code GSE132979. Source s41556-019-0416-0.
data for Figs. 1–7 and Extended Data Figs. 1–7 have been provided as Statistics
Correspondence and requests for materials should be addressed to A.X.
Source Data. All other data supporting the findings of this study are available from
the corresponding author upon reasonable request. Reprints and permissions information is available at www.nature.com/reprints.

Nature Cell Biology | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles

Extended Data Fig. 1 | See next page for caption.

Nature Cell Biology | www.nature.com/naturecellbiology


Articles NATURE CELL BIoLoGy

Extended Data Fig. 1 | Bioinformatics analysis of in vitro transcription-sequencing APA sites (IVT-SAPAS) data of VSV-infected macrophages and
previously collected viral infection microarray data. a, IVT-SAPAS revealed the genes with transcriptional changes in MDMs infected with VSV for 0,
2, 4, 8, 16, 24 hrs. b-c, The percentage of GFP+ cells of FACS analysis of A549 cells transfected with control siRNA or the indicated siRNAs followed by
VSV-eGFP infection for another 6 hrs (n = 3 independent experiments). d, qRT-PCR revealed the mRNA expression of target gene in A549 cells transfected
with indicated siRNAs for 48 hrs (n = 3 independent experiments). e-f, RNA levels of Znfx1 and Rig-I are significantly increased after different virial
infection in different cell types. g, Schematic representation of ZNFX1 promoter containing core region bound by STAT1, STAT2, IRF1 and IRF9. For e, n = 3
independent experiments. Data in f, n = 4 wells for SeV infected epithelial cells and n = 6 wells for uninfected cells; n = 5 samples for IVA infected pDCs;
n = 4 independent experiments for SeV infected monocytoid cells; n = 2 independent experiments for SeV or HIV infected Macrophages or mDCs. All data
are shown as the mean ± s.d. Statistical differences were detected using two-tailed unpaired Student’s t-tests.

Nature Cell Biology | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles

Extended Data Fig. 2 | ZNFX1 deficiency impairs antiviral immune response in vitro and in vivo. a, Quantitative RT-PCR (qRT-PCR) analysis of Znfx1
mRNA expression in A549 and L929 cells transfected with control siRNA or ZNFX1 siRNA 1#, 2# or 3# for 48 hrs (n = 3 independent experiments).
b, Western blot analysis of ZNFX1 protein expression in A549 cells transfected with control siRNA or human ZNFX1 siRNA 1# for 48 hrs. c, ELISA of IFN-α
or IFN-β production in cell supernatants from A549 cells with target gene knockdown for 48 hrs followed by VSV infection or poly I:C stimulation for
another 12 hrs (n = 4 independent experiments). d, qRT-PCR analysis of VSV mRNA expression (left panel) and plaque assay analysis of VSV titer (right
panel) of A549 cells transfected with RIG-I, ZNFX1 expressing plasmids or empty vector plasmid for 24 hrs and then infected with VSV at an MOI of 2 for
16 hrs (n = 3 independent experiments). e, FACS analysis of Znfx1+/+ and Znfx1-/- 293T cells followed by VSV-eGFP infection at 0.5 MOI for the indicated
time points (n = 3 independent experiments). f, Znfx1-/- 293T and A549 clones were generated by the CRISPR-Cas9 method. Deficiency of target genes in
the KO clones were confirmed by immunoblotting analysis. g, qRT-PCR analysis of viral mRNA transcripts in VSV, EMCV, H1N1 and HSV-1 infected A549
cells with control siRNA (si Control) or Znfx1-specific siRNA (si ZNFX1) (n = 3 independent experiments). h, ELISA of IFN-α in supernatants of BMDMs
from WT and Znfx1-/- mice infected with VSV or HSV-1 for 16 hrs (n = 5 independent experiments). All data are shown as the mean ± s.d. P values were
calculated using two-tailed unpaired Student’s t-test. For b, f, the experiments were repeated three times, independently, with similar results obtained.

Nature Cell Biology | www.nature.com/naturecellbiology


Articles NATURE CELL BIoLoGy

Extended Data Fig. 3 | ZNFX1 positively regulates IFN-β signaling. a, Illustration of the CRISPR-Cas9 strategy to generate Znfx1-deficient mice and primer
design used in (b). b, Genotyping of the ZNFX1 mutant pups. c, Immunoblot analysis of ZNFX1 protein levels in Znfx1+/+ and Znfx1-/- MEFs cells. d, qRT-
PCR analysis of Ifnb1 and ISGs mRNA levels in A549 cells transfected with siControl (si Ctrl) or si ZNFX1 for 48 hrs and then infected with VSV for the
indicated time points (n = 3 independent experiments). e, qRT-PCR analysis of Ifnb1 and ISGs mRNA expression in Znfx1+/+ and Znfx1-/- 293T cells followed
by VSV infected with increasing MOI (0.5 and 1) for 0, 8 and 16 hrs (n = 3 independent experiments). For b, c, the experiments were repeated three times,
independently, with similar results obtained. Data in d, e are the mean ± s.d. P values were calculated using two-tailed unpaired Student’s t-test.

Nature Cell Biology | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles

Extended Data Fig. 4 | ZNFX1 localizes to mitochondria and interacts with MAVS. a, FACS analysis of Znfx1-/- A549 cells transfected with ZNFX1 and
its mutants expressing plasmids or empty vector (EV) for 24 hrs followed by VSV-eGFP infection at an MOI of 2 for 6 hrs. b, Endogenous level of ZNFX1
protein in mitochondrial fractions in WT and Mavs-/- 293T with or without VSV infection at 1 MOI for 6 hrs. COX-IV was used as the loading control. Data
are representative of at least three independent experiments.

Nature Cell Biology | www.nature.com/naturecellbiology


Articles NATURE CELL BIoLoGy

Extended Data Fig. 5 | The expression of Znfx1 in different tissues and cell types, and the phylogenetic tree of Znfx1. a-c, The expression of Znfx1, Rig-I
and Mda5 in different tissues and cell types as per BioGPS. d, Phylogenetic tree of ZNFX1 and RIG-I using an amino acid sequence alignment among
different species.

Nature Cell Biology | www.nature.com/naturecellbiology


NATURE CELL BIoLoGy Articles

Extended Data Fig. 6 | Alignment of ZNFX1 amino acid sequences in human, mouse, rat and zebrafish. Shading indicates sequence conservation, with
darker gray indicating a higher degree of conservation.

Nature Cell Biology | www.nature.com/naturecellbiology


Articles NATURE CELL BIoLoGy

Extended Data Fig. 7 | Work model of mitochondria-localized ZNFX1 functions as a dsRNA sensor to initiate antiviral responses through MAVS. Upon
RNA virus infection, ZNFX1 induces type I interferon response by interacting with MAVS in the early stage, thus primes the expression of a number of
ISGs, including RIG-I and MDA5. The induced sensors further enhance the antiviral immune response by amplifying ISGs expression.

Nature Cell Biology | www.nature.com/naturecellbiology


nature research | reporting summary
Corresponding author(s): Anlong Xu
Last updated by author(s): 28th August 19

Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see Authors & Referees and the Editorial Policy Checklist.

Statistics
For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section.
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient)
AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Our web collection on statistics for biologists contains articles on many of the points above.

Software and code


Policy information about availability of computer code
Data collection Fluorescent images: Leica TCS SP5 confocal laser microscopy
Flow cytometry: Cytoflex (Beckmam Coulter).
qRT-PCR: LightCycler 480 instrument (Roche).

Data analysis Data representation and statistical analysis: Graphpad Prism 7 and Microsoft Office Excel 2017. Flow cytometry data were analyzed with
CytExpert 1.2 software. Laser scanning confocal microscopy: LAS X Core. Images were processed with Adobe Photoshop.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors/reviewers.
We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.

Data
Policy information about availability of data
All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability
October 2018

Accession codes of GEO database for collected microarray data: GDS4231/4214/1271/3210/6082/6063.


Accession codes of GEO database for raw data of RNA sequencing : GSE132979.
Statistical source data for Fig. 1-7 and Supplementary Fig. 1-6 are provided in Supplementary Table. Uncropped blots are provided in supplement figure.

1
nature research | reporting summary
Field-specific reporting
Please select the one below that is the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/documents/nr-reporting-summary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size Sample sizes were determined based on the Authors' experience of what is necessary to generate a convincing and compelling result. All
experiments included at least 3 independent experiments.The number of independent experiment was indicated in each figure legend.
Western blots are representative of three independently performed experiments.

Data exclusions No data were excluded from the analyses.

Replication All experimental findings were reliably reproduced in multiple independent experiments as indicated in the figure legends.

Randomization Animals were randomly assigned to the respective body weight matched groups.

Blinding Animals were randomly assigned to the respective body WT groups and KO ZNFX1 groups were infected with or without VSV. No blinding was
done in this study. Virtually all the data are quantitative, most measurements are made using a machine, and not easily subject to operator
bias.

Reporting for specific materials, systems and methods


We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material,
system or method listed is relevant to your study. If you are not sure if a list item applies to your research, read the appropriate section before selecting a response.

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Antibodies ChIP-seq
Eukaryotic cell lines Flow cytometry
Palaeontology MRI-based neuroimaging
Animals and other organisms
Human research participants
Clinical data

Antibodies
Antibodies used anti-ZNFX1 (IB:1:1000; ab179452, Abcam),
anti-MAVS (IB:1:1,000; IP:1:100, ab31334, Abcam),
anti-IRF3 (IB:1:1,000, sc-33641, SL-12, Santa Cruz),
anti-RIG-I (IB:1:1000, D14G6, #3743, Cell Signaling),
anti-MDA5 (IB:1:1000, D74E4, #5321, Cell Signaling)
anti-phosphorylated IRF3 (IB:1:1,000; 4D4G; #4947, Cell Signaling),
anti-TBK1 (IB:1:1000; D1B4; #3504, Cell Signaling),
anti-phosphorylated TBK1 (IB:1:1000; D52C2, #5483, Cell Signaling),
anti-GAPDH (IB:1:10,000; ET1601-4; HuaBio),
anti-b-actin (IB:1:10,000; 60008-1-Ig; Proteintech),
anti-Lamin B1 (IB:1:2000; 12987-1-AP; Proteintech),
anti-COX-IV (IB:1:3000; 66110-1-Ig; Proteintech),
anti-HA (IB:1: 5,000; clone HA-7, H6533; Sigma),
October 2018

anti-Flag (IB:1:5000; clone M2, F1804, Sigma),


Goat anti-mouse IgG-HRP (IB:1:10000; HA1006, HuaBio),
Goat anti-Rabbit IgG-HRP (IB:1:10000; HA1001, HuaBio),
anti-mouse ZNFX1 protein was prepared by Dia-An Biotech, Inc, in Wuhan, China (IP:1:100; IB:1:1000).

Validation All antibodies used in this study were obtained from commercial sources and validated according to manufacturers's instruction.

2
Eukaryotic cell lines

nature research | reporting summary


Policy information about cell lines
Cell line source(s) The A549, MEF, L929 and 293T cell lines were purchased from ATCC.

Authentication All the cell lines were authenticated by short tandem repeat profiling prior to use. And microscopic inspection (these cells are
easily distinguished based on morphology).

Mycoplasma contamination All the cell lines were tested negative for mycoplasma contamination.

Commonly misidentified lines No commonly misidentified cell lines were used.


(See ICLAC register)

Animals and other organisms


Policy information about studies involving animals; ARRIVE guidelines recommended for reporting animal research
Laboratory animals The following inbred mouse species were used: C57BL/6J. Supplier and age (6-8 weeks) of the mice as well as housing conditions
are provided for each experiment in the Methods section. Male and female mice were used.

Wild animals We declare the study did not involve wild animals.

Field-collected samples No field-collected samples were used in this study.

Ethics oversight The study is compliant with all relevant ethical regulations for animal experiments. All the experimental protocols were approved
by the Institutional Animal Care and Use Committee of Sun Yat-sen University.
Note that full information on the approval of the study protocol must also be provided in the manuscript.

Flow Cytometry
Plots
Confirm that:
The axis labels state the marker and fluorochrome used (e.g. CD4-FITC).
The axis scales are clearly visible. Include numbers along axes only for bottom left plot of group (a 'group' is an analysis of identical markers).
All plots are contour plots with outliers or pseudocolor plots.
A numerical value for number of cells or percentage (with statistics) is provided.

Methodology
Sample preparation For gene knockdown, knockout or overexpression, A549 and L929 cells were transiently transfected with siRNAs or expression
plasmids and then challenged with VSV-eGFP. The cells were washed once in cold PBS and resuspended in cold staining buffer
(PBS with 1% fetal bovine serum).

Instrument Cytoflex (Beckmam Coulter) system.

Software CytExpert 1.2 software was used for data collection and CytExpert 1.2 for data analysis.

Cell population abundance Purity of the sorted A549 and L929 cell fractions was confirmed by flow cytometry resulting in a purity of the sorted cells of
>95%.

Gating strategy A representative plot and gating is provided with every flow cytometry figure. We gated the GFP positive cells in the VSV-
uinfected group less than 1% and calculated the ratios of GFP positive cells after eGFP-VSV infection.

Tick this box to confirm that a figure exemplifying the gating strategy is provided in the Supplementary Information.
October 2018

You might also like