ArticlePDF Available

Extracellular fibrinogen-binding protein released by intracellular Staphylococcus aureus suppresses host immunity by targeting TRAF3

Authors:

Abstract and Figures

Many pathogens secrete effectors to hijack intracellular signaling regulators in host immune cells to promote pathogenesis. However, the pathogenesis of Staphylococcus aureus secretory effectors within host cells is unclear. Here, we report that Staphylococcus aureus secretes extracellular fibrinogen-binding protein (Efb) into the cytoplasm of macrophages to suppress host immunity. Mechanistically, RING finger protein 114, a host E3 ligase, mediates K27-linked ubiquitination of Efb at lysine 71, which facilitates the recruitment of tumor necrosis factor receptor associated factor (TRAF) 3. The binding of Efb to TRAF3 disrupts the formation of the TRAF3/TRAF2/cIAP1 (cellular-inhibitor-of-apoptosis-1) complex, which mediates K48-ubiquitination of TRAF3 to promote degradation, resulting in suppression of the inflammatory signaling cascade. Additionally, the Efb K71R mutant loses the ability to inhibit inflammation and exhibits decreased pathogenicity. Therefore, our findings identify an unrecognized mechanism of Staphylococcus aureus to suppress host defense, which may be a promising target for developing effective anti-Staphylococcus aureus immunomodulators.
Efb inhibits host pro-inflammatory responses a Quantitative polymerase chain reaction (qPCR) analysis of Tnf, Il1b, Il6, and Il12p40 mRNA from alveolar macrophage (MH-S) infected with Newman, ΔEfb, or ΔEfb + Flag − Efb for indicated times (MOI = 25; **P = 0.0014, 0.0052, ****P < 0.0001, ***P = 0.0003 in sequence, Tnf; ****P < 0.0001, Il1b; ****P < 0.0001, <0.0001, <0.0001, ***P = 0.0001, ****P < 0.0001, ***P = 0.0002, in sequence, Il6; **P = 0.0019, *P = 0.0126, 0.0391, 0.0118, **P = 0.0020, 0.0020, in sequence, Il12p40). b ELISA quantification of TNF-α, IL-1β, IL-6, and IL-12 levels in lung tissue homogenized in 1 ml PBS 24 h after infection with Newman, ΔEfb, or ΔEfb + Flag − Efb. C57BL/6 mice were infected by intratracheal administration with the test strains (2 × 10⁸ CFUs per mouse) for the indicated times (**P = 0.0095, ****P < 0.0001, in sequence, TNF-α; *P = 0.0109, ****P < 0.0001, in sequence, IL-1β; *P = 0.0187, ***P = 0.0008, in sequence, IL-6; *P = 0.0206, ***P = 0.0002, in sequence, IL-12). c Quantification of the bacterial CFUs of lung tissue homogenates obtained in b (***P = 0.0001, ****P < 0.0001). d, e Histopathology of lung tissues was assessed in H&E sections stained from mice infected for 24 h; scale bars, 1,000 μm (top) and 200 μm (bottom), the boxed areas at the top are enlarged below (**P = 0.0036, *P = 0.0110). Student’s two-tailed unpaired t-test (a, b, d) or two-tailed Mann–Whitney U test (c) was used for statistical analysis. Data are representative of three experiments with at least three independent biological replicates. The bars show the mean and standard deviation of n = 3 (a), n = 9 (b, c), and n = 5 (d) per group. Source data are provided as a Source Data file.
… 
This content is subject to copyright. Terms and conditions apply.
Article https://doi.org/10.1038/s41467-022-33205-z
Extracellular brinogen-binding protein
released by intracellular Staphylococcus
aureus suppresses host immunity by
targeting TRAF3
Xiaokai Zhang
1,6
, Tingrong Xiong
1,6
,LinGao
1,6
,YuWang
1,2,6
,LuxuanLiu
3
,
Tian Tian
1
,YunShi
4
, Jinyong Zhang
1
, Zhuo Zhao
1
, Dongshui Lu
1
,PingLuo
1
,
Weijun Zhang
1
,PingCheng
1
, Haiming Jing
1
,QiangGou
1
, Hao Zeng
1,7
,
Dapeng Yan
5,7
& Quanming Zou
1,7
Many pathogens secrete effectors to hijack intracellular signaling regulators in
host immune cells to promote pathogenesis. However, the pathogenesis of
Staphylococcus aureus secretory effectors within hostcellsisunclear.Here,we
report that Staphylococcus aureus secretes extracellular brinogen-binding
protein (Efb) into the cytoplasm of macrophages to suppress host immunity.
Mechanistically, RING nger protein 114, a host E3 ligase, mediates K27-linked
ubiquitination of Efb at lysine 71, which facilitates the recruitment of tumor
necrosis factor receptor associated factor (TRAF) 3. The binding of Efb to
TRAF3 disrupts the formation of the TRAF3/TRAF2/cIAP1 (cellular-inhibitor-of-
apoptosis-1) complex, which mediates K48-ubiquitination of TRAF3 to pro-
mote degradation, resulting in suppression of the inammatory signaling
cascade. Additionally, the Efb K71R mutant loses the ability to inhibit inam-
mation and exhibits decreased pathogenicity. Therefore, our ndings identify
an unrecognized mechanism of Staphylococcus aureus to suppress host
defense, which may be a promising target for developing effective anti-Sta-
phylococcus aureus immunomodulators.
S
taphylococcus aureus (S. aureus) is one of the most common bacterial
strains causing hospital- and community-acquired pneumonia asso-
ciated with signicant mortality worldwide1,2. It is estimated that ~85%
of the human population carries or previously carried S. aureus3.The
nasal cavity is the main site of S. aureus colonization in the human
body4, providing a pathogen reservoir that signicantly increases the
chance of secondary S. aureus pulmonary infection. However, the
morbidity associated with S. aureus pneumonia is far below the bac-
terial carrying rate due to the powerful host immune system. Resident
macrophages, an important class of innate immune cells, are the rst
to encounter invading pathogens5. Besides killing the invading
pathogens, macrophages can produce pro-inammatory cytokines,
Received: 31 January 2022
Accepted: 8 September 2022
Check for updates
1
National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third
Military Medical University, Chongqing 400038, China.
2
Department of Basic Courses, NCO School, Third Military Medical University, Shijiazhuang 050081,
China.
3
Collegeof Medicine,Southwest Jiaotong University,Chengdu 610083, China.
4
Institute of Biopharmaceutical Research, WestChina Hospital, Sichuan
University, Chengdu, Sichuan 610041, China.
5
Department of Immunology, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and
Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
6
These authors contributed equally: Xiaokai Zhang,
Tingrong Xiong, Lin Gao, Yu Wang.
7
These authors jointly supervised this work: Hao Zeng, Dapeng Yan, Quanming Zou. e-mail: zeng1109@163.com;
dapengyan@fudan.edu.cn;qmzou2007@163.com
Nature Communications | (2022) 13:5493 1
1234567890():,;
1234567890():,;
Content courtesy of Springer Nature, terms of use apply. Rights reserved
chemokines, and lipid mediators that recruit other innate immune
cells, such as neutrophils, monocytes, and dendritic cells, to orches-
trate immune responses and ght infections6. Therefore, resident
macrophages represent a critical defense line that S. aureus must
overcome in order to propagate in the host7.
S. aureus secretes multiple effectors that functionally evade or
inhibit host immune responses8. Previous pathogenic studies on S.
aureus have mainly focused on the effect of its virulent factors on the
cytomembrane and its receptors in innate immune cells. Several
hemolysins, leukocidins, and phenol-soluble modulins have been
identied to lyse cells by forming pores on the membranes of mac-
rophages and other innate immune cells9. Other bacterial factors,
including staphylococcal superantigen-like protein 3 and lipoylated
E2 subunit of the pyruvate dehydrogenase complex, released from S.
aureus can suppress macrophage activation through inhibiting Toll-
like receptors (TLR) activation10,11. Recently, it has been demonstrated
that S. aureus can invade and survive within macrophages and other
host cells12. Even in a hostile environment, such as that in macro-
phages, S. aureus can develop specic countermeasures to evade the
immune response. Numerous intracellular bacteria have been shown
to weaken host immune defenses bysecreting virulent factors capable
of hijacking macrophage signaling pathways13. However, which effec-
tors are secreted by intracellular S. aureus to manipulate the signaling
pathways of macrophages and their underlying mechanisms remains
unclear.
To inhibit the intracellular survival of invading pathogens, the
inammatory signaling pathway promotes the expression of various
pro-inammatory cytokines including tumor necrosis factor (TNF),
interleukin 1β(IL-1β), IL-6, and IL-1214,15.Inthepresentstudy,wefound
that extracellular brinogen-binding protein (Efb) released by intra-
cellular S. aureus inhibits pro-inammatory cytokine expression in
macrophages. Previous studies have suggested that Efb can block the
function of C3b, inhibit the formation of platelet-leukocyte complexes,
and bind brinogen to prevent neutrophil activation16,17. However, lit-
tle is known about the role of intracellular Efb in modulating host
inammatory signaling pathways.
In this work, we demonstrate that intracellular S. aureus secretes
Efb into the cytoplasm of macrophages to inhibit expression of pro-
inammatory cytokines and suppress host immunity by interacting
with tumor necrosis-associated factor 3 (TRAF3). Mechanistically, the
interaction between Efb and TRAF3 requires K27-linked ubiquitination
of Efb mediated by a host E3 ubiquitin ligase, RING nger protein 114
(RNF114).
Results
Efb inhibits host pro-inammatory responses
To identify the anti-inammatory components secreted by intracel-
lular S. aureus, we tested mature chains of 78 secretory proteins of S.
aureus on nuclearfactor-κB(NF-κB) activation in HEK293T cells using a
dual-luciferase reporter gene assay (SupplementaryFig. 1a, b). Efb was
one of the proteins shown to inhibit NF-κB activation induced by TNF-α
(Supplementary Fig. 1b). Efb was co-expressed with an NF-κBreporting
gene in HEK293T cells to verify the inhibitory role of intracellularly
expressed Efb on activation of the NF-κB pathway (Supplementary
Fig. 1c). Next, we found that adenoviral vector-mediated intracellular
overexpression of Efb strongly suppressed phosphorylation of p65
(Supplementary Fig. 1d) and mRNA levels of pro-inammatory cyto-
kines, including Tnf,Il1β,Il6,andIl12p40 (Supplementary Fig. 1e), in
peritoneal macrophages (PMs) stimulated with heat-killed S. aureus.
These results suggest that intracellular Efb inhibits the transcription of
pro-inammatory cytokines likely through inhibiting the NF-κB
pathway.
To validate the anti-inammatory activity of physiological Efb in
an infection model, we generated an S. aureus Newman strain with an
Efb-deletional mutant (ΔEfb) and a complementation strain (ΔEfb +
Flag Efb). As shown in Supplementary Fig. 2a, western blot analysis
with anti-Efb or anti-Flag antibodies and silver staining of poly-
acrylamide gel electrophoresis (PAGE) revealed Efb expression in the
culture supernatant of the corresponding strains. Consistent with a
previous report18, the absence of Efb did not affect S. aureus growth
(Supplementary Fig. 2b).
Next, we found that macrophages infected with the ΔEfb strain
signicantly increased the mRNA levels of Tnf,Il1β,Il6,andIl12p40 in
infected macrophages, while the ΔEfb + Flag Efb strain restored the
inhibitory effect of Efb (Fig. 1a, Supplementary Fig. 3a). However, the
inhibitory effect of Efb on the expression of inammatory cytokines
was lost after stimulation with only the culture supernatants of S.
aureus (Supplementary Fig. 3b) or non-contact co-culture of mac-
rophages and S. aureus in Transwells (Supplementary Fig. 3c). These
data suggest that Efb released by intracellular S. aureus, not Efb
released by extracellular S. aureus, exerts anti-inammatory func-
tions. In the control experiments, we found no difference in intra-
cellular S. aureus loading among macrophages infected with
Newman, ΔEfb, or ΔEfb + Flag Efb strains for 6 h (Supplementary
Fig. 3d, e, GFP did not affect the expression of Efb in S. aureus;
Supplementary Fig. 3f), indicating that the above-observed differ-
ences in mRNA levels of inammatory cytokines are Efb-dependent.
Compared to the Newman strain, the ΔEfb strain also exhibited
accelerated phosphorylation of p65, p38, JNK, and ERK in macro-
phages (Supplementary Fig. 4a, b), suggesting that Efb may also
inhibit the mitogen-activated protein kinase (MAPK) pathway in
addition to NF-κB pathway. These results provide further evidence
that physiological Efb released by intracellular S. aureus suppresses S.
aureus-triggered inammatory responses in macrophages.
To investigate the role of Efb in vivo, we established a trachea
cannula infection model by challenging 6-week-old C57BL/6 mice with
Newman, ΔEfb, or ΔEfb + Flag Efb strains. At 24 h post infection, there
was increased expression of TNF-α,IL-1β, IL-6, and IL-12 in the lungs of
mice infected with the ΔEfb strain compared to those infected with the
Newman and ΔEfb + Flag Efb strains (Fig. 1b). In agreement, the
bacterial burden in the lungs of Newman and ΔEfb + Flag Efb infected
mice was signicantly higher than that of ΔEfb infected mice (Fig. 1c).
There were also more intact alveolar spaces, along with less inltration
of neutrophils and lymphocytes, in the lungs of ΔEfb infected mice
(Fig. 1d, e). We also found that ΔEfb directly led to an increase in
survival rates in lethal pneumonia (Supplementary Fig. 5a) and bac-
teremia (Supplementary Fig. 5b) models, as well as alleviated festering
areas inthe skin infection model (Supplementary Fig. 5c, d), compared
to Newmanand ΔEfb + Flag Efb infected mice. Together, these results
suggest that Efb released by intracellular S. aureus may act as an inhi-
bitor of host inammatory responses, making Efb an important factor
for S. aureus infection.
Efb interacts with TRAF3
Immunoelectronmicroscopy and immunouorescent confocal micro-
scopy showed that Efb was secreted by intracellular S. aureus into the
cytoplasm of the macrophages and gradually accumulated over time
during infection (Supplementary Fig. 6a, b). TLR signaling, especially
TLR2, plays an important role in detecting S. aureus and inducing the
expression of pro-inammatory cytokines during infection10,19. There-
fore, using a co-immunoprecipitation assay, we screened Efb-
interactive proteins from a list of key signaling molecules in the TLR
pathway. TRAF3 appeared to be the only Efb-associated protein
(Supplementary Fig. 7a and Fig. 2a, b). Further experiments in ΔEfb +
Flag Efb infected macrophages demonstrated that Efb released by
intracellular S. aureus interacted physiologically with TRAF3 (Fig. 2c, d
and Supplementary Fig. 7b). The in vitro GST-pull down assay also
demonstrated a direct interaction between puried Efb and TRAF3
(Supplementary Fig. 7c, d). In addition, the zinc nger domain of
TRAF3 was responsible for the interaction (Fig. 2e, f).
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Efb inhibits pro-inammatory responses by stabilizing TRAF3
Next, we examined whether TRAF3 mediated the inhibition effects of
Efb on host pro-inammatory responses. We generated conditional
knockout mice for TRAF3 (genotype: TRAF3[ox/ox, Lyz2-Cre]) using
CRISPR/Cas9-mediated genome editing and isolating the PMs from
these mice. TRAF3 deciency effectively abrogated Efbs inhibitory
effects on the mRNA levels of Tnf,Il1β,Il6,andIl12p40 in PMs (Fig. 3a).
Furthermore, Efbs inhibitory effects were lost in lungs of TRAF3[ox/ox,
Lyz2-Cre] mice, but notin TRAF3[ox/ox] mice (Fig. 3b).Consistentwiththis
result, there were no signicant differences in the pulmonary bacteria
burdens and histopathology between TRAF3[ox/ox, Lyz2-Cre] mice infec-
ted with the Newman and ΔEfb strains (Fig. 3ce). TRAF3 mediatesNF-
κB, MAPK, and type I interferon pathways20. Therefore, we used NIK
SMI1 (NIK inhibitor), GSK8612 (TBK1 inhibitor), and 5Z-7-oxozeaeno
(TAK1 inhibitor) to test which pathway mainly mediates pro-
inammatory cytokine production of macrophages infected with S.
aureus. The results show that 5Z-7-oxozeaeno, but not NIK SMI1 and
GSK8612, inhibited pro-inammatory cytokine production. TAK1
mainly mediated the canonical NF-kB and MAPK pathways21. There-
fore, the canonical NF-kB and MAPK pathways may play a dominant
role in this mechanism. And the anti-inammatory effects of Efb could
still be observed in macrophages treated with NIK SMI1 and GSK8612,
but not with 5Z-7-oxozeaeno (Supplementary Fig. 8ac). Collectively,
these in vitro and in vivo results indicate that Efb may suppress host
inammatory responses via inhibiting TRAF3-mediated canonical NF-
κB and MAPK pathway activation.
Previous studies have demonstrated that K48 and K63 ubiquiti-
nation of TRAF3 is essential for activation of the NF-κBandMAPK
pathways2225. Here, we found that, compared to the Newman strain,
the ΔEfb strain markedly reduced the amounts of TRAF3 in macro-
phages, suggesting that Efb may stabilize TRAF3 (Fig. 4a and Supple-
mentary Fig. 9a). Consistently, both overexpressed and physiological
Efb signicantly reduced the K48-linked ubiquitination of TRAF3 in
HEK293T cells and macrophages (Fig. 4b, c, Supplementary Fig. 9b).
However, we found little evidence of K63-linked ubiquitination of
TRAF3 in macrophages infected with S. aureus (Supplementary
Fig. 9c), indicating that Efb may mainly affect the K48-linked ubiqui-
tination of TRAF3. The degradative ubiquitination of TRAF3 is typically
mediated by cIAP1/2, and the modication effects require TRAF2 to act
as a bridge between TRAF3 and cIAP1/226,27. We found that TRAF2-
mediated K48-linked ubiquitination of TRAF3 in macrophages infected
by S. aureus (Supplementary Fig. 9d), and Efb could prevent TRAF3
from interacting with TRAF2 and cIAP1 (Fig. 4d, e, Supplementary
Fig. 9e), resulting in inhibition of K48-linked ubiquitination of TRAF3 in
both a cell-free ubiquitylation reaction system and HEK293T cells
(Fig. 4f, Supplementary Fig. 9f). We further demonstrated that the zinc
nger domain of TRAF3 was responsible for its binding to TRAF2
(Supplementary Fig. 9g), suggesting that Efb could competitively bind
TRAF3 and affect TRAF2 binding to TRAF3.
Lysines 106 and 155 of TRAF3 were reported as the main K48-
linked ubiquitination sites28, and our results showed that Efb mainly
inhibited the K48-linked ubiquitination of lysine 155 in the zinc nger
Fig. 1 | Efb inhibits host pro-inammatory responses. a Quantitative polymerase
chain reaction (qPCR) analysis of Tnf,Il1b,Il6,andIl12p40 mRNA from alveolar
macrophage (MH-S) infected with Newman, ΔEfb, or ΔEfb + Flag Efb for indicated
times (MOI = 25; **P= 0.0014, 0.0052, ****P<0.0001, ***P=0.0003insequence,
Tnf;****P<0.0001, Il1b;****P< 0.0001, <0.0001, <0.0001, ***P=0.0001,
****P< 0.0001, ***P= 0.0002, in sequence, Il6;**P=0.0019,*P=0.0126, 0.0391,
0.0118, **P= 0.0020, 0.0020, in sequence, Il12p40). bELISA quantication of TNF-
α, IL-1β, IL-6, and IL-12 levels in lung tissue homogenized in 1ml PBS 24 h after
infection with Newman, ΔEfb, or ΔEfb + Flag Efb. C57BL/6 mice were infected by
intratracheal administration with the test strains (2 × 108CFUs per mouse) for the
indicated times (**P= 0.0095, ****P<0.0001, in sequence, TNF-α;*P=0.0109,
****P<0.0001, in sequence, IL-1β;*P= 0.0187, ***P= 0.0008, in sequence, IL-6;
*P= 0.0206, ***P= 0.0002, in sequence, IL-12). cQuantication of the bacterial
CFUs of lung tissue homogenates obtained in b (***P=0.0001, ****P< 0.0001).
d,eHistopathologyof lung tissues was assessed in H&E sections stained from mice
infectedfor 24 h; scale bars, 1,000μm(top)and200μm (bottom), the boxedareas
at the top are enlarged below (**P=0.0036, *P= 0.0110). Students two-tailed
unpaired t-test (a,b,d) or two-tailed MannWhitney Utest (c) was used for sta-
tistical analysis. Data are representative of three experiments with at least three
independent biological replicates. The bars show the mean and standard deviation
of n=3(a), n=9(b,c), and n=5(d) pergroup. Source dataare provided as a Source
Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved
domain of TRAF3 (Fig. 4g and Supplementary Fig. 9h). Next, we
expressed wild-type TRAF3 in TRAF3 knockout macrophages to
restore the function of TRAF3 and expressed TRAF3 K155R in TRAF3
knockout macrophages to mimic the effects of Efb on TRAF3. After
infection, the pro-inammatory cytokines induced by the ΔEfb stain
remained higher than the Newman str ain in wild-type TRAF3, but not in
TRAF3 K155R-expressing macrophages (Supplementary Fig. 9i). Taken
together, these results suggest that Efb disturbs the TRAF3/TRAF2/
cIAP1 complex, inhibits the K48-linked ubiquitination of TRAF3 at
lysine 155, and prevents TRAF3 degradation in macrophages during S.
aureus infections.
RNF114 mediated K27-linked ubiquitination of Efb
Several studies have suggested that bacteria can utilize the post-
translational modication system of the host to modify their effectors
for pathogenesis29,30. Given that Efb is small in size (mature protein,
15kD) and contains 20 lysine residues that are common sites for ubi-
quitination, we investigated the ubiquitination of intracellular Efb.
Plasmids expressing different ubiquitins (Ub, K6, K11, K27, K29, K33,
K48, and K63) were co-transfected with Efb into HEK293T cells. Co-
immunoprecipitation results revealed multiple types of ubiquitination
of Efb, with K27-linked polyubiquitination being most dominant
(Supplementary Fig. 10a). Furthermore, we detected the endogenous
K27-linked polyubiquitin conjugates of intracellular Efb in macro-
phages infected with the ΔEfb + Flag Efb strain (Fig. 5a, Supplemen-
tary Fig. 10b), indicating that Efb is polyubiquitinated by K27 in host
cells. We also carried out proteomics analysis on immunoprecipitated
overexpressed Efb in HEK293T cells using tandem mass spectrometry
(MS/MS) in Q ExactiveTM Plus (Thermo) coupled online to UPLC
(Supplementary Table 3). We identied a protein named RNF114 as an
E3 ligase with K27-linked polyubiquitination activity31. Overexpression
co-immunoprecipitation (Fig. 5b, c) and endogenous co-
immunoprecipitation (Fig. 5d, Supplementary Fig. 10c) consistently
demonstrated reproducible Efb interaction with RNF114. Over-
expression of RNF114 also increased the K27-linked (but not other
types), polyubiquitination of Efb in HEK293T cells (Fig. 5e, Supple-
mentary Fig. 11), and RNF114 KD by specic siRNA reduced the K27-
linked polyubiquitination of Efb in macrophages (Fig. 5f). These results
collectively suggest that RNF114 is an important ubiquitinase that
mediates K27-linked polyubiquitination of Efb.
To determine which lysine residue of Efb was modied by K27-
linked polyubiquitination, we constructed 20Efb mutants by replacing
lysine with arginine and co-transfected these mutants with a plasmid
expressing K27 ubiquitin in HEK293T cells. The immunoprecipitation
results revealed that the K71R mutant signicantly reduced the K27-
linked polyubiquitination of Efb (Supplementary Fig. 12a). Con-
sistently, RNF114 did not promote the K27-linked ubiquitination of
K71R mutant of Efb in HEK293T cells (Fig. 5g). Next, we replaced all
other Efb lysines with arginines, except lysine 71, and RNF114 still
maintained the K27-linked polyubiquitination of Efb at lysine 71 (Sup-
plementary Fig. 12b, c). Moreover, physiological K27-linked ubiquiti-
nationof the K71R mutant of Efb disappeared in macrophages (Fig. 5h).
These results suggest that RNF114 can mediate K27-linked poly-
ubiquitinion of Efb at lysine 71.
Fig. 2 | Efbinteracts withTRAF3. a,bImmunoblots of wholecell lysates (WCL)and
immunoprecipitation (IP) products of WCL from HEK293T cells transfected with
indicated plasmids. cImmunoblots of WCL and IP products of WCL from MH-S
infected with ΔEfb+ Flag Efb for indicated times (MOI = 25).
dImmunouorescence assay of MH-S infected with ΔEfb + FlagEfb for 4 h
(MOI = 25), scale bars, 2 μm. eTruncation of TRAF3. fImmunoblots of WCL and IP
products of WCL from HEK293T cells transfectedwith indicated plasmids; WT, full-
length TRAF3. Data are representative of three experiments with at least three
independent biological replicates. Source data are provided as a Source Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 4
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Ubiquitination of Efb facilitates the stabilization of TRAF3
To investigate whether RNF114 is involved in suppressing
TRAF3 signalingby Efb, we observed theenhancing effectof RNF114 on
Efb inhibiting K48-linked ubiquitination of TRAF3 (Supplementary
Fig. 13a). When compared to wild-type Efb, the K71R mutant appeared
to reverse the inhibition of Efb on the K48-linked ubiquitination of
TRAF3 (Supplementary Fig. 13b). The results obtained from ΔEfb +
Flag Efb and ΔEfb + Flag Efb K71R infected macrophages (Supple-
mentary Fig. 13ce) showed that the K71R-Efb mutant reduced the
inhibitory effects on TRAF2 and cIAP1 mediated K48-linked poly-
ubiquitination of TRAF3 at physiological levels. Furthermore, we found
that ΔEfb infection in RNF114 KD macrophages resulted in an even
lower level of pro-inammatory cytokine expression compared to
Newman infection (Supplementary Fig. 13f), and we found that RNF114
affected the TRAF3 expression level only when Efb was present (Sup-
plementary Fig. 13g), suggesting that RNF114 is involved in the reg-
ulation of inammatory cytokine production during S.aureus infection
in macrophages independent of Efb and TRAF3. Together, these
results suggest that RNF114 promotes K27-linked ubiquitination of Efb
to facilitate TRAF3 stabilization.
Ubiquitinated Efb inhibits host pro-inammatory responses
Next, the reconstitution experiments using ΔEfb infected macro-
phages demonstrated that wild-type Efb, but not the K71R-mutant
strain, was able to restore inhibition of infection-induced pro-inam-
matory cytokine production (Fig. 6a, Supplementary Fig. 13c). To
investigate the function of the K71R mutant on the innate immune
response in vivo, we infected 6-week-old mice with the Newman, ΔEfb,
ΔEfb + Flag Efb, or ΔEfb + Flag Efb K71R strains and evaluated the
expression levels of pro-inammatory cytokines in the lungs. Com-
pared with ΔEfb infection, ΔEfb + Flag Efb, but not ΔEfb + FlagEfb
(K71R), signicantly suppressed pro-inammatory cytokine produc-
tion in the affected lungs (Fig. 6b). The bacterial load of ΔEfb and
ΔEfb + Flag Efb (K71R) strains in the lungs was signicantly lower than
that of the Newman and ΔEfb + Flag Efb strains (Fig. 6c). Consistently,
histopathology results revealed more severe lung damage and more
neutrophil and lymphocyte inltration in the lungs of ΔEfb mice
reconstituted with wild-type but not the K71R-mutant Efb strains
(Fig. 6d, e). These results suggest that the pathogenesis of S. aureus
might rely on the K27-linked ubiquitination of Efb at its lysine 71
residue.
Discussion
Opsonophagocytic killing conducted by macrophages and neu-
trophils is critical for the host to combat S. aureus, a classical
extracellular pathogen7. However, in recent years, accumulating
reports revealed that S. aureus can invade and survive within mac-
rophages, neutrophils, and other host cells12. As a reservoir of S.
aureus, macrophages play an important role in regulating the
inammatory responses that are critical for eliminating invading
pathogens6,12. To achieve a successful infection, S. aureus must
overcome activation of the inammatory pathways of macrophages.
Fig. 3 | Efb inhibits host pro-inammatory responses via TRAF3.a qPCR analysis
of Tnf,Il1b,Il6,andIl12p40 mRNA of PMs infected with Newman, ΔEfb (MOI = 25 )
for 4 h; wild-type PMs (WT) were isolated from TRAF3[ox/ox] mice; TRAF3knockout
(KO) PMs were isolated from TRAF3[ox/ox, Lyz2-Cre] mice (**P= 0.0096, Tnf;
***P=0.0001, Il1b;**P= 0.0011, Il6;**P= 0.0015, Il12p40). bELISA quantication of
TNF-α, IL-1β, IL-6, and IL-12 levels in lung tissues from TRAF3[ox/ox] (Flox) or
TRAF3[ox/ox, Lyz2-Cre] (TRAF3 KO) mice, homogenized in 1 ml PBS and infected with
Newman, ΔEfb for 24 h (2 × 10 8CFUs per mouse; ****P< 0.0001, TNF-α;*P=0.0184,
IL-1β;*P= 0.0427,IL-6; **P= 0.0089,IL-12). cQuantication of the bacterial CFUs of
lung tissue homogenates obtained in b(**P=0.0030).d,eHistopathology of
lung tissues was assessed in H&E sections stained from mice infected for 24 h;
scale bars, 1000 μm(top)and200μm (bottom), the boxed areas at the top are
enlarged below (****P<0.0001).Students two-tailed unpaired t-test (a,b,d)or
two-tailed MannWhitney Utest (c) was used for statistical analysis. Data are
representative of three experiments with at least three independent biological
replicates. The bars show the mean and standard deviation of n=3(a), n=9
(b,d), and n=5(e) mice per group. Source data are provided as a Source
Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 5
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Secreting multiple effectors is a unique strategy that S. aureus has
adopted to cope with environmental challenges9. However, whether
and how the secreted effectors of intracellular S. aureus interfere
with the inammatory pathways of macrophages remains unclear. In
the present study, we screened 78 secreted effectors on activation of
the NF-κB pathway. The results showed that more than half of the
secretory proteins decreased NF-kB activation. We conducted sev-
eral bioinformatics and literature searches on these proteins,
especially α-hemolysin (WP-000857483.1), serine protease SplE
(WP-001038759.1), and cysteine protease staphopain A (WP-
000827748.1). We found that the inhibitory effects of these three
proteins were false positives due to the fact that overexpressing
these proteins in HEK293T cells led to cell deformation or death
(Supplementary Fig. 1f). Moreover, this validation work is still in
progress. Efb was the rst protein we discovered that could inhibit
both NF-κB and MAPK, as well as inhibit S. aureus-induced expression
of pro-inammatory cytokines, including TNFα,IL-1β, IL-6, and IL-12,
in vitro and in vivo, respectively. And we determined that the Efb
does not inuence the in vitro killing ability of isolated macrophages
(Supplementary Fig. 3df) or neutrophils (Supplementary Fig. 14)
in vitro. In the early stages of infection, increased levels of TNFαand
IL-1βare critical for eliminating bacterium in human S. aureus
bacteremia32.IL-1βhas also been shown to be a key cytokine for the
eradication of S. aureus in experimental models33. Our results indi-
cate that decreased bacterial burdens in lung tissues infected with
ΔEfb could be attributed to elevated TNF-αand IL-1βlevels,
which may lead to more neutrophil or other immune cell chemotaxis
or activation. Thus, inhibiting host pro-inammatory responses may
be one way by which Efb contributes to the pathogenicity of S.
aurues.
Although the post-translational modication system in bacteria is
very simple in comparison to eukaryotes34, accumulating studies have
shown that some bacteria effectors can be modied by the host post-
translational modication system29. Under certain conditions, some
bacteria effectors can disturb host post-translational modication35 to
facilitate infection. Our present study found that a host E3 ligase,
RNF114, ubiquitinates Efb of S. aureus to suppress host immunity. K27-
linkedubiquitination serves a varietyof functions, including enhancing
proteinprotein interactions, promoting proteasomal degradation,
and providing binding platforms for DNA repair proteins36. RNF114 has
been reported as a host E3 ligase with K27-linked polyubiquitination
activity with degradation effects in porcine and sea perch37,38.How-
ever, our results showed that K27-linked ubiquitination of Efb by
RNF114 did not lead to degradation of Efb but promoted the interac-
tion between Efb and TRAF3.
Efb has been identied as an immune evasion effector, and its
complement inhibitory effect depends on its C-terminal16. However,
our results demonstrate that replacing lysine 71 with arginine in the
N-terminal of Efb can eliminateits inhibitoryeffects in vivo and in vitro,
suggesting that the inhibitory effects of Efb are independent of its
complement effects in vivo. On the other hand, the immunosuppres-
sive abilities of Efb were shown to be correlated with inhibiting the
Fig. 4 | Efb stabilizes TRAF3. a Immunoblots of WCL from MH-S infected with
Newman andΔEfb (MOI = 25). bImmunoblots of WCL and IP products of WCL from
HEK293T cells transfected with indicated plasmids. Ub, wild-type ubiquitin; K48,
Ub with a single 48 lysine residue left; K63 Ub with a single 63 lysine residue left.
cImmunoblots of IP products of WCL from MH-S infected with Newman and ΔEfb
(MOI = 25). dImmunoblots of WCL and IPproducts from HEK293T cellstransfected
with indicated plasmids. eImmunoblots of IP products from MH-S infected with
Newman and ΔEfb (MOI = 25). fEfb inhibits K48-linked ubiquitin chains on TRAF3
in vitro. gImmunoblots of WCL and IP products from HEK293T cells transfected
with indicated plasmids. K/R, replace all lysine of TRAF3 with arginine; R106K,
replace 106 arginine of K/R with lysine; R155K, replace 155 arginine of K/R with
lysine. R106/155K, replace 106 and 155 arginine of K/R with lysine. Data are repre-
sentative of three experiments withat least three independent biological replicates.
Source data are provided as a Source Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved
formation of platelet-leukocyte complexes and binding brinogen to
prevent neutrophil activation, which depends on the N-terminal of
Efb17. However, lysine 71 of Efb was not the key residue in the inter-
action between Efb and brinogen according to a previous report39.
Platelets have been reported to play an important role in
inammation40. However, there is no detailed information as to which
amino acid of Efb plays a key role in the interaction between the Efb
N-terminal and platelets. Our results demonstrate that the direct
inhibitory effect of Efb on pro-inammatory cytokines was accom-
plished through interaction with TRAF3 in myeloid cells, including
mature macrophages, monocytes, and granulocytes. The anti-
inammatory effects of Efb are critical to the in vivo change in S.
aureus load and pathogenicity in mice. However, it is still possible that
our in vivo results were partly due to the interaction between Efb and
platelets or other unknown mechanisms.
TRAF3 is a tri-faced immune regulator that has distinct roles
depending on its targeted receptors, even within the same cell, and is
also highly cell-type-dependent20. TRAF3 has been reported to posi-
tively regulate type I interferon production while negatively regulating
NF-κB and MAPK pathways22. Our results demonstrate that TRAF3
regulates S. aureus-induced activation of NF-κBandMAPKpathwaysin
macrophages. S. aureus infection induces the degradation of TRAF3 in
macrophages. However, Efb can inhibit degradation by disturbing the
K48-linked ubiquitination of TRAF3 in HEK293T cells, PMs, and
alveolar macrophages (MH-S). K48-linked ubiquitination of TRAF3 for
degradation is accomplished by cIAP1/2, which requires the bridging
effects of TRAF2. Our dataindicate thatthe interaction between TRAF3
and TRAF2 was disturbed by Efb, and the inhibition on K48-linked
ubiquitination of TRAF3 by Efb was accomplished by disrupting the
TRAF3-TRAF2-cIAP1/2 complex.
As stated in previous reports, TLR2 and MyD88 in innate
immune cells play a key role in detecting S. aureus4143. We demon-
strated that TLR2 and MyD88 were critical in promoting pro-
inammatory responses of macrophages infected by S. aureus
(Supplementary Fig. 15a). As stated in a report by Perkins et al.44,
macrophages stimulated by P3C, a TLR2 ligand, overnight (24 h)
upregulated TRAF3 protein expression. However, their results also
showed that the TRAF3 mRNA levels were not signicantly increased
until 8 h post treatment. In the present study, we measured TRAF3
protein expression immediately after S. aureus infection using wild-
type, TLR2/, and MyD88/macrophages. We found that the
decrease in TRAF3 protein expression was related to the TLR2-
MyD88 pathway (Supplementary Fig. 15b).
The emergence of multi-drug resistant strains and the lack of a
vaccines has made S. aureus a global concern. Immune evasion has
been proven to be the main pathogeneses for S. aureus infection9,
and is also one of the main challenges for vaccine development45.We
identied that K27-linked ubiquitination of Efb by RNF114 binding to
TRAF3 disrupts the formation of the TRAF3/TRAF2/cIAP1 complex
and prevents K48-ubiquitination-mediated TRAF3 degradation,
resulting in suppressed pro-inammatory cytokine production
in vivo and in vitro. The present ndings identify a previously
unrecognized mechanism that S. aureus uses to suppress host
immunity (Supplementary Fig. 16). In addition, the Efb-RNF114
interface may be a promising target for the development of effec-
tive anti-S. aureus treatments.
Fig. 5 | Efb is K27-ubiquitinated at K71 by host RNF114. a Immunoblots of IP
products of WCL from MH-S infected with ΔEfb + Flag Efb for indicated times
(MOI = 2 5). b,cImmunoblots of WCLand IP products from WCL of HEK 293 T cells
transfected with indicated plasmids. dImmunoblots of IP products of WCL from
MH-S infected with ΔEfb + Flag Efb for indicated times (MOI = 25). eImmunoblots
of WCL and IP products from HEK293T cells transfected with indicated plasmids.
fImmunoblots of WCL and IP products from MH-S or RNF114 knockdown (KD)
MH-S infected with ΔEfb + Flag Efb for indicated times (MOI = 25).
gImmunoblots of WCL and IP products from HEK293T cells transfected with
indicated plasmids. K71R, replace 71 lysine of Efb wi th arginine. hImmunoblots
of WCL and IP products from MH-S infected with ΔEfb + Flag Efb or ΔEfb +
Flag Efb K71R for indicated times (MOI = 25). Data are representative of three
experiments with at least three independent biological replicates. Source data
are provided as a Source Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Methods
Bacterial strains and cells
Bacterial strains adopted in the present study are described in Sup-
plementary Table 1. The Escherichia coli strains DH5αand BL21 (Tian-
gen Biotech, China) were grown in LB medium. When required, the
antibiotics ampicillin (100 μg/ml) (Sanggon Biotech) or kanamycin
(50 μg/ml) (Sanggon Biotech) were used for the E. coli strain selection.
The S. aureus strains were grown in trypticase soy broth (OXOID,
CM0129B). Chloramphenicol (25 μg/ml) (Sanggon Biotech) was used
for the selection of S. aureus strains when required. Unless noted, all
bacteria were grown at 37°C in a shaking incubator at 200 rpm in
tubes kept at a 45° angle.
HEK293T cells (CRL-3216), obtained from the American Type
Culture Collection, were maintained in Dulbeccosmodied Eagles
medium (DMEM; HyClone) supplemented with 10% (v/v) fetal bovine
serum (FBS, HyClone) and 100 U/ml penicillin and streptomycin
(HyClone). MH-S cells (CRL-2019), obtained from the American Type
CultureCollection, were maintained in Roswell Park MemorialInstitute
(RPMI)1640 medium (HyClone) supplemented with 10% FBS. Perito-
neal macrophages (PMs) were obtained from 6-week-old wild-type or
mutant C57BL/6J mice as follows: mutant mice and their wild-type
littermates were injected with 2 ml Thioglycolate Broth (4%) intraper-
itoneally. Three days later, the peritoneal lavage uid was collected
from the mice and washed with PBS three times. PMs were grown in
DMEM supplemented with 10% FBS. Neutrophils were isolated from
mice blood using anti-Ly6G MicroBeads (Miltenyi, 130-120-337) and
cultured in RPMI-1640 medium (HyClone) containing 10% FBS.
Plasmids, reagents, and antibodies
Expressing plasmids were constructed by inserting a synthetic gene
segment in the vector, the names of which are listed in Supple-
mentary Table 1. The following antibodies were used for western
blot, immunoprecipitation, or immunouorescence assays: rabbit
anti-TRAF3 (PA5-20165, Invitrogen; ab36988, Abcam), mouse anti-
TRAF3 (sc6933, Santa Cruz), rabbit anti-TRAF2 (4724, Cell signaling
technology, CST), mouse anti-Flag (F1804, Sigma-Aldrich), rabbit
anti-HA (3724, CST), rabbit anti-Myc (2040, CST), rabbit anti-
phospho-p65 (3033, CST), rabbit anti-phospho-p38 (9215, CST),
rabbit anti-phospho-Erk1/2 (9101,CST),rabbitanti-phospho-JNK
(4668, CST), rabbit anti-GFP (2956, CST), rabbit anti-K27 (ab181537,
Abcam), rabbit anti-K48 (8081, CST), rabbit anti-K63 (5621, CST),
rabbit anti-cIAP1 (ab2399, Abcam), rabbit anti-RNF114 (ab97303,
Fig. 6 | Efb inhibits host immunity depending on K27-ubiquitination. a qPCR
analysis of Tnf,Il1b,Il6,andIl12p40 mRNA from PMs infected with Newman,
ΔEfb, ΔEfb + Flag Efb, or ΔEfb + Flag Efb K71R for indicated time s (MOI = 2 5;
***P= 0.0001, ****P< 0.0001, ***P= 0.0004, in sequence, Tnf;
*P=0.0128,**P= 0.0034, 0.0034, in sequence, Il1b;**P=0.0024,***P= 0.0010,
0.0010, in sequence, Il6;***P= 0.0010, 0.0001, 0.0002, in sequence, Il12p40).
bELISA quantication of TNF-α, IL-1β, IL-6, and IL-12 levels in lung tissues
homogenized in 1 ml PBS, infected with Newman, ΔEfb, ΔEfb + Flag Efb, or
ΔEfb + Flag Efb K71R (2 × 108CFUs per mouse) for 24 h (****P<0.0001,
***P =0.0001, 0.0003, in sequence, TNF-α;****P< 0.0001, <0.0001,
*P= 0.0163, in sequence, IL-1β;**P= 0.0065, ****P< 0.0 001, *P= 0.0111, in
sequence, IL-6; ***P=0.0008, **P=0.0019, *P= 0.0105, in sequence, IL-12).
cQuantication of the bacterial CFUs in lung tissue homogenates obtained in
b(**P= 0.0079, 0.0013, 0.0065). d,eHistopathology of lung tissues was
assessed in H&E sections stained from mice infected for 24 h; scale bars, 1,000
μm(top)and200μm (bottom), the boxed areas at the top are enlarged below
(***P= 0.0001, 0.0007, **P= 0.0020). Students two-tailed unpaired t-test
(a,b) or two-tailed MannWhitney Utest (c) was used for statistical analysis.
Data are representative of three experiments with at least three independent
biological replicates. The bars show the mean and standard deviation of n=3
(a), n=9(b,c), and n=5(d) per group. Source data are provided as a Source
Data le.
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 8
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Abcam), Alexa Fluor Plus 488 conjugated goat anti-mouse IgG
(A32723, Invitrogen), Alexa Fluor Plus 555 conjugated goat anti-
mouse IgG (A32727, Invitrogen), Alexa Fluor Plus 647 conjugated
goat anti-rabbit IgG (A32733, Invitrogen); Anti-Efb was generatedby
immunization of rabbits with the protein of Efb, rabbit anti-GST
(CW0085M, Cwbio), rabbit anti-His (CW0083M Cwbio); rabbit anti-
GAPDH (G9545, Sigma-Aldrich), mouse anti-Flag M2 Afnity Gel
(A2220, Sigma-Aldrich), mouse anti-HA Magnetic Beads (88836,
Thermo Fisher), mouse anti-TRAF3 agarose beads (sc6933 AC,
Santa Cruz), and goat anti-rabbit IgG (5127, CST), goat anti-mouse
IgG (96714, CST). For western blot assays, all primary antibodies
weredilutedat1:1000andsecondaryantibodiesweredilutedata
1:5000. For immunouorescence, monoclonal mouse anti-Flag M2
antibody was used at 1:500 dilution and rabbit anti-TRAF3 at a 1:200
dilution. Corresponding Alexa Fluor Plus 488 labeled goat anti-
mouse IgG, Alexa Fluor Plus 555 labeled goat anti-mouse IgG or
Alexa Fluor Plus 647 labeled goat anti-rabbit IgG were used as at a
1:200 dilution. DAPI Stain Solution for nuclear strain was from
Sangon Biotech (E607303). For ow cytometry, rat anti-mouse
Ly6G PE (551461, BD), rat anti-mouse CD11b-FITC (557396, BD) were
used at a 1:100 dilution.
Construction of S. aureus strains
The pBT2 vector (provided by X. Rao, Army Medical University,
Chongqing) was used to generate the S. aureus Newman strain with a
deletion of the gene encoding Efb (ΔEfb) using an allelic replacement
strategy as previously described46. The primersfor the construction of
the Efb knockout vector are listed in Supplementary Table 2. The
deletion of Efb was conrmed by PCR and Sanger sequencing. The
pLI50 vector (provided by X. Rao, Army Medical University, Chongq-
ing) was used to complement the ΔEfb strain with the wild-type Efb or
Efb K71R gene driven by the original promotor of Efb. The expression
of Efb or its mutants in the supernatant of S. aureus was examined by
immunoblot assays.
Transfection and confocal microscopy
HEK293T cells were transiently transfected using PEI (23966-2; Poly-
sciences) according to instructions of the manufacturer. PMs were
transduced or transfected with adenovirus (Hanbio) or jetMSSENGER
(150-07, Polyplus). MH-S cells were transiently transfected using the
INVI DNA RNA Transfection Reagent (IV1216100, Invigentech). Con-
focal microscopy was performed as described previously46.MH-SCells
were xed with4% formaldehyde for 10 minat 25 °C, permeabilized for
30 min in PBScontaining 0.3% Triton X-100, and then blocked for 1 h at
4 °C in a blocking buffer (1% BSA in PBS). Next, the cells were incubated
with the indicated antibodies at 4 °C overnight and secondary anti-
bodies at room temperature for 1 h. After staining with DAPI, images
were obtained using a Zeiss LSM 780 confocal laser microscopy
system.
Immunoelectronmicroscopy
After 4h infection with Newman or ΔEfb strains, MH-S cells were xed
in immunoelectronmicroscopy xative (Wanwu; G1124) for 2 h at 4 °C.
Ultrathin cryosections (70 nm) were prepared as previously
described47, and sections were sequentially labeled with rabbit anti-
Flag antibody, followed by sheep anti-rabbit antibody coupled with
10 nm gold particles. The stained sections were observed under a
Hitachi electron microscope HT7800.
Luciferase assay
HEK293T cells were transiently transfected with pNF-κBluc, pRLTK,
and the indicated plasmids for 24 h. After TNF-α(210-TA, R&D) sti-
mulation for 6 h, the dual-luciferase reporter assay system (RG028,
Beyotime) was used to detect luciferase activity according to the
instruction of the manufacturer.
Immunoprecipitation and immunoblot assays
Immunoprecipitation and immunoblot assays were performed as
previously described. Briey, HEK293T cells were transiently trans-
fected with plasmids using PEI. After 48 h, culture supernatants were
removed at corresponding times, and the cells were washed three
times with PBS. Cells were lysed in cell lysis buffer (Beyotime) sup-
plemented with 1% protease inhibitor cocktail (4693116001, Roche).
After centrifugation, the supernatants of cell lysates were incubated
with indicated gels at 4 °C overnight. For endogenous immunopreci-
pitations, PMs and MH-S were infected with S. aureus for the indicated
times.The cell lysates were subsequently incubated with indicated gels
at 4 °C overnight. After being centrifuged, the gels were then washed
three times with cell lysis buffer and boiled with SDS loading buffer.
Equivalent amounts of total proteins were separated by SDS-PAGE and
electro-blotted onto PVDF membranes. The membranes were then
probed with antibodies, followed by exposure conducted using an ECL
reagent (32209 or 34095, Thermo Fisher).
Ubiquitination assay
HA-TRAF3 was puried from HEK293T cells using an anti-Flag M2
Afnity Gel. The beads were washed three times with cell lysis buffer.
Beads were incubated in E3 ligase buffer, which contained Flag-TRAF2
and Flag-cIAP1 overexpressing cell lysate, ubiquitin, Mg-ATP, with or
without E1, E2, or Flag-Efb overexpressing cell lysate. All samples were
incubated at 37 °C for 1h in a metal bath by gently shaking. After
removing the supernatants, 50 μl2×SDS-PAGEgelloadingbufferwas
added to the obtained beads followed by heating to 95 °C for 5 min
prior to western blot analysis.
Real-time PCR analysis
RNA preparation and qPCR analysis were performed as described
previously30 using gene-specic primers (Supplementary Table 2).
Total RNA of cells was isolated using RNAiso Plus (9109; Takara). Next,
RNA (1 µg) was reverse-transcribed using the PrimeScriptRT Reagent
Kit (RR037; Takara) to generate cDNA. SYBR RT-PCR Kit (QPK-212;
Toyobo) were used for the quantitative real-time RT-PCR analysis.
Gene amplication was performed using the ΔΔCt method, and gene
expression was normalized to that of GAPDH.
Mice infection model
Six-week-old female specic-pathogen-free C57BL/6 mice were pur-
chased from Beijing HFK Bioscience CO., LTD. Traf3[ox/ox] and
Traf3[ox/ox, Lyz2-Cre] mice were purchased from Cyagen Biosciences.
They were housed under 12 light/12 dark cycles, ~1823 °C, 4060%
humidity and specic-pathogen-free (SPF) conditions at the National
Engineering Research Center of Immunological Products. All animal
experiments were reviewed and approved by the Animal Experiment
Administration Committee of Army Medical University and were
conducted in accordance with governmental guidelines and institu-
tional policies for the Care and Use of Laboratory Animals. Six-week-
old C57BL/6 mice were divided randomly into cages and infected by
intratracheal administration with 2 × 108CFUs or 6 × 108CFUs of dif-
ferent S. aureus strains in 20 μl PBS for 24 h or a few days. After the
mice were sacriced at the indicated times, the lungs were collected
and homogenized in 1 ml of PBS for ELISA and CFU assays. S. aureus
burden was determined by plating serial dilutions of each tissue
homogenate on tryptic soy agar (TSA) plates, which were incubated
at 37 °C. Colonies were counted after 12 h of incubation. For histo-
logical analysis, lungs were removed and xed in 4% paraformalde-
hyde in PBS and embedded in parafn. Sections were cut and stained
with hematoxylin and eosin (H&E) according to standard protocols.
Imaging was performed using microscopy (ECLIPSE 80i, Nikon). For
skin and blood infection models, the doses of S. aureus were 2 × 108
and 1 × 108CFUs. All mice were age- and sex-matched in each
experiment. The sample size was determined based on data from
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved
pilot experiments. For in vitro S. aureus infections, macrophages
were infected with a single-cell suspension of bacteria at an moieties
of infection (MOI) of 25.
Statistics and reproducibility
Data are expressed as mean ± s tandard deviation (SD). GraphPad Prism
8 was used for statistical analysis. The sample sizes, reproducibility of
experiments and the statistical tests used are presented in the gure
legends. ZEN 2.1 on Zeiss LSM 780 confocal laser microscopy system
was used for immunohistochemistry data collection and analysis. CFX
Manager Software v3.0 on BioRad CFX96Touch was used for qRT-PCR
data collection and analysis. Bio-Rad ChemDoc Touch was used for
western blot data collection and analysis. BD biosciences FACSDiva
software on FACSCanto was used for Flow cytometry data collection
and analysis.
Reporting summary
Further information on research design is available in the Nature
Research Reporting Summary linked to this article.
Data availability
All data are available within the present article and Supplementary
Information, or in the Source Data les. Source data are provided with
this paper.
References
1. Wardenburg, J. B., Schneewind, O., Bae, T., Otto, M. & De Leo, F.
Vaccine protection against Staphylococcus aureus pneumonia.
Crit. Care Med. 35S,A48(2007).
2. Aliberti, S. et al. Global initiative for meticillin-resistant Staphylo-
coccus aureus pneumonia (GLIMP): an international, observational
cohort study. Lancet Infect. Dis. 16,1364(2016).
3. Lee, A. S. et al., Methicillin-resistant Staphylococcus aureus.Nat.
Rev. Dis. Primers 4, 18033 (2018).
4. Krismer, B., Weidenmaier, C., Zipperer, A. & Peschel, A. The com-
mensal lifestyle of Staphylococcus aureus and its interactions with
the nasal microbiota. Nat. Rev. Microbiol. 15, 675 (2017).
5. Joshi, N., Walter, J. M. & Misharin, A. V. Alveolar macrophages. Cell
Immunol. 330,86(2018).
6. Wang, L. et al.Oxidization of TGF beta-activated kinase by MPT53 is
required for immunity to Mycobacterium tuberculosis.Nat. Micro-
biol. 4,1378(2019).
7. Pozzi, C. et al. Phagocyte subsets and lymphocyte clonal deletion
behind ineffective immune response to Staphylococcus aureus.
FEMS Microbiol. Rev. 39, 750 (2015).
8. Lakhundi, S. & Zhang, K. Methicillin-resistant Staphylococcus aur-
eus: molecular characterization, evolution, and epidemiology. Clin.
Microbiol. Rev. 31, e00020 (2018).
9. Cole, J., Aberdein, J., Jubrail, J. & Dockrell, D. H. In Academic Press,
Advances in Microbial Physiology, Vol 65 (ed Poole, R. K.) p.
125 (2014).
10. Grayczyk, J. P., Harvey, C. J., Laczkovich, I. & Alonzo, F. I. A lipoy-
lated metabolic protein released by Staphylococcus aureus sup-
presses macrophage activation. Cell Host Microbe 22, 678 (2017).
11. Yokoyama, R. et al. Staphylococcal superantigen-like Protein 3
binds to the toll-like receptor 2 extracellular domain and inhibits
cytokine production induced by Staphylococcus aureus, cell wall
component, or lipopeptides in murine macrophages. Infect.
Immun. 80,2816(2012).
12. Assis, L. M., Nedeljkovic, M. & Dessen, A. New strategies for tar-
geting and treatment of multi-drug resistant Staphylococcus aur-
eus.Drug Resist. Update.31,1(2017).
13. Schubert, K. A., Xu, Y., Shao, F. & Auerbuch, V. The Yersinia type III
secretion system as a tool for studying cytosolic innate immune
surveillance. Annu. Rev. Microbiol. 74, 221 (2020).
14. Wang, J. et al. Mycobacterium tuberculosis suppresses innate
immunity by coopting the host ubiquitin system. Nat. Immunol. 16,
189 (2015).
15. Raymond, B. et al. Subversion of trafcking, apoptosis, and innate
immunity by type III secretion system effectors. Trends Microbiol.
21, 430 (2013).
16. Hammel, M. et al. A structural basis for complement inhibition by
Staphylococcus aureus.Nat. Immunol. 8, 430 (2007).
17. Posner, M. G. et al. Extracellular brinogen-binding protein (Efb)
from Staphylococcus aureus Inhibits the formation of platelet-
leukocyte complexes. J. Biol. Chem. 291,2764(2016).
18. Jongerius, I. et al., Staphylococcus aureus virulence is enhanced by
secreted factors that block innate immune defenses. J. Innate
Immun. 4, 301 (2012).
19. Nandi, A. et al. Differential induction of inammatory cytokines and
reactive oxygen species in murine peritoneal macrophages and
resident fresh bone marrow cells by acute Staphylococcus aureus
infection: contribution of Toll-Like Receptor 2 (TLR2). Inammation
38, 224 (2015).
20. Yi,Z.,Lin,W.W.,Stunz,L.L.&Bishop,G.A.RolesforTNF-receptor
associated factor 3 (TRAF3) in lymphocyte functions. Cytokine
Growth Factor Rev. 25, 147 (2014).
21. Mihaly, S. R., Ninomiya-Tsuji, J. & Morioka, S. TAK1 control of cell
death. Cell Death Differ. 21,1667(2014).
22. Lalani, A. I., Luo, C., Han, Y. & Xie, P. TRAF3: a novel tumor sup-
pressor gene in macrophages. Macrophage 2, e1009 (2015).
23. Lalani, A. I. et al. Myeloid cell TRAF3 regulates immune responses
and inhibits inammation and tumor development in mice. J.
Immunol. 194, 334 (2015).
24. Gupta, P. et al. Leishmania donovani targets tumor necrosis factor
receptor-associated factor (TRAF) 3 for impairing TLR4-mediated
host response. FASEB J. 28,1756(2014).
25. Zhou, W. et al. Hypothermic oxygenated perfusion inhibits HECTD3-
mediated TRAF3 polyubiquitination to alleviate DCD liver ischemia-
reperfusion injury. Cell Death Dis. 12, 211 (2021).
26. Vallabhapurapu, S. et al. Nonredundant and complementary func-
tions of TRAF2 and TRAF3 in a ubiquitination cascade that activates
NIK-dependent alternative NF-kappaB signaling. Nat. Immunol. 9,
1364 (2008).
27. Zarnegar, B. J. et al. Noncanonical NF-kappa B activation requires
coordinated assembly of a regulatory complex of the adaptors
cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase NIK. Nat. Immunol. 9,
1371 (2008).
28. Tseng, P. et al. Different modes of ubiquitination of the adaptor
TRAF3 selectively activate the expression of type I interferons and
proinammatory cytokines. Nat. Immunol. 11,70(2010).
29. Wang, L., Wu, J., Li, J., Yang, H. & Ge, B. Host-mediated ubiquiti-
nation of a mycobacterial protein suppresses immunity. Nature 577,
682 (2020).
30. Yan,D.,Wang,X.,Luo,L.,Cao,X.&Ge,B.InhibitionofTLRsignaling
by a bacterial protein containing immunoreceptor tyrosine-based
inhibitory motifs. Nat. Immunol. 13,1063(2012).
31. Zhao, Y. et al. RNF114 suppresses metastasis through regulation of
PARP10 in cervical cancer cells. Cancer Commun. 41, 187 (2021).
32. Miller,L.S.Jr.,Fowler,V.G.,Shukla,S.K.,Rose,W.E.&Proctor,R.A.
Development of a vaccine against Staphylococcus aureus invasive
infections: evidence based on human immunity, genetics and
bacterial evasion mechanisms. FEMS Microbiol. Rev. 44,123(2020).
33. Giai, C. et al. Staphylococcus aureus induces shedding of IL-1RII in
monocytes and neutrophils. J. Innate Immun. 8, 284 (2016).
34. Macek, B. et al. Protein post-translational modications in bacteria.
Nat. Rev. Microbiol. 17,651(2019).
35. Ding, J. et al. Structural and functional insights into host death
domains inactivation by the bacterial arginine GlcNAcyltransferase
effector. Mol Cell 74,922(2019).
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 10
Content courtesy of Springer Nature, terms of use apply. Rights reserved
36. Tracz, M. & Bialek, W., Beyond K48 and K63: non-canonical protein
ubiquitination. Cell. Mol. Biol. Lett. 26,1(2021).
37. Zhang, Y., Zhang, H., Zheng, G. L., Yang, Q. & Qiu, H. J. Porcine RING
nger protein 114 inhibits classical swine fever virus replication via
the K27-linked polyubiquitination of viral NS4B. J. Virol. 93,
e1219 (2019).
38. Xiang, Y. et al. E3 ubiquitin ligase RNF114 inhibits innate immune
response to red-spotted grouper nervous necrosis virus infection in
sea perch by targeting MAVS and TRAF3 to mediate their degra-
dation. J. Immunol. 206,77(2021).
39. Ko, Y., Liang, X., Smith, C. W., Degen, J. L. & Hoeoek, M. Binding of
Efb from Staphylococcus aureus to brinogen blocks neutrophil
adherence. J. Biol. Chem. 286,9865(2011).
40. Sreeramkumar, V. et al. Neutrophils scan for activated platelets to
initiate inammation. Science 346, 1234 (2014).
41. Schmaler, M. et al. Lipoproteins in Staphylococcus aureus mediate
inammation by TLR2 and iron-dependent growth in vivo. J.
Immunol. 182, 7110 (2009).
42. Wang, X., Eagen, W. J. & Lee, J. C. Orchestration of human macro-
phage NLRP3 inammasome activation by Staphylococcus aureus
extracellular vesicles. Proc.NatlAcad.Sci.USA117, 3174 (2020).
43. Nakagawa, S. et al. Staphylococcus aureus Virulent PSMalpha
peptides induce keratinocyte alarmin release to orchestrate IL-17-
dependent skin inammation. Cell Host Microbe 22,667(2017).
44. Perkins, D. J. et al. Reprogramming of murine macrophages through
TLR2 confers viral resistance via TRAF3-mediated, enhanced inter-
feron production. PLoS Pathog. 9, e1003479 (2013).
45. Miller,L.S.Jr.,Fowler,V.G.,Shukla,S.K.,Rose,W.E.&Proctor,R.A.
Development of a vaccine against Staphylococcus aureus invasive
infections: Evidence based on human immunity, genetics and
bacterial evasion mechanisms. FEMS Microbiol. Rev. 44,123(2020).
46. Shang, W. et al. beta-Lactam antibiotics enhance the pathogenicity
of methicillin-resistant Staphylococcus aureus via SarA-controlled
lipoprotein-like cluster expression. mBio 10, e00880 (2019).
47. Mestre,M.B.,Fader,C.M.,Sola,C.&Colombo,M.I.Alpha-
hemolysin is required for the activation of the autophagic pathway
in Staphylococcus aureus-infected cells. Autophagy 6, 110 (2010).
Acknowledgements
We thank Xiancai Rao (Third Military Medical University, China) for pro-
viding the pBT2 and pLI50 vectors and S. aureus Newman strain; Lixin
Zheng (LISB/NIAID/NIH, USA) for critical reading of the manuscript; and
Weilong Shang (Third Military Medical University, China), Yi Yang (Third
Military Medical University, China), and Xianzhi Meng (Southwest Uni-
versity, China) for technical assistance. This work was supported by
grants from the National Natural Science Foundation of China (Nos.
81902036, 31970138) and the National Natural Science Foundation of
Chongqing (cstc2019jcyj-msxmX0377).
Author contributions
Q.Z., D.Y., H.Z., and X.Z. designed the experiments. X.Z. and T.X. wrote
the manuscript. X.Z., T.X., L.G., Y.S., J.Z., and Z.Z. analyzed experimental
results. X.Z., T.X., L.G., Y.W., L.L., and T.T. carried out the experiments.
D.L., P.L., W.Z., P.C., H.J., and Q.G. provided technical help. All authors
discussed the results and commented on the manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Supplementary information The online version contains
supplementary material available at
https://doi.org/10.1038/s41467-022-33205-z.
Correspondence and requests for materials should be addressed to
Hao Zeng, Dapeng Yan or Quanming Zou.
Peer review information Nature Communications thanks Friedrich Götz
and the other anonymous reviewer(s) for their contribution to the peer
review of this work. Peer review reports are available.
Reprints and permission information is available at
http://www.nature.com/reprints
Publishers note Springer Nature remains neutral with regard to jur-
isdictional claims in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons license, and indicate if
changes were made. The images or other third party material in this
article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not
included in the articles Creative Commons license and your intended
use is not permitted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from the copyright
holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
© The Author(s) 2022
Article https://doi.org/10.1038/s41467-022-33205-z
Nature Communications | (2022) 13:5493 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Several studies have revealed that STING serves as a dominant regulatory molecule during S. aureus infection, exerting conflicting effects in different infection models (10,11). Our previous study found that tumor necrosis factor α (TNF-α), interleukin (IL)-1β, IL-6, and IL-12 secreted by macrophages hinder S. aureus colonization in the early stages of infection (12). This study found that STING is also involved in this process. ...
... EsxB Interacts with STING. Secretory effectors released by S. aureus can manipulate the activation of macrophages by interfering with intracellular signal transduction (12). We speculated that the STING-mediated proinflammatory responses of macrophages may counteract S. aureus infection. ...
... The STING pathway is a critical pathway that plays a vital role in regulating the inflammatory responses of macrophages during microbial infections (5). Several studies have identified STING as a dominant regulatory molecule during S. aureus infection (10,12). However, the role of STING in S. aureus infection remains contro versial. ...
Article
Full-text available
Staphylococcus aureus ( S. aureus ) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING’s palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.
... As mentioned, S. aureus Efb also interacts with Fg and belongs to SERAMs (9,23,27,34). It is reported to inhibit complement activation by engaging C3b (35)(36)(37)(38), block platelet aggregation and their interaction with leukocytes (39,40) and interact with immune cells blocking cellular-mediated immunity (23,27,41,42). Furthermore, Efb can also bind to Complement Receptor 2 on B cells, further tackling adaptive responses of the host (43). ...
Article
Full-text available
Staphylococcus aureus pathology is caused by a plethora of virulence factors able to combat multiple host defence mechanisms. Fibrinogen (Fg), a critical component in the host coagulation cascade, plays an important role in the pathogenesis of this bacterium, as it is the target of numerous staphylococcal virulence proteins. Amongst its secreted virulence factors, coagulase (Coa) and Extracellular fibrinogen-binding protein (Efb) share common Fg binding motives and have been described to form a Fg shield around staphylococcal cells, thereby allowing efficient bacterial spreading, phagocytosis escape and evasion of host immune system responses. Targeting these proteins with monoclonal antibodies thus represents a new therapeutic option against S. aureus. To this end, here we report the selection and characterization of fully human, sequence-defined, monoclonal antibodies selected against the C-terminal of coagulase. Given the functional homology between Coa and Efb, we also investigated if the generated antibodies bound the two virulence factors. Thirteen unique antibodies were isolated from naïve antibodies gene libraries by antibody phage display. As anticipated, most of the selected antibodies showed cross-recognition of these two proteins and among them, four were able to block the interaction between Coa/Efb and Fg. Furthermore, our monoclonal antibodies could interact with the two main Fg binding repeats present at the C-terminal of Coa and distinguish them, suggesting the presence of two functionally different Fg-binding epitopes.
... ECP, also referred to as nonclassical secretion or protein moonlighting, has important implications for how bacterial cells interact with their external environment. Indeed, some of these excreted proteins have been reported to mediate host-pathogen interactions (24), biofilm formation (23), suppress macrophage activation (25), and are linked to survival and pathogenicity (26). Yet, the mechanisms of ECP are poorly understood (27), and it was previously unknown how mscL and arfA collectively mediate ECP in response to stress. ...
Article
Full-text available
Excretion of cytoplasmic protein (ECP) is a commonly observed phenomenon in bacteria, and this partial extracellular localisation of the intracellular proteome has been implicated in a variety of stress response mechanisms. In response to hypoosmotic shock and ribosome stalling in Escherichia coli , ECP is dependent upon the presence of the large-conductance mechanosensitive channel and the alternative ribosome–rescue factor A gene products. However, it is not known if a mechanistic link exists between the corresponding genes and the respective stress response pathways. Here, we report that the corresponding mscL and arfA genes are commonly co-located on the genomes of Gammaproteobacteria and display overlap in their respective 3′ UTR and 3′ CDS. We show this unusual genomic arrangement permits an antisense RNA–mediated regulatory control between mscL and arfA , and this modulates MscL excretory activity in E. coli . These findings highlight a mechanistic link between osmotic, translational stress responses and ECP in E. coli , further elucidating the previously unknown regulatory function of arfA sRNA.
... Recently it was shown that macrophage-ingested S. aureus secretes Efb into the cytoplasm, suppresses the secretion of inflammatory cytokines, and subdues host immunity by engaging with tumor necrosis-associated factor 3 (TRAF3). Ubiquitination of Efb by host ubiquitin ligase is essential for Efb-TRAF3 interaction (Zhang et al., 2022). ...
Article
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Article
Full-text available
Ischemia-reperfusion injury (IRI) is an inevitable and serious clinical problem in donations after heart death (DCD) liver transplantation. Excessive sterile inflammation plays a fateful role in liver IRI. Hypothermic oxygenated perfusion (HOPE), as an emerging organ preservation technology, has a better preservation effect than cold storage (CS) for reducing liver IRI, in which regulating inflammation is one of the main mechanisms. HECTD3, a new E3 ubiquitin ligase, and TRAF3 have an essential role in inflammation. However, little is known about HECTD3 and TRAF3 in HOPE-regulated liver IRI. Here, we aimed to investigate the effects of HOPE on liver IRI in a DCD rat model and explore the roles of HECTD3 and TRAF3 in its pathogenesis. We found that HOPE significantly improved liver damage, including hepatocyte and liver sinusoidal endothelial cell injury, and reduced DCD liver inflammation. Mechanistically, both the DOC and HECT domains of HECTD3 directly interacted with TRAF3, and the catalytic Cys (C832) in the HECT domain promoted the K63-linked polyubiquitination of TRAF3 at Lys138. Further, the ubiquitinated TRAF3 at Lys138 increased oxidative stress and activated the NF-κB inflammation pathway to induce liver IRI in BRL-3A cells under hypoxia/reoxygenation conditions. Finally, we confirmed that the expression of HECTD3 and TRAF3 was obviously increased in human DCD liver transplantation specimens. Overall, these findings demonstrated that HOPE can protect against DCD liver transplantation-induced-liver IRI by reducing inflammation via HECTD3-mediated TRAF3 K63-linked polyubiquitination. Therefore, HOPE regulating the HECTD3/TRAF3 pathway is a novel target for improving IRI in DCD liver transplantation.
Article
Full-text available
Protein ubiquitination has become one of the most extensively studied post-translational modifications. Originally discovered as a critical element in highly regulated proteolysis, ubiquitination is now regarded as essential for many other cellular processes. This results from the unique features of ubiquitin (Ub) and its ability to form various homo- and heterotypic linkage types involving one of the seven different lysine residues or the free amino group located at its N-terminus. While K48- and K63-linked chains are broadly covered in the literature, the other types of chains assembled through K6, K11, K27, K29, and K33 residues deserve equal attention in the light of the latest discoveries. Here, we provide a concise summary of recent advances in the field of these poorly understood Ub linkages and their possible roles in vivo.
Article
Full-text available
RIG-I-like receptor (RLR)-mediated antiviral signaling is critical to trigger the immune response to virus infection; however, the antiviral responses are also tightly regulated to avoid uncontrolled production of type I IFN by various mechanisms, including ubiquitination. In this study, an E3 ubiquitin ligase ring finger protein 114 (RNF114) from sea perch (Lateolabrax japonicus) (LjRNF114) was identified as a suppressor of RLR signaling pathways during red-spotted grouper nervous necrosis virus (RGNNV) infection. RGNNV infection promoted the expression of LjRNF114. Overexpression of LjRNF114 enhanced RGNNV replication, whereas knockdown of LjRNF114 led to opposite effects. Type I IFN production induced by RGNNV was suppressed by LjRNF114, which is dependent on its ubiquitin ligase activity. Moreover, LjRNF114 inhibited IFN promoter activation induced by key signaling molecules in RLR signaling pathways. We observed the interactions between LjRNF114 and both sea perch mitochondrial antiviral signaling protein (MAVS) and TNFR-associated factor 3 (TRAF3). Domain mapping experiments indicated that the RING and ubiquitin interacting motif domains of LjRNF114 were required for its interaction with TRAF3 and MAVS. We found that LjRNF114 targeted MAVS and TRAF3 for K27- and K48-linked ubiquitination and degradation, resulting in the inhibition of IFN production. Taken together, our study reveals, to our knowledge, a novel mechanism that LjRNF114 targets and promotes K27- and K48-linked ubiquitination of MAVS and TRAF3 to negatively regulate the RLR signaling pathways, promoting viral infection.
Article
Full-text available
Mycobacterium tuberculosis is an intracellular pathogen that uses several strategies to interfere with the signalling functions of host immune molecules. Many other bacterial pathogens exploit the host ubiquitination system to promote pathogenesis1,2, but whether this same system modulates the ubiquitination of M. tuberculosis proteins is unknown. Here we report that the host E3 ubiquitin ligase ANAPC2—a core subunit of the anaphase-promoting complex/cyclosome—interacts with the mycobacterial protein Rv0222 and promotes the attachment of lysine-11-linked ubiquitin chains to lysine 76 of Rv0222 in order to suppress the expression of proinflammatory cytokines. Inhibition of ANAPC2 by specific short hairpin RNA abolishes the inhibitory effect of Rv0222 on proinflammatory responses. Moreover, mutation of the ubiquitination site on Rv0222 impairs the inhibition of proinflammatory cytokines by Rv0222 and reduces virulence during infection in mice. Mechanistically, lysine-11-linked ubiquitination of Rv0222 by ANAPC2 facilitates the recruitment of the protein tyrosine phosphatase SHP1 to the adaptor protein TRAF6, preventing the lysine-63-linked ubiquitination and activation of TRAF6. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.
Article
Full-text available
Invasive Staphylococcus aureus infections are a leading cause of morbidity and mortality in both hospital and community settings, especially with the widespread emergence of virulent and multi-drug resistant methicillin-resistant S. aureus strains. There is an urgent and unmet clinical need for non-antibiotic immune-based approaches to treat these infections as the increasing antibiotic resistance is creating a serious threat to public health. However, all vaccination attempts aimed at preventing S. aureus invasive infections have failed in human trials, especially all vaccines aimed at generating high titers of opsonic antibodies against S. aureus surface antigens to facilitate antibody-mediated bacterial clearance. In this review, we summarize the data from humans regarding the immune responses that protect against invasive S. aureus infections as well as host genetic factors and bacterial evasion mechanisms, which are important to consider for the future development of effective and successful vaccines and immunotherapies against invasive S. aureus infections in humans. The evidence presented form the basis for a hypothesis that staphylococcal toxins (including superantigens and pore-forming toxins) are important virulence factors, and targeting the neutralization of these toxins are more likely to provide a therapeutic benefit in contrast to prior vaccine attempts to generate antibodies to facilitate opsonophagocytosis.
Article
Full-text available
Over the past decade the number and variety of protein post-translational modifications that have been detected and characterized in bacteria have rapidly increased. Most post-translational protein modifications occur in a relatively low number of bacterial proteins in comparison with eukaryotic proteins, and most of the modified proteins carry low, substoichiometric levels of modification; therefore, their structural and functional analysis is particularly challenging. The number of modifying enzymes differs greatly among bacterial species, and the extent of the modified proteome strongly depends on environmental conditions. Nevertheless, evidence is rapidly accumulating that protein post-translational modifications have vital roles in various cellular processes such as protein synthesis and turnover, nitrogen metabolism, the cell cycle, dormancy, sporulation, spore germination, persistence and virulence. Further research of protein post-translational modifications will fill current gaps in the understanding of bacterial physiology and open new avenues for treatment of infectious diseases. Post-translational modifications of bacterial proteins have a role in various cellular processes such as protein synthesis and turnover, metabolism, the cell cycle, morphogenesis and virulence. In this Review, Macek and colleagues introduce the major types of protein post-translational modifications in bacteria and discuss their roles in various aspects of bacterial cell biology.
Article
Full-text available
In the host, many RING-domain E3 ligases have been reported to inhibit viral replication through various mechanisms. In a previous screen, we found that the porcine RING finger protein 114 (pRNF114), an RING-domain E3 ubiquitin ligase, inhibits classical swine fever virus (CSFV) replication. This study aimed to clarify the underlying antiviral mechanism of pRNF114 against CSFV. Upon CSFV infection, the pRNF114 mRNA was upregulated both in vitro and in vivo . CSFV replication was significantly suppressed in PK-pRNF114 cells stably expressing pRNF114 by lentivirus-delivered system, whereas CSFV growth was enhanced in PK-15 cells with RNF114 knockout by the CRISPR/Cas9 system. The RING domain of pRNF114, which has the E3 ubiquitin ligase activity, is crucial for its antiviral activity. Mechanistically, pRNF114 interacted with the CSFV NS4B protein through their C-terminal domains, which led to the K27-linked polyubiquitination and degradation of NS4B through a proteasome-dependent pathway. Collectively, these findings indicate that pRNF114 as a critical regulator of CSFV replication and uncover a mechanism by which pRNF114 employs its E3 ubiquitin ligase activity to inhibit CSFV replication. IMPORTANCE The porcine RING finger protein 114 (pRNF114) is a member of RING-domain E3 ligases. In this study, pRNF114 is a potential anti-CSFV factor and the anti-CSFV effect of pRNF114 depends on its E3 ligase activity. Notably, pRNF114 targets and catalyzes the K27-linked polyubiquitination of the NS4B protein and then promotes proteasome-dependent degradation of NS4B, inhibiting the replication of CSFV. To our knowledge, pRNF114 is the first E3 ligase to be identified as being involved in anti-CSFV activity and targeting NS4B could be a crucial route for antiviral development.
Article
Microbial pathogens have evolved complex mechanisms to interface with host cells in order to evade host defenses and replicate. However, mammalian innate immune receptors detect the presence of molecules unique to the microbial world or sense the activity of virulence factors, activating antimicrobial and inflammatory pathways. We focus on how studies of the major virulence factor of one group of microbial pathogens, the type III secretion system (T3SS) of human pathogenic Yersinia, have shed light on these important innate immune responses. Yersinia are largely extracellular pathogens, yet they insert T3SS cargo into target host cells that modulate the activity of cytosolic innate immune receptors. This review covers both the host pathways that detect the Yersinia T3SS and the effector proteins used by Yersinia to manipulate innate immune signaling. Expected final online publication date for the Annual Review of Microbiology, Volume 74 is September 8, 2020. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Release of extracellular vesicles (EVs) is a common feature among eukaryotes, archaea, and bacteria. However, the biogenesis and downstream biological effects of EVs released from gram-positive bacteria remain poorly characterized. Here, we report that EVs purified from a community-associated methicillin-resistant Staphylococcus aureus strain were internalized into human macrophages in vitro and that this process was blocked by inhibition of the dynamin-dependent endocytic pathway. Human macrophages responded to S. aureus EVs by TLR2 signaling and activation of NLRP3 inflammasomes through K ⁺ efflux, leading to the recruitment of ASC and activation of caspase-1. Cleavage of pro–interleukin (IL)-1β, pro-IL-18, and gasdermin-D by activated caspase-1 resulted in the cellular release of the mature cytokines IL-1β and IL-18 and induction of pyroptosis. Consistent with this result, a dose-dependent cytokine response was detected in the extracellular fluids of mice challenged intraperitoneally with S. aureus EVs. Pore-forming toxins associated with S. aureus EVs were critical for NLRP3-dependent caspase-1 activation of human macrophages, but not for TLR2 signaling. In contrast, EV-associated lipoproteins not only mediated TLR2 signaling to initiate the priming step of NLRP3 activation but also modulated EV biogenesis and the toxin content of EVs, resulting in alterations in IL-1β, IL-18, and caspase-1 activity. Collectively, our study describes mechanisms by which S. aureus EVs induce inflammasome activation and reveals an unexpected role of staphylococcal lipoproteins in EV biogenesis. EVs may serve as a novel secretory pathway for S. aureus to transport protected cargo in a concentrated form to host cells during infections to modulate cellular functions.