You are on page 1of 13

42 Hongrui Li et al. DOI: 10.1002/eji.201847603 Eur. J. Immunol. 2019.

49: 42–53
Basic

Innate immunity

Research Article
USP14 promotes K63-linked RIG-I deubiquitination
and suppresses antiviral immune responses
Hongrui Li1 , Zizhao Zhao1 , Jing Ling1 , Linhui Pan1 , Xibao Zhao1,2 ,
Huihui Zhu1 , Juan Yu1 , Bin Xie1 , Jianzhong Shen3 and Weilin Chen1,2

1
Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
2
Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong
Sheng, China
3
Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn
University, Auburn, AL

Retinoic acid-inducible gene I (RIG-I) is a critical RNA virus sensor that initiates antiviral
immune response through K63-linked ubiquitination. In this study, we demonstrated
USP14, a deubiquitinating enzyme, as a negative regulator in antiviral responses by
directly deubiquitinating K63-linked RIG-I. USP14 knockdown significantly enhanced RIG-
I-triggered type I IFN signaling and inhibited vesicular stomatitis virus (VSV) replication
both in mouse peritoneal macrophages and THP1 cells. USP14 overexpression in HeLa
cells attenuated RIG-I-triggered IFN-β expression and promoted VSV replication. Besides,
USP14-specific inhibitor, IU1, increased RIG-I-mediated type I IFN production and antivi-
ral responses in vitro and in vivo. In addition, USP14 could interact with RIG-I and remove
RIG-I K63-linked polyubiquitination chains. This article is the first to report that USP14
acts as a negative regulator in antiviral response through deubiquitinating K63-linked
RIG-I. These findings provide insights into a potential new therapy targeting USP14 for
RNA virus-related diseases.

Keywords: Deubiquitination r RIG-I-like receptors r RNA virus sensor r USP14 r Vesicular


stomatitis virus (VSV)

 Additional supporting information may be found online in the Supporting Information section
at the end of the article.

Introduction associated gene 5, go through conformational changes and acti-


vate mitochondrial antiviral signaling protein (MAVS, also known
Vertebrates are continuously challenged with pathogenic microbes as VISA, IPS-1 and CARDIF) [4, 5] when infected by RNA virus.
that induce a variety of damages into host cells. In return, eukary- Subsequently, activated MAVS would recruit TBK1, then inter-
otic cells express PRRs that detect microbial PAMPs and acti- feron regulatory factor 3 (IRF3) or NF-κB to produce type I IFNs
vate immune responses [1]. PRRs, including TLRs, retinoic acid- or inflammatory cytokines [6].
inducible gene (RIG-I)-like receptors, NOD-like receptors (NLRs), Ubiquitin is a small, 76-amino acid protein that regulates pro-
and C-type lectin receptors, recognize specific microbes [2, 3]. tein stability, activity, and harbors at least seven types (K6, K11,
RIG-I-like receptors, such as RIG-I and melanoma differentiation- K27, K29, K33, K48 and K63) of ubiquitination modifications
[7, 8]. RIG-I ubiquitination is indispensable in antiviral immu-
nity [9, 10] and its K63-linked ubiquitination could be catalyzed
Correspondence: Prof. Weilin Chen
e-mail: cwl@zju.edu.cn by TRIM25 [11, 12], Riplet (RNF135, REUL) [13], TRIM4 [14],


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 43

and Mex3c [15] in cellular antiviral responses. Deubiquitination after being infected by VSV for 12 or 24 h. Data showed that USP14
of RIG-I could be regulated by deubiquitinating enzyme (DUBs). deficiency attenuated VSV replication in Mϕ (Fig. 1E) and THP1
USP25 was reported to negatively regulate Sendai virus (Sev)- cells (Fig. 1F). Above Data indicate USP14 deficiency elevates IFN-
induced type I IFN signaling by deubiquitinating RIG-I, TRAF2, β production and promotes cell antiviral ability exhibited by Mϕ
and TRAF6 [16]. USP3 inhibits vesicular stomatitis virus (VSV)- and THP1 cells.
induced type I IFN signaling by deubiquitinating K63-linked RIG-
I [17].
Ubiquitin-specific protease 14 (USP14) belongs to USP fam- IU1 enhances RIG-I-mediated IFN-β production and
ily of DUBs [18] and deubiquitinates proteasome-bound sub- inhibits VSV replication in macrophages
strates that were ubiquitinated at multiple sites [19–21]. Until
now, USP14 has been demonstrated to interact with Tau [22], Specific or nonspecific inhibitors targeting USP14 are signifi-
CXCR4 [23], prion [24], and NLRC5 [25]. Multiple studies illus- cant to both scientific studies and clinical therapies. Nonspecific
trated that USP14 regulated innate immune response. As an exam- inhibitors include WP1130 (Degrasyn) [19] and b-AP15 [30]. Spe-
ple, USP14 promotes dengue virus replication [26] and IL-1β pro- cific inhibitors contain VLX1570 [31] and IU1 screened by Lee et
duction [27, 28]. al [32]. IU1 specificity had been confirmed by quite a few stud-
To hunt DUBs interacting with RIG-I, the binding proteins of ies [26, 33–37]. IU1 structure is shown in Supporting Information
RIG-I were enriched in RIG-I overexpressed HEK293T cells using Fig. S2A. Previous studies showed IU1 inhibited flaviviruses repli-
co-IP assay. The immunoprecipitates were separated by SDS-PAGE cation [26]. In this study, we speculated whether IU1 suppressed
gel, and then the proteins were cut from the gel and analyzed by RNA virus propagation. First, we chose the optimal concentra-
mass spectrometric (MS) analysis. Among the RIG-I-interacting tions of IU1 by comparing IFN-β mRNA expressions induced by
proteins, we focused on USP14 and the highest frequency of spe- VSV. RT-qPCR analysis showed that IU1-pretreated cells at the
cific peptides also indicated USP14 to be a candidate for RIG-I concentration of 100 μM exhibited the highest IFN-β mRNA lev-
interacting partner (Supporting Information Fig. S1A and Table els (Supporting Information Fig. S2B). Cell toxicity of IU1 was
S1).In the present study, we demonstrated that USP14 negatively calibrated and exhibited minor influence (Supporting Information
regulated RNA virus-triggered type I IFN signaling. USP14-specific Fig. S2C). In summary, 100 μM was treated as the working con-
inhibitor, IU1, increased RIG-I-mediated type I IFN production and centration.
antiviral responses in vitro and in vivo. As a DUB, USP14 inter- To verify whether IU1 influenced cell antiviral ability, we
acted with RIG-I and removed its K63-linked polyubiquitination treated cells with IU1/DMSO before challenged with VSV or intra-
chains. In conclusion, our study is the first to confirm USP14 as a cellular poly(I:C). IFN-β mRNA production, mRNA and protein
negative regulator in RIG-I-mediated antiviral immune response levels, was measured. Except for VSV induced higher levels of IFN-
through K63-linked RIG-I deubiquitination. β in IU1-treated groups, cells transfected with poly(I:C) produced
more IFN-β mRNA, likewise in Mϕ (Fig. 2A) and THP1 cells (Fig.
2C). IFN-β secretion by IU1-treated cells was significantly higher
Results than DMSO treated ones in Mϕ (Fig. 2B) and THP1 cells (Fig. 2D),
and similar difference was seen in IFN-α expression (Supporting
USP14 knockdown increases IFN-β production and Information Fig. S2D). Furthermore, to investigate whether IU1
inhibits VSV replication in macrophages regulated VSV replication, cell supernatants were collected and
analyzed by TCID50. Less VSV replications in IU1-treated cells
To understand USP14 function in RIG-I signaling, IFN-β produc- were detected in Mϕ and THP1 cells (Fig. 2E). Two more meth-
tions were measured in mouse peritoneal macrophages and THP1 ods were applied as follows in THP1 cells. Immunofluorescence
macrophages. First, specific siRNAs targeting mouse and human assay indicated that IU1-treated groups exhibited less quantity
USP14 genes were designed, and knockdown efficiency was deter- of green fluorescence (Fig. 2F). THP1 cells were pretreated with
mined by RT-qPCR and immunoblot analysis separately (Support- IU1/DMSO followed by VSV-eGFP infection. Flow cytometry anal-
ing Information Fig. S1B and S1C). RIG-I can be activated by ysis showed that the percentage of GFP positive cells was lower
short ssRNA, dsRNA, or RNAs with 5’pp or 5’ppp end [29], such as in IU1-treated THP1 cells than that of control (Fig. 2G, Support-
VSV, ssRNA virus, and intratransfected polyinosinic:polycytidylic ing Information Fig. S2E). Data of this part demonstrate that IU1
acid (poly[I:C]) with low molecular weight. IFN-β production was could enhance IFN-β production and attenuates VSV replication.
higher in USP14 knockdown mouse peritoneal macrophage (Mϕ)
than control cells infected with VSV or transfected with poly(I:C)
both in mRNA and protein levels (Fig. 1A and B). USP14 siRNA USP14 overexpression attenuates IFN-β production
also elevated IFN-α mRNA levels and protein expression that is and promotes VSV replication in HeLa cells
triggered by VSV (Supporting Information Fig. S1D and S1E).
Similar difference of IFN-β expression was detected in THP1 cells HeLa cell is an immortal cell line that had been used in studies
(Fig. 1C and D). To examine USP14 function on VSV replication, on virus [38–40]. First, USP14 overexpression in HeLa cells was
cell supernatants were collected and analyzed for TCID50 analysis measured (Supporting Information Fig. S3A). To weigh the role


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
44 Hongrui Li et al. Eur. J. Immunol. 2019. 49: 42–53

Figure 1. Knockdown of USP14 increases RIG-I-mediated IFN-β production and inhibits VSV replication in macrophages. (A–D) Mouse peritoneal
macrophage or THP1 cells were infected with VSV or intracellular poly(I:C) after transfected with scrambled siRNA or USP14 siRNA for 36 h. IFN-β
mRNA levels (A) and protein levels (B) were measured by RT-qPCR or ELISA in mouse peritoneal macrophage (Mϕ). IFN-β mRNA levels (C) and
protein levels (D) were measured by RT-qPCR or ELISA in THP1 cells. (E–F) TCID50 analysis of Mϕ (E) and THP1 cells (F) with VSV infected for 12 to 24
h were conducted in HEK293T cells. (A–F) Data are shown as mean ± SD of three samples per group and are from one experiment representative of
three independent experiments. Significance was calculated in relation to the control group. *p < 0.05, **p < 0.01, ***p < 0.001 (two-sided Student’s
t-test).

of USP14 overexpression in RIG-I mediated antiviral response, cated that IU1-treated mice exhibited less inflammation (Fig. 4D).
IFN-β mRNA levels were determined and showed a lower level in Challenged with the lethal dose of VSV, mice treated with IU1
USP14 overexpressed HeLa cells infected with VSV or intracellu- were more resistant to VSV infection than control in a durable
lar poly(I:C) (Fig. 3A). IFN-β secretions were measured by ELISA observation (Fig. 4E). Data demonstrated that IU1-treated mice
and exhibited similar difference between control and USP14 over- gained greater antiviral ability against VSV infection by producing
expression cells (Fig. 3B). VSV replications were determined by more IFN-β.
three assays. TCID50 analyses showed that USP14 overexpression
increased virus titers (Fig. 3C). HeLa cells undergone fluorescence
microscopy (Fig. 3D) and flow cytometry analysis (Fig. 3E, Sup- USP14 targets and interacts with RIG-I
porting Information Fig. S3B) to detect GFP-positive cells after
being infected by VSV-eGFP. Results indicated that USP14 over- Next, we dissected the mechanisms on which USP14 regulated
expression increased the percentage of GFP-positive cells. Data RIG-I mediated type I IFN production. By knocking down USP14
suggest that USP14 is in favor of VSV infection in HeLa cells. or IU1 inhibiting in Mϕ, we detected IFN-β mRNA following stim-
ulation with LPS/poly(I:C)/VSV. Data indicated that USP14 had
no significant influence on TLR3- and TLR4-activated IFN-β (Fig.
IU1 enhances the antiviral response of mice in vivo 5A) and IFN-α (Supporting Information Fig. S4) production. To
investigate the role of USP14 in regulating RIG-I mediated sig-
To prove that IU1 influenced antiviral response in vivo, IU1 and naling, we therefore used VSV to stimulate Mϕ and THP1 cells.
DMSO were preintraperitoneally injected into mice before VSV We observed that IRF3 and TBK1 phosphorylation was elevated in
infection. Higher IFN-β mRNA levels were detected in IU1-treated USP14 knockdown cells (Fig. 5B). Next, dual-luciferase reporter
mice than that of control in lung and spleen (Fig. 4A). VSV repli- assay was applied to investigate the target of USP14 in type I
cations were significantly reduced in IU1-treated mice compared IFN signaling pathway. Data showed that USP14 just suppressed
to control in lung and spleen (Fig. 4B). IU1-treated mice produced RIG-I-N-activated IFN-β and ISRE luciferase activation, instead
more IFN-β in serum (Fig. 4C). The H & E staining of lungs indi- of MAVS, TBK1, and IRF3-5D (Fig. 5C). This illustrated that


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 45

Figure 2. IU1 elevated RIG-I-mediated IFN-β production and inhibited VSV replication in macrophages. (A–D) Mϕ and THP1 cells were pretreated
with IU1or DMSO for 2 h followed stimulation by VSV or intracellular poly(I:C). IFN-β mRNA levels (A) and protein levels (B) were measured by
RT-qPCR or ELISA in Mϕ. IFN-β mRNA levels (C) and protein levels (D) were measured by RT-qPCR or ELISA in THP1 cells. (E) TCID50 analysis of Mϕ
and THP1 cells with VSV infected for 12 to 24 h were conducted in HEK293T cells. (F) THP1 cells infected with VSV-eGFP undergone fluorescence
microscopy (left) for 12 (top) to 16 h (bottom) (40-fold), bar = 100 μm, and average fluorescence intensity by histogram (right). (G) THP1 cells
infected with VSV-eGFP for 12 to 16 h were prepared for flow cytometry (left) at MOI = 0.01 (top), 0.025 (middle), and 0.05 (bottom), and green
fluorescence intensity is shown by histogram (right). (A–E) Data are shown as mean ± SD of three samples per group and are from one experiment
representative of three independent experiments. Significance was calculated in relation to the control group. *p < 0.05, **p < 0.01, ***p < 0.001
(two-sided Student’s t-test).


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
46 Hongrui Li et al. Eur. J. Immunol. 2019. 49: 42–53

Figure 3. USP14 overexpression attenuated IFN-β mRNA expression and promoted VSV replications in HeLa cells. (A–E) Hela cells were transfected
with Flag-USP14 plasmids (Flag-USP14) or empty vector (control) for 24 h followed by VSV or intracellular poly(I:C) for 0, 4, 8, 12, or 16 h. (A) IFN-β
mRNA was quantified as described in the Materials and methods. (B) IFN-β production was measured by ELISA at 0 and 12 h. (C) TCID50 analysis
of VSV replication in cells infected with VSV for 12 to 24 h. (D) HeLa cells infected with VSV-eGFP for 16 h undergone fluorescence microscopy
examination (40 ×) (left), bar = 100 μm. Images are representative of three independent experiments. Histogram shows the average fluorescence
intensity (right). (E) Cells were infected with VSV-eGFP for 16 h and then analyzed by flow cytometry. Data were analyzed by FlowJo software (left)
and histogram (right) showed green fluorescence intensity. Similar results were obtained from three independent experiments. (A–E) Data are
shown as mean ± SD of three samples per group and are from one experiment representative of three independent experiments. Significance was
calculated in relation to the control group. *p < 0.05, **p < 0.01, ***p < 0.001 (two-sided Student’s t-test).

USP14 participated in antiviral response through RIG-I. For fur- at N-terminal was responsible for DUB activity. To determine the
ther verification, IPs between exogenous Flag-USP14 and Myc- domain responsible for the interaction, we generated two deleted
RIG-I/MAVS/TBK1/IRF3 were conducted. As expected, USP14 constructs of USP14. HEK293T cells were cotransfected with plas-
interacted with RIG-I (Fig. 5D). Together, these data indicated mids expressing USP14-FL/USP14-Ubl/ USP14-USP and RIG-I.
that USP14 regulated RIG-I activation when stimulated by RNA Co-IP and immunoblot analysis revealed that the USP domain
virus. of USP14, instead of Ubl domain, interacted with RIG-I (Fig. 5G).
Next, we examined the interaction between USP14 and RIG- These results show that USP14 regulates RNA virus-induced type
I with two IP assays. Plasmids expressing USP14 or RIG-I were I IFN signaling pathway through interacting with RIG-I by its USP
cotransfected into HEK293T cells. The co-IP was operated by domain.
anti-Myc antibodies and analyzed by immunoblot, and the inter-
action was validated (Fig. 5E). Endogenous USP14 from Mϕ
lysates infected with VSV for 4 h were immunoprecipitated by USP14 deubiquinates K63-linked RIG-I
anti-USP14 antibodies. The immunoblot analysis indicated that
USP14 and RIG-I interacted with each other. Additionally, as We hypothesized that USP14 promotes RIG-I deubiquitination.
time went by, the interaction between USP14 and RIG-I turned To test the hypothesis, IP and immunoblot were conducted in
intense (Fig. 5F). USP14 contains two domains, ubiquitin-like HEK293T cells. Results showed that USP14 reduced RIG-I ubiq-
(Ubl) domain, which was responsible for interaction with protea- uitination (Fig. 6A). Further, we cotransfected HEK293T cells
some at C-terminal and ubiquitin-specific protease (USP) domain with plasmids expressing Myc-RIG-I, Flag-USP14, and HA-Ub with


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 47

Figure 4. IU1 enhances the antiviral response of mice in vivo. (A–D) Mice (6 weeks old, n = 8) were infected with VSV by intraperitoneal injection
for 12 h after IU1 or DMSO treatment for 2 h. (A) Quantified IFN-β mRNA expression of lungs and spleens as detailed in the Materials and
methods. (B) VSV replications from mice lungs and spleens were quantified by TCID50. (C) ELISA analyzed the concentrations of IFN-β in serum
from IU1- or DMSO-treated mice. (D) Hematoxylin–Eosin staining of lungs (200 ×) from mice in (A), bar = 100 μm. Images are representative of
three independent experiments. (E) Mice (6 weeks old, n = 12) pretreated with IU1 or DMSO for 2 h were injected with a lethal dose of VSV by
intraperitoneal injection, and survival was followed for 60 h. (A–C) Data are shown as mean ± SD of a single experiment from three independent
experiments (eight mice/group/experiment). (E) Data are shown as mean ± SD of a single experiment from two independent experiments (12
mice/group/experiment). **p < 0.01, ***p < 0.001.

WT/K6/K11/K27/K29/K33/K48 or K63 and immunoprecipitated Previous studies of USP14 focused mainly on neuron diseases
with anti-Myc antibodies. Immunblot analysis showed that USP14 and cancer. USP14 loss in ax (J) mice causes a deficit in paired-
attenuated K63-linked RIG-I ubiquitination (Fig. 6B and C). To pulse facilitation at hippocampal synapses [51]. USP14 regulates
confirm the discovery above, endogenous experiment was con- presynaptic function independently of its role in deubiquitina-
ducted in Mϕ that was infected with VSV. Cell lysates were tion [52]. USP14’s catalytic activity is required for nervous system
immunoprecipitated with anti-RIG-I antibodies. Immunoblot anal- structure, function, and neuromuscular junction synaptic trans-
ysis suggested that USP14 knockdown increased RIG-I K63-linked mission [53]. USP14 is also important in cancer. The deficiency
ubiquitination (Fig. 6D). USP domain was responsible for the of USP14 inhibits breast cancer cells proliferation, metastasis, and
interaction between USP14 and RIG-I. Further, we examined the promotes apoptosis [54]. In esophageal squamous cell carcinoma
truncated mutants of USP14 on RIG-I ubiquitination. Data demon- cells, proliferation and tumor genesis were inhibited by reduced
strated that USP domain of USP14 was in charge of K63-linked expression of USP14 [55].
RIG-I deubiquitination (Fig. 6E). These results suggest that USP14 In innate immune response, when induced by LPS, USP14
promotes K63-linked RIG-I deubiquitination through USP domain. increased the production of IL-6 and TNF-α, ERK1/2 phospho-
rylation, and NF-кB activation [56]. But Meng et al. showed
that USP14 negatively regulates NF-кB signaling by removing the
polyubiquitin chains from NLRC5 to enhance NLRC5-mediated
Discussion
inhibition of NF-κB activation [25]. We discovered that USP14
suppressed VSV-induced RIG-I activation through deubiquitinat-
DUBs play crucial roles in the control of immune pathways. For
ing K63-linked RIG-I.
example, DUBs could regulate innate immunity in TLRs- and
And USP14 promoted IL-6 and TNF-α production triggered
NLRs-mediated immune signaling. USP2a [41], USP10 [42], and
by VSV or transfected poly(I:C). Immunoblotting analysis indi-
USP20 [43] regulate immune response through targeting TRAF6.
cated that phosphorylation of P65 enhanced significantly in USP14
USP4 [44] and USP18 [45] target TAK1, USP25 [46] targets
knockdown cells and negligible difference was observed in ERK,
TRAF3, A20 [47] and OTULIN [48] could target RIPK1/2 [49]. In
JNK, and P38 phosphorylation. The inconsistency of conclu-
antiviral immune response, RIG-I, which mediated type I IFN pro-
sions from different labs may result from different experiment
duction, is undependable for controlling virus propagation. DUBs
conditions.
directly regulate RIG-I K63-linked ubiquitination that includes
The present study identified USP14 as a negative regulator in
USP3, USP15, USP21, and CYLD. RIG-I K48-linked ubiquitination
antiviral response through its ability to bind to and deubiquitinate
could be regulated by USP4 [50].


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
48 Hongrui Li et al. Eur. J. Immunol. 2019. 49: 42–53

Figure 5. USP14 targets and interacts with RIG-I. (A) Mϕ transfected with scrambled siRNA/USP14 siRNA or DMSO/IU1 for 36 h were infected with
poly(I:C) for 4 h, LPS for 4 h, and VSV for 8 h. IFN-β mRNA expression was measured by RT-qPCR as detailed in the Materials and methods. (B) Mϕ
and THP1 cells were transfected with control siRNA or USP14 siRNA for 36 h and then infected with VSV for indicated hours. Cell lysates were
separated by SDS–PAGE and immunoblotted with the indicated antibodies. (C) Dual-luciferase reporter assay was used to analyze the activation of
IFN-β and ISRE by RIG-I-N, MAVS, TBK1, and IRF3-5D with or without USP14 overexpression as detailed in Materials and methods. (D) HEK293T cells
were cotransfected with Flag-USP14 and Myc-RIG-I/MAVS/TBK1/IRF3. Cell lysates were immunoprecipitated with anti-Flag antibodies. (E) HEK293T
cells were cotransfected with Flag-USP14, Myc-RIG-I, and followed by immuneprecipitation with anti-Myc antibodies. (F) Mϕ were infected with
VSV for 0, 4, 8 h, then cell lyslates were immunoprecipitated with anti-USP14 antibodies and immunoblot analyzed the interaction between USP14
and RIG-I. (G) Schematic structure of USP14 and the derivatives are shown. HEK293T cells were cotransfected with Flag-USP14, Flag-USP14-Ubl,
Flag-USP14-USP, Myc-RIG-I, followed by immuneprecipitation with anti-Flag antibodies. (A, C) Data are shown as mean ± SD of three samples per
group and are from one experiment representative of three independent experiments. Significance was calculated in relation to the control group.
*p < 0.05, ***p < 0.001 (two-sided Student’s t-test). (B, D–G) Data are from one experiment representative of three independent experiments.


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 49

Figure 6. USP14 removed K63-linked polyubiquitination conjugates from RIG-I. (A) HEK293T cells were co-transfected with Flag-USP14, Myc-RIG-I
and HA-Ub. Cell lyslates were precipitated with anti-Myc antibodies. Immunoblot analyzed the ubiquitination of RIG-I. (B) HEK293T cells were
co-transfected with Flag-USP14, Myc-RIG-I, HA- Ub including wild type (WT), K6, K11, K27, K29, K33, and K48. Cell lyslates were precipitated
with anti-Myc antibodies. Immunoblot analyzed the ubiquitination of RIG-I. (C) Plasmids expressing Flag-USP14, Myc-RIG-I and HA-Ub including
WT, K63 and K48 were co-transfected into HEK293T cells. Immunoprecipitation and immunoblot analyzed the ubiquitination of RIG-I. (D) Mϕ
was transfected with scrambled siRNA or USP14 siRNA for 36 h, then infected with VSV for 0 and 4 h. Cell lyslates were precipitated with
anti-RIG-I antibodies and immunoblot analyzed the ubiquitination of wild type and K63. (E) The structure of USP14 truncates (top). HEK293T
cells cotransfected with Flag-USP14, Flag-USP14-Ubl, Flag-USP14-USP, Myc-RIG-I, and HA-Ub-K63 for 24 h, followed by immunoprecipitation with
anti-Myc antibodies. (A–E) Blots are from one experiment representative of three independent experiments.


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
50 Hongrui Li et al. Eur. J. Immunol. 2019. 49: 42–53

RIG-I. Knockdown of USP14 by siRNA significantly enhanced Antibodies and virus


RIG-I-triggered type I IFN signaling and inhibited VSV replication
both in Mϕ and THP1 macrophages. Inhibition of USP14 by IU1 Anti-c-Myc (sc-40), anti-HA (sc-805), and anti-Flag (sc-807) were
increased RIG-I-mediated type I IFN production and antiviral purchased from Santa Cruze Biotechnology (Dallas, Tx, USA).
responses in vitro and in vivo. USP14 overexpression in HeLa Antibodies specific to p-IRF3 (4947), IRF3 (4302), p-TBK1 (5483),
cells attenuated cell antiviral ability. In addition, we found that TBK1 (3504), USP14 (11931), ubiquitin (3936), and K63-linked
USP14 acted by interacting with RIG-I and removing K63-linked ubiquitin (12930) were purchased from Cell Signaling Technology
polyubiquitination chains from RIG-I. (Danvers, MA, USA). Antibody against DDX58 (RIG-I) (20056-1-
We found that IU1 treatment exhibited stronger influence on AP) was purchased from Proteintech (Rosemont, IL, USA). VSV
IFN-β production induced by VSV than USP14 knockdown. The and VSV-eGFP belong to our lab.
answer may be that IU1 directly inhibits the activity of USP14, but
USP14 siRNA works by reducing USP14 expression that indirectly
prevents the activity of USP14. However, there are some flaws in
this study, i.e. no USP14-null mice were included. In the future, VSV and reagents treatment
we wish to create a new mouse line with systemic or tissue/cell-
specific deletion of USP14 to further confirm our findings. In sum- MOI of Mϕ, THP1 cells, and HeLa cells were 1, 0.01, and 1 sep-
mary, our study has identified a previously unrecognized role for arately. The regular dose of VSV for mice was 1× 108 PFU/g and
USP14 in the negative regulation of antiviral response through lethal dose was 5 × 108 PFU/g. Dosage of IU1 in cells lines
deubiquitinating K63-linked RIG-I. These findings provide insights and mice were 100 μM and 10 mg/kg separately. Intracellular
into a potential new therapy specifically targeting USP14 for RNA and extracellular infection of poly(I:C) was 1 μg and 20 μg/mL,
virus-related diseases. respectively, and LPS was 100 ng/mL.

Cell transfection and dual-luciferase reporter assay


Materials and methods
Transfection and dual-luciferase reporter assay were performed
Mice and cell culture as described previously [58]. Cells transfection was performed
in accordance with jetPEI (Polyplus transfection, Alsace, France)
C57BL/6J mice (female, 5–6 weeks) were procured from Joint instructions. Dual-luciferase reporter assay was performed in 96-
Ventures Sipper BK Experimental Animals, Shanghai, China. Mice well plate with 100 ng of firefly luciferase reporter plasmid and
were raised in pathogen-free conditions. All animal experiments 10 ng of Renilla luciferase reporter plasmid. All samples were set
were preceded according to the National Institute of Health Guide as triplicate. The folds were calculated relative to baseline control
for the Care and Use of Laboratory Animals with approval of the in each experiment.
Zhejiang University, Hangzhou.
Mϕ extraction and culture were as described before [57].
THP1 cells were cultured in 1640 medium with 10% FBS and
treated with PMA (1 μg/mL) for 24 h to be induced into THP1 RNA interference
macrophages. HeLa cells, HEK293T cells, and Vero cells were cul-
tured in DMEM medium with 10% FBS. Mϕ and THP1 cells were transfected according to Inter-
ferin instructions (Polyplus transfection). siRNAs were syn-
thesized by Gene Pharma (Shanghai, China). Mouse USP14
siRNA 5’-GGUAGAGAGACGGAUUCUUTT-3’, human USP14 siRNA
Plasmids and reagents 5’-GGAGAAAUUUGAAGGUGUA-3’, and scrambled siRNA 5’-
UUCUCCGAACGUGUCACGUdTdT-3’.
Myc-tagged RIG-I (Myc-RIG-I), Myc-tagged RIG-I (N) (Myc-RIG-
I-N), Myc-tagged MAVS (Myc-MAVS), Myc-tagged TBK1 (Myc-
TBK1), Myc-tagged IRF3 (Myc-IRF3), Myc-tagged IRF3 (N) (Myc-
IRF3-5D), Flag-tagged USP14 (Flag-USP14), Flag-tagged USP Mass spectrometry
domain of USP14 (Flag-USP14-USP), and Flag-tagged Ubl domain
of USP14 (Flag-USP14-Ubl) plasmids were constructed by our lab. MS analysis was performed as described by Song et al [59].
HA-Ub, HA-K63-Ub, HA-K48-Ub, HA-K33-Ub, HA-K29-Ub, HA- HEK293T cells were transfected with control vector, Flag-RIG-I,
K27-Ub, HA-K11-Ub, HA-K6-Ub plasmids were gifts from Prof. or Myc-RIG-I plasmids for 24 h and then infected with VSV for
Xuetao Cao (National Key Laboratory of Medical Immunology, 8 h before collecting. The cell lysates were immunoprecipitated
Shanghai, China). IU1 (S7134) was purchased from Selleck.cn by antibodies to Flag or Myc tag and undergo MS analysis to iden-
(Shanghai, China). tify RIG-I interacted DUBs.


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 51

RT-qPCR and ELISA Grant No. LZ17H100001 and the National Natural Science Foun-
dation of China (31670914, 31870908).
Total RNA of cells was extracted using Trizol, and cDNAs were We are grateful to Prof. Xuetao Cao (National Key Laboratory of
synthesized from RNAs according to manufacturer’s instructions Medical Immunology, Shanghai, China) for providing IRF3-5D,
(Takara, Japan). The products were subjected to quantitative RT- Ub, K48-Ub, and K63-Ub expression plasmids.
PCR (RT-qPCR) analysis using Hieff qPCR SYBR Green Master Mix
(No Rox) (YEASEN, Shanghai, China) and gene-specific primers
(Supporting Information Table S2). Calculation was conducted Conflict of interest: The authors declare no conflicts of interest.
relative to β-Actin by 2−t method with three replicates in each
three samples. The supernatants from infected cells or mouse
serum were subjected to ELISA using commercial mouse IFN-β References
kit (Biolegend, San Diego, CA, USA) and human IFN-β kit (PBL
assay science, NJ, USA) with three samples in each experiment. 1 Iwasaki, A. and Medzhitov, R., Regulation of adaptive immunity by the
innate immune system. Science 2010. 327: 291–295.

2 Long, L., Deng, Y., Yao, F., Guan, D., Feng, Y., Jiang, H., Li, X. et al.,
Recruitment of phosphatase PP2A by RACK1 adaptor protein deactivates
Immunoprecipitation and ubiquitination analysis
transcription factor IRF3 and limits type I interferon signaling. Immunity
2014. 40: 515–529.
IP and ubiquitination analysis were conducted in HEK293T cells
3 Huang, G., Li, L. and Zhou, W., USP14 activation promotes tumor pro-
and Mϕ, as we previously described [57]. HEK293T cells or Mϕ
gression in hepatocellular carcinoma. Oncol. Rep. 2015. 34: 2917–2924.
were seeded into 6 cm dishes for 24 h. HEK293T cells were trans-
4 Xia, P., Wang, S., Xiong, Z., Ye, B., Huang, L. Y., Han, Z. G. and
fected with plasmids for 24 h, and Mϕ were infected with VSV for
Fan, Z., IRTKS negatively regulates antiviral immunity through PCBP2
4 to 8 h. Cells then were lysed with NP40 lysis buffer. Lysates were sumoylation-mediated MAVS degradation. Nat. Commun. 2015. 6: 8132.
clarified by centrifugation and incubated with specific antibodies http//doi.org/10.1038/ncomms9132
for 1 h at 4°C followed by interacting with Protein A/G Sepharose 5 Pauli, E. K., Chan, Y. K., Davis, M. E., Gableske, S., Wang, M. K., Feister,
(Santa Cruze Biotechnology) overnight. The immunocomplexes K. F. and Gack, M. U., The ubiquitin-specific protease USP15 promotes
were recovered by brief centrifugation and washed five times with RIG-I-mediated antiviral signaling by deubiquitylating TRIM25. Sci. Signal.
2014. 7: ra3. http//doi.org/10.1126/scisignal.2004577
lysis buffer and resuspended in 2 × loading buffer. Protein lysates
were subjected to SDS–PAGE and electrophoretically transferred 6 Zhou, Y., He, C., Yan, D., Liu, F., Liu, H., Chen, J., Cao, T. et al., The kinase
CK1varepsilon controls the antiviral immune response by phosphorylat-
to nitro cellulose membranes (Bio-Rad, Hercules, CA, USA). And
ing the signaling adaptor TRAF3. Nat. Immunol. 2016. 17: 397–405.
subsequent immunoblot analysis was performed with indicated
7 McDowell, G. S. and Philpott, A., Non-canonical ubiquitylation: mecha-
antibodies.
nisms and consequences. Int. J. Biochem. Cell Biol. 2013. 45: 1833–1842.

8 Ikeda, F., Crosetto, N. and Dikic, I., What determines the specificity and
outcomes of ubiquitin signaling? Cell 2010. 143: 677–681.
Flow cytometric assay and immunofluorescence assay
9 Chattopadhyay, S., Fensterl, V., Zhang, Y., Veleeparambil, M.,
Yamashita, M. and Sen, G. C., Role of interferon regulatory factor 3-
Flow cytometric assay were conducted according to the guidelines
mediated apoptosis in the establishment and maintenance of persistent
[60]. Cells were infected with VSV-eGFP or PBS for 12 or 16 infection by Sendai virus. J. Virol. 2013. 87: 16–24.
h, and undergone pancreatin digestion following twice cold PBS
10 Shi, Y., Yuan, B., Zhu, W., Zhang, R., Li, L., Hao, X., Chen, S. et al.,
washing. Results were analyzed by FlowJo software. Cells were Ube2D3 and Ube2N are essential for RIG-I-mediated MAVS aggre-
pleated into 12-well plate and then infected with VSV-eGFP for gation in antiviral innate immunity. Nat. Commun. 2017. 8: 15138.
12 or 16 h, then the cells were washed twice with PBS, and then http//doi.org/10.1038/ncomms15138

plates were examined by fluorescence microscope. 11 Gack, M. U., Shin, Y. C., Joo, C. H., Urano, T., Liang, C., Sun, L., Takeuchi,
O. et al., TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-
mediated antiviral activity. Nature 2007. 446: 916–920.

Statistical analysis 12 Sanchez, J. G., Chiang, J. J., Sparrer, K. M., Alam, S. L., Chi, M., Roganow-
icz, M. D., Sankaran, B. et al., Mechanism of TRIM25 catalytic activation
in the antiviral RIG-I pathway. Cell Rep. 2016. 16: 1315–1325.
Statistical significances between groups were determined by two-
13 Oshiumi, H., Miyashita, M., Matsumoto, M. and Seya, T., A distinct role
tailed Student’s t-test and two-way ANOVA test.
of riplet-mediated K63-linked polyubiquitination of the RIG-I repressor
domain in human antiviral innate immune responses. PLoS Pathog. 2013.
9: e1003533. http//doi.org/10.1371/journal.ppat.1003533

14 Yan, J., Li, Q., Mao, A. P., Hu, M. M. and Shu, H. B., TRIM4 modulates type
I interferon induction and cellular antiviral response by targeting RIG-I
for K63-linked ubiquitination. J. Mol. Cell Biol. 2014. 6: 154–163.
Acknowledgements: This work was supported by grants from 15 Kuniyoshi, K., Takeuchi, O., Pandey, S., Satoh, T., Iwasaki, H., Akira, S.
Zhejiang Provincial Natural Science Foundation of China under and Kawai, T., Pivotal role of RNA-binding E3 ubiquitin ligase MEX3C in


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
52 Hongrui Li et al. Eur. J. Immunol. 2019. 49: 42–53

RIG-I-mediated antiviral innate immunity. Proc. Natl. Acad. Sci. USA. 2014. and induces apoptosis of multiple myeloma cells. Sci. Rep. 2016. 6: 26979.
111: 5646–5651. http//doi.org/10.1038/srep26979

16 Zhong, H., Wang, D., Fang, L., Zhang, H., Luo, R., Shang, M., 32 Lee, B. H., Lee, M. J., Park, S., Oh, D. C., Elsasser, S., Chen, P. C., Gartner, C.
Ouyang, C. et al., Ubiquitin-specific proteases 25 negatively regulates et al., Enhancement of proteasome activity by a small-molecule inhibitor
virus-induced type I interferon signaling. PLoS One 2013. 8: e80976. of USP14. Nature 2010. 467: 179–184.
http//doi.org/10.1371/journal.pone.0080976
33 Lahaie, N., Kralikova, M., Prezeau, L., Blahos, J. and Bouvier, M.,
17 Cui, J., Song, Y., Li, Y., Zhu, Q., Tan, P., Qin, Y., Wang, H. Y. et al., Post-endocytotic deubiquitination and degradation of the metabotropic
USP3 inhibits type I interferon signaling by deubiquitinating RIG-I-like gamma-aminobutyric acid receptor by the ubiquitin-specific protease 14.
receptors. Cell Res. 2014. 24: 400–416. J. Biol. Chem. 2016. 291: 7156–7170.

18 Shinji, S., Naito, Z., Ishiwata, S., Ishiwata, T., Tanaka, N., Furukawa, K., 34 McKinnon, C., Goold, R., Andre, R., Devoy, A., Ortega, Z., Moonga, J., Line-
Suzuki, H. et al., Ubiquitin-specific protease 14 expression in colorectal han, J. M. et al., Prion-mediated neurodegeneration is associated with
cancer is associated with liver and lymph node metastases. Oncol. Rep. early impairment of the ubiquitin-proteasome system. Acta Neuropathol.
2006. 15: 539–543. 2016. 131: 411–425.

19 Kapuria, V., Peterson, L. F., Fang, D., Bornmann, W. G., Talpaz, M. and 35 Ponnappan, S., Palmieri, M., Sullivan, D. H. and Ponnappan, U., Com-
Donato, N. J., Deubiquitinase inhibition by small-molecule WP1130 trig- pensatory increase in USP14 activity accompanies impaired proteasomal
gers aggresome formation and tumor cell apoptosis. Cancer Res. 2010. 70: proteolysis during aging. Mech. Ageing Dev. 2013. 134: 53–59.
9265–9276.
36 Santos, A. R., Mele, M., Vaz, S. H., Kellermayer, B., Grimaldi, M., Colino-
20 Xu, D., Shan, B., Sun, H., Xiao, J., Zhu, K., Xie, X., Li, X. et al., USP14 Oliveira, M., Rombo, D. M. et al., Differential role of the proteasome in
regulates autophagy by suppressing K63 ubiquitination of Beclin 1. Genes the early and late phases of BDNF-induced facilitation of LTP. J. Neurosci.
Dev. 2016. 30: 1718–1730. 2015. 35: 3319–3329.

21 Lee, B. H., Lu, Y., Prado, M. A., Shi, Y., Tian, G., Sun, S., Elsasser, S. et al., 37 Xu, Y. H. and Guo, N. L., USP14 inhibitor IU1 prevents ventilator-induced
USP14 deubiquitinates proteasome-bound substrates that are ubiquiti- lung injury in rats. Cell. Mol. Biol. (Noisy-le-grand) 2014. 60: 50–54.
nated at multiple sites. Nature 2016. 532: 398–401.
38 Tirumuru, N., Zhao, B. S., Lu, W., Lu, Z., He, C. and Wu, L., N(6)-
22 Lee, J. H., Shin, S. K., Jiang, Y., Choi, W. H., Hong, C., Kim, D. E. and Lee, methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag
M. J., Facilitated Tau degradation by USP14 aptamers via enhanced pro- protein expression. Elife 2016. 5: e15528.http//doi.org/10.7554/eLife.15528
teasome activity. Sci. Rep. 2015. 5: 10757. http//doi.org/10.1038/srep10757
39 Barrows, N. J., Campos, R. K., Powell, S. T., Prasanth, K. R., Schott-Lerner,
23 Mines, M. A., Goodwin, J. S., Limbird, L. E., Cui, F. F. and Fan, G. H., G., Soto-Acosta, R., Galarza-Munoz, G. et al., A screen of fda-approved
Deubiquitination of CXCR4 by USP14 is critical for both CXCL12-induced drugs for inhibitors of zika virus infection. Cell Host Microbe 2016. 20:
CXCR4 degradation and chemotaxis but not ERK ativation. J. Biol. Chem. 259–270.
2009. 284: 5742–5752.
40 Feeley, E. M., Sims, J. S., John, S. P., Chin, C. R., Pertel, T., Chen,
24 Homma, T., Ishibashi, D., Nakagaki, T., Fuse, T., Mori, T., Satoh, L. M., Gaiha, G. D. et al., IFITM3 inhibits influenza A virus infec-
K., Atarashi, R. et al., Ubiquitin-specific protease 14 modulates tion by preventing cytosolic entry. PLoS Pathog. 2011. 7: e1002337.
degradation of cellular prion protein. Sci. Rep. 2015. 5: 11028. http//doi.org/10.1371/journal.ppat.1002337
http//doi.org/10.1038/srep11028
41 He, X., Li, Y., Li, C., Liu, L. J., Zhang, X. D., Liu, Y. and Shu, H. B., USP2a
25 Meng, Q., Cai, C., Sun, T., Wang, Q., Xie, W., Wang, R. and Cui, J., negatively regulates IL-1beta- and virus-induced NF-kappaB activation
Reversible ubiquitination shapes NLRC5 function and modulates NF- by deubiquitinating TRAF6. J. Mol. Cell Biol. 2013. 5: 39–47.
kappaB activation switch. J. Cell Biol. 2015. 211: 1025–1040.
42 Wang, W., Huang, X., Xin, H. B., Fu, M., Xue, A. and Wu, Z. H., TRAF fam-
26 Nag, D. K. and Finley, D., A small-molecule inhibitor of deubiquitinating ily member-associated NF-kappaB activator (TANK) inhibits genotoxic
enzyme USP14 inhibits Dengue virus replication. Virus Res. 2012. 165: nuclear factor kappaB activation by facilitating deubiquitinase USP10-
103–106. dependent deubiquitination of TRAF6 ligase. J. Biol. Chem. 2015. 290:
13372–13385.
27 Lopez-Castejon, G., Luheshi, N. M., Compan, V., High, S., Whitehead,
R. C., Flitsch, S., Kirov, A. et al., Deubiquitinases regulate the activity of 43 Jean-Charles, P. Y., Zhang, L., Wu, J. H., Han, S. O., Brian, L., Freedman,
caspase-1 and interleukin-1beta secretion via assembly of the inflamma- N. J. and Shenoy, S. K., Ubiquitin-specific protease 20 regulates the recip-
some. J. Biol. Chem. 2013. 288: 2721–2733. rocal functions of beta-Arrestin2 in toll-like receptor 4-promoted nuclear
factor kappaB (NFkappaB) activation. J. Biol. Chem. 2016. 291: 7450–
28 Mialki, R. K., Zhao, J., Wei, J., Mallampalli, D. F. and Zhao, Y., Overex-
7464.
pression of USP14 protease reduces I-kappaB protein levels and increases
cytokine release in lung epithelial cells. J. Biol. Chem. 2013. 288: 15437– 44 Fan, Y. H., Yu, Y., Mao, R. F., Tan, X. J., Xu, G. F., Zhang, H., Lu, X. B.
15441. et al., USP4 targets TAK1 to downregulate TNFalpha-induced NF-kappaB
activation. Cell Death Differ. 2011. 18: 1547–1560.
29 Goubau, D., Schlee, M., Deddouche, S., Pruijssers, A. J., Zillinger, T.,
Goldeck, M., Schuberth, C. et al., Antiviral immunity via RIG-I-mediated 45 Liu, X., Li, H., Zhong, B., Blonska, M., Gorjestani, S., Yan, M., Tian, Q.
recognition of RNA bearing 5’-diphosphates. Nature 2014. 514: 372–375. et al., USP18 inhibits NF-kappaB and NFAT activation during Th17 differ-
entiation by deubiquitinating the TAK1-TAB1 complex. J. Exp. Med. 2013.
30 Chitta, K., Paulus, A., Akhtar, S., Blake, M. K., Caulfield, T. R., Novak,
210: 1575–1590.
A. J., Ansell, S. M. et al., Targeted inhibition of the deubiquitinating
enzymes, USP14 and UCHL5, induces proteotoxic stress and apoptosis 46 Zhong, B., Liu, X., Wang, X., Liu, X., Li, H., Darnay, B. G., Lin, X., et al.,
in Waldenstrom macroglobulinaemia tumour cells. Br. J. Haematol. 2015. Ubiquitin-specific protease 25 regulates TLR4-dependent innate immune
169: 377–390. responses through deubiquitination of the adaptor protein TRAF3. Sci.
Signal. 2013. 6: ra35. http//doi.org/10.1126/scisignal.2003708
31 Wang, X., Mazurkiewicz, M., Hillert, E. K., Olofsson, M. H., Pier-
rou, S., Hillertz, P., Gullbo, J. et al., The proteasome deubiquitinase 47 Hitotsumatsu, O., Ahmad, R. C., Tavares, R., Wang, M., Philpott, D.,
inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 Turer, E. E., Lee, B. L. et al., The ubiquitin-editing enzyme A20 restricts


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu
Eur. J. Immunol. 2019. 49: 42–53 Innate immunity 53

nucleotide-binding oligomerization domain containing 2-triggered sig- 57 Zhao, X., Zhu, H., Yu, J., Li, H., Ge, J. and Chen, W., c-Cbl-mediated ubiq-
nals. Immunity 2008. 28: 381–390. uitination of IRF3 negatively regulates IFN-beta production and cellular
antiviral response. Cell. Signal. 2016. 28: 1683–1693.
48 Elliott, P. R., Nielsen, S. V., Marco-Casanova, P., Fiil, B. K., Keusekotten,
K., Mailand, N., Freund, S. M. et al., Molecular basis and regulation of 58 Versteeg, G. A., Rajsbaum, R., Sanchez-Aparicio, M. T., Maestre, A. M.,
OTULIN-LUBAC interaction. Mol. Cell 2014. 54: 335–348. Valdiviezo, J., Shi, M., Inn, K. S. et al., The E3-ligase TRIM family of pro-
teins regulates signaling pathways triggered by innate immune pattern-
49 Lopez-Castejon, G. and Edelmann, M. J., Deubiquitinases: novel ther-
recognition receptors. Immunity 2013. 38: 384–398.
apeutic targets in immune surveillance? Mediators Inflamm. 2016. 2016:
3481371. http//doi.org/10.1115/2016/3481371 59 Song, Y., Lai, L., Chong, Z., He, J., Zhang, Y., Xue, Y., Xie, Y. et al., E3
ligase FBXW7 is critical for RIG-I stabilization during antiviral responses.
50 Kumari, P. and Kumar, H., Viral deubiquitinases: role in evasion of anti-
Nat. Commun. 2017. 8: 14654. http//doi.org/10.1038/ncomms14654
viral innate immunity. Crit. Rev. Microbiol. 2018. 44: 304–317.
60 Cossarizza, A., Chang, H. D., Radbruch, A., Akdis, M., Andra, I., Annunzi-
51 Marshall, A. G., Watson, J. A., Hallengren, J. J., Walters, B. J., Dobrunz,
ato, F., Bacher, P. et al., Guidelines for the use of flow cytometry and cell
L. E., Francillon, L., Wilson, J. A. et al., Genetic background alters
sorting in immunological studies. Eur. J. Immunol. 2017. 47: 1584–1797.
the severity and onset of neuromuscular disease caused by the loss
of ubiquitin-specific protease 14 (usp14). PLoS One 2013. 8: e84042.
http//doi.org/10.1371/journal.pone.0084042

52 Walters, B. J., Hallengren, J. J., Theile, C. S., Ploegh, H. L., Wilson, S. M. Abbreviations: DUB: deubiquitinating enzyme · IRF3: interferon regula-
and Dobrunz, L. E., A catalytic independent function of the deubiquiti- tory factor 3 · Mϕ: mouse peritoneal macrophage · MAVS: mitochondrial
nating enzyme USP14 regulates hippocampal synaptic short-term plas- antiviral signaling protein · NF-κB: nuclear factor-kappa B · NLRs: NOD-
ticity and vesicle number. J. Physiol. 2014. 592: 571–586.
like receptors · poly(I:C): polyinosinic:polycytidylic acid · RIG-I: retinoic
53 Vaden, J. H., Bhattacharyya, B. J., Chen, P. C., Watson, J. A., Marshall, A. acid-inducible gene I · TBK1: TANK-binding kinase 1 · Ubl: ubiquitin-like
G., Phillips, S. E., Wilson, J. A. et al., Ubiquitin-specific protease 14 reg-
domain · USP14: ubiquitin-specific protease 14 · VSV: vesicular stom-
ulates c-Jun N-terminal kinase signaling at the neuromuscular junction.
atitis virus
Mol. Neurodegener. 2015. 10: 3. https//doi.org/10.1186/1750-1326-10-3

54 Zhu, L., Yang, S., He, S., Qiang, F., Cai, J., Liu, R., Gu, C. et al., Downregu-
Full correspondence: Prof. Weilin Chen, Institute of Immunology,
lation of ubiquitin-specific protease 14 (USP14) inhibits breast cancer cell
Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang,
proliferation and metastasis, but promotes apoptosis. J. Mol. Histol. 2016.
China
47: 69–80.
e-mail: cwl@zju.edu.cn
55 Zhang, J., Zhang, D. and Sun, L., Knockdown of ubiquitin-specific
protease 14 (USP14) inhibits the proliferation and tumorigenesis in
esophageal squamous cell carcinoma cells. Oncol. Res. 2017. 25: 249–257.

56 Liu, N., Kong, T., Chen, X., Hu, H., Gu, H., Liu, S., Chen, X. et al., Ubiquitin- Received: 15/3/2018
specific protease 14 regulates LPS-induced inflammation by increasing Revised: 15/10/2018
ERK1/2 phosphorylation and NF-kappaB activation. Mol. Cell. Biochem. Accepted: 19/11/2018
2017. 431: 87–96. Accepted article online: 22/11/2018


C 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.eji-journal.eu

You might also like