ArticlePDF AvailableLiterature Review

Immunocompetent and Immunodeficient Mouse Models for Enterovirus 71 Pathogenesis and Therapy

MDPI
Viruses
Authors:

Abstract and Figures

Enterovirus 71 (EV71) is a global health threat. Children infected with EV71 could develop hand-foot-and-mouth disease (HFMD), encephalitis, paralysis, pulmonary edema, and death. At present, no effective treatment for EV71 is available. We reviewed here various mouse models for EV71 pathogenesis and therapy. Earlier studies relied on the use of mouse-adapted EV71 strains. To avoid artificial mutations arising de novo during the serial passages, recent studies used EV71 clinical isolates without adaptation. Several human receptors for EV71 were shown to facilitate viral entry in cell culture. However, in vivo infection with human SCARB2 receptor transgenic mice appeared to be more limited to certain strains and genotypes of EV71. Efficacy of oral infection in these transgenic models is extremely low. Intriguingly, despite the lack of human receptors, immunodeficient neonatal mouse models can still be infected with EV71 clinical isolates via oral or intraperitoneal routes. Crossbreeding between SCARB2 transgenic and stat1 knockout mice generated a more sensitive and user-friendly hybrid mouse model. Infected hybrid mice developed a higher incidence and earlier onset of CNS disease and death. Different pathogenesis profiles were observed in models deficient in various arms of innate or humoral immunity. These models are being actively used for antiviral research.
Content may be subject to copyright.
viruses
Review
Immunocompetent and Immunodeficient Mouse
Models for Enterovirus 71 Pathogenesis and Therapy
Chiaho Shih 1, *, Chun-Che Liao 1, Ya-Shu Chang 1, Szu-Yao Wu 1, Chih-Shin Chang 1,2 and
An-Ting Liou 1
1
Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; jfliao@ibms.sinica.edu.tw (C.-C.L.);
daph7516@gmail.com (Y.-S.C.); guguma126@yahoo.com.tw (S.-Y.W.); sony721108@hotmail.com (C.-S.C.);
sh3333@msn.com (A.-T.L.)
2Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 11221, Taiwan
*Correspondence: cshih@ibms.sinica.edu.tw; Tel.: +886-2-2652-3996; Fax: +886-2-2652-3597
Received: 1 November 2018; Accepted: 26 November 2018; Published: 28 November 2018


Abstract:
Enterovirus 71 (EV71) is a global health threat. Children infected with EV71 could
develop hand-foot-and-mouth disease (HFMD), encephalitis, paralysis, pulmonary edema, and death.
At present, no effective treatment for EV71 is available. We reviewed here various mouse models
for EV71 pathogenesis and therapy. Earlier studies relied on the use of mouse-adapted EV71 strains.
To avoid artificial mutations arising de novo during the serial passages, recent studies used EV71
clinical isolates without adaptation. Several human receptors for EV71 were shown to facilitate
viral entry in cell culture. However,
in vivo
infection with human SCARB2 receptor transgenic mice
appeared to be more limited to certain strains and genotypes of EV71. Efficacy of oral infection
in these transgenic models is extremely low. Intriguingly, despite the lack of human receptors,
immunodeficient neonatal mouse models can still be infected with EV71 clinical isolates via oral or
intraperitoneal routes. Crossbreeding between SCARB2 transgenic and stat1 knockout mice generated
a more sensitive and user-friendly hybrid mouse model. Infected hybrid mice developed a higher
incidence and earlier onset of CNS disease and death. Different pathogenesis profiles were observed
in models deficient in various arms of innate or humoral immunity. These models are being actively
used for antiviral research.
Keywords: Enterovirus 71; EV71; EV-A71; animal models; pathogenesis; therapy
1. Introduction
Enterovirus 71 (EV71 or EV-A71) was first isolated from the stool of encephalitis patients in
California in 1969 [
1
]. It is a positive-strand RNA virus, belonging to the Picornaviridae family [
2
,
3
].
The genomic structure of EV71 is as outlined in Figure 1. Similar to poliovirus and other plus-strand
RNA viruses, it contains an IRES element, which drives the translation of a polyprotein from a 7.4 kb
genomic mRNA template. Upon protease cleavage, this polyprotein precursor can then convert into
various smaller-size structural and non-structural proteins, leading to capsid assembly. As the focus in
this review is about pathogenesis and animal models, readers interested in the EV71 replication cycle
are advised to consult other reviews in literature.
Viruses 2018,10, 674; doi:10.3390/v10120674 www.mdpi.com/journal/viruses
Viruses 2018,10, 674 2 of 20
Viruses 2018, 10, x FOR PEER REVIEW 2 of 18
Figure 1. The genomic organization of EV71. The plus-strand RNA of EV71 genome encodes a single
polyprotein precursor. Upon proteolytic processing by virus-encoded proteases 2A and 3C, this
polyprotein precursor converts into several smaller mature products, including 4 structural proteins
(VP1-VP4 capsid protein) and 7 non-structural proteins (2A, 2B, 2C, 3A, 3B, 3C and 3D). 3B encodes
a VPg protein, which is covalently bound to the 5 end of the RNA genome. 3D encodes RNA-
dependent RNA polymerase. IRES: internal ribosome entry site. This cartoon illustration is based on
gt B5 clinical isolates in Taiwan [4,5]. However, it is representative of the general genomic structure
of all genotypes isolated elsewhere.
Adults infected with EV71 could develop subclinical or cold-like syndrome. However, children
younger than five years of age could develop hand-foot-and-mouth disease (HFMD) and herpangina.
In severe cases, EV71-infected children could develop aseptic meningitis, encephalitis or myocarditis
[6]. In a long-term follow-up study, EV71-infected patients with CNS involvement and/or
cardiopulmonary failure was associated with delayed neurodevelopment and reduced cognitive
functioning [7]. Unfortunately, some patients were still not saved due to pulmonary edema and/or
heart failure [8].
Although EV71 was first discovered in the US decades ago, recent major outbreaks occurred
frequently in the Asia-Pacific region, including Taiwan, Singapore, China, Malaysia, and Japan [6,9
12]. China alone reported 1894 deaths between 2008 and 2011 [12]. Sporadic cases were also reported
from Europe [13,14], Russia [15] and Australia [16], indicating that EV71 is a global health threat. Like
poliovirus in the last century, EV71 is emerging rapidly as another pathogenic picornavirus for
children in this century.
Intravenous immunoglobulin (IVIG) is a therapeutic strategy for EV71-associcated brainstem
encephalitis [17]. However, an antibody-dependent enhancement (ADE) phenomenon was observed
in EV71-infected patients [1821], suggesting that IVIG may not be the most adequate treatment in
clinical practice. Milrinone, a phosphodiesterase inhibitor, was used to treat patients with EV-71
induced pulmonary edema (PE). While Milrinone is not an antiviral per se, mortality was
significantly decreased in the Milrinone-treated group, suggesting that it could be a useful
therapeutic drug for EV71-associated PE [22].
To investigate the pathogenesis and therapy for EV71 infection, we will need both in vitro and
in vivo models of EV71 infection. In this review, we cataloged here from the literature a total of 16
cell lines (Table 1) and 22 animal models (Table 2), which had been tested for EV71 infection and
pathogenesis. We compare below their respective advantages, limitations, and caveats in using these
models.
EV71 RNA ≈ 7.4 kb
Figure 1.
The genomic organization of EV71. The plus-strand RNA of EV71 genome encodes a
single polyprotein precursor. Upon proteolytic processing by virus-encoded proteases 2A and 3C,
this polyprotein precursor converts into several smaller mature products, including 4 structural proteins
(VP1-VP4 capsid protein) and 7 non-structural proteins (2A, 2B, 2C, 3A, 3B, 3C and 3D). 3B encodes a
VPg protein, which is covalently bound to the 5
0
end of the RNA genome. 3D encodes RNA-dependent
RNA polymerase. IRES: internal ribosome entry site. This cartoon illustration is based on gt B5 clinical
isolates in Taiwan [
4
,
5
]. However, it is representative of the general genomic structure of all genotypes
isolated elsewhere.
Adults infected with EV71 could develop subclinical or cold-like syndrome. However, children
younger than five years of age could develop hand-foot-and-mouth disease (HFMD) and herpangina.
In severe cases, EV71-infected children could develop aseptic meningitis,
encephalitis or myocarditis [6]
.
In a long-term follow-up study, EV71-infected patients with CNS involvement and/or cardiopulmonary
failure was associated with delayed neurodevelopment and reduced cognitive functioning [
7
].
Unfortunately, some patients were still not saved due to pulmonary edema and/or heart failure [8].
Although EV71 was first discovered in the US decades ago, recent major outbreaks occurred
frequently in the Asia-Pacific region, including Taiwan, Singapore, China, Malaysia, and Japan [
6
,
9
12
].
China alone reported 1894 deaths between 2008 and 2011 [
12
]. Sporadic cases were also reported
from Europe [
13
,
14
], Russia [
15
] and Australia [
16
], indicating that EV71 is a global health threat.
Like poliovirus in the last century, EV71 is emerging rapidly as another pathogenic picornavirus for
children in this century.
Intravenous immunoglobulin (IVIG) is a therapeutic strategy for EV71-associcated brainstem
encephalitis [17]. However, an antibody-dependent enhancement (ADE) phenomenon was observed
in EV71-infected patients [
18
21
], suggesting that IVIG may not be the most adequate treatment in
clinical practice. Milrinone, a phosphodiesterase inhibitor, was used to treat patients with EV-71
induced pulmonary edema (PE). While Milrinone is not an antiviral per se, mortality was significantly
decreased in the Milrinone-treated group, suggesting that it could be a useful therapeutic drug for
EV71-associated PE [22].
To investigate the pathogenesis and therapy for EV71 infection, we will need both
in vitro
and
in vivo
models of EV71 infection. In this review, we cataloged here from the literature a total of
16 cell lines (Table 1) and 22 animal models (Table 2), which had been tested for EV71 infection
and pathogenesis. We compare below their respective advantages, limitations, and caveats in using
these models.
Viruses 2018,10, 674 3 of 20
Table 1. Summary of 16 cell lines used for EV71 in vitro infection.
Cell Line Species Tissue Origin EV71 Genotype CPE #References
RD
Human
Rhabdomyosarcoma A, B, C, D +[2330]
SK-N-SH Neuroblastoma C2, D +[24,27]
SH-SY5Y Neuroblastoma C4 +[31,32]
NSC-34 motor neuron cell line B2, B4 & C2 [33]
SF268 glioblastoma * +[34]
Neuro-2A Mouse Neuroblastoma D +[24,35]
HT29
Human
intestinal epithelial cell B4, C +[25,26,28]
Caco-2 colorectal carcinoma B, C2 +[27,29]
C2BBel
mature intestinal epithelial cell (derived from Caco-2)
C+[28]
HeLa Human cervical carcinoma EV71:BS (Accession NO KF514878), C4 +[35,36]
HEK293T Human embryonic kidney EV71:BS (Accession NO KF514878), C4 +[35,37]
THP-1 Human monocytic cell line C2, C4 [19,37]
human PBMC C2 [19]
Vero Monkey African green monkey kidney A, EV71:BS (Accession NO KF514878) +[30,35]
COS-7 African green monkey kidney (SV40 transformed) EV71:BS (Accession NO KF514878) +[35]
NIH/3T3 Mouse mouse embryonic fibroblast mouse-adapted strain (EV:TLLm) +[35]
* information not available. #CPE, cytopathic effect. Genotype D is a C4 subgenotype.
Viruses 2018,10, 674 4 of 20
Table 2. Summary of 22 animal models used for EV71 in vivo infection.
Mouse Strain Host Age @Route Genotype Titer/Mouse Phenotype Virus-Replicating Tissue Reference
Balb/C
1-day-old i.p. B4 1×104PFU limb paralysis & death intestine [38]
2-day-old
i.p./i.c.
C4 103TCID50 * brain, liver and intestine [39]
5-day-old i.p. C4 (mouse adapted strain) 104TCID50 limb paralysis & death spleen, heart, liver, muscle and brain [40]
7-day-old
i.m.
i.p.
i.c.
B3 (CHO cell adapted strain;
CHO-26M) (mouse adapted strain;
MP-26M)
103TCID50 death
muscle (CHO-26M) blood, spleen,
heart, liver, muscle and brain
(MP-26M)
[41]
9-day-old i.p. C4 104.5 TCID50 limb paralysis
skeletal muscle, spinal cord, brainstem,
lung, and jejunum [24]
ICR
1-day-old oral C (mouse adapted strain) 106–107PFU paralysis & skin lesion skin, intestine and spinal cord [42]
1-day-old i.p. C4 (Fuyang-0805a) 2×105TCID50 none muscles, intestines, lungs, and brain [43]
7-day-old oral C (mouse adapted strain M2) 5×106PFU death * [44]
7-day-old i.p. C2 (mouse adapted strain MP4) 2.5 ×106PFU death brain, spinal cord, lung and muscle [21]
3-day-old i.p. C2 (clinical isolate) 4×106PFU death * [45]
7-day-old oral C2 (mouse adapted strain MP4) 5×105PFU death * [45]
12–14-day-old i.p. C (mouse adapted strain M2) 5×104–105PFU paralysis & death * [46]
14-day-old i.p. B3 (mouse adapted strain) 100 CCID50 paralysis brain and muscle [47]
14-day- &
28-day-old
i.p.
i.m.
oral
s.c.
B3 (mouse adapted strain) 105CCID50 paralysis mainly in CNS [48]
C57BL/6 9-day-old oral C (mouse adapted strain M2) 3×107PFU * * [44]
C57BL/6J 14-day-old i.p. C (mouse adapted strain M2) 3×105PFU hunched posture,
paralysis & death brain [49]
PSGL-1 Tg 10-day-old i.p. C4 (clinical isolate) 108TCID50 no disease onset brain, spinal cord and skeletal muscle [50]
C4 (mouse adapted strain MP10) 5×106TCID50 hindlimb paralysis *
Viruses 2018,10, 674 5 of 20
Table 2. Cont.
Mouse Strain Host Age @Route Genotype Titer/Mouse Phenotype Virus-Replicating Tissue Reference
hSCARB2 Tg 3-week-old
i.v. C (Isehara strain) 102–6 TCID50
ataxia, paralysis & death
CNS [51]
i.c. A, B and C 102–6 TCID50
i.p. C (Isehara strain) 103–6 TCID50
oral C (Isehara strain) 106–7 TCID50
pEF-1α-hSCARB2 1-day-old s.c. B 1×107PFU
HFMD-like syndrome &
paralysis CNS, intestine, muscle and skin [52]
7-day-old s.c. C 3×104PFU
AG129 younger than
2-week-old
i.p. B4 5×105–107PFU limb paralysis & death CNS [53]
oral 106–107PFU
AG129 10-week-old i.p. B2 (mouse adapted strain) 1.3 ×105
TCID50/mL limb paralysis & death ND [54]
A129
G129 7-day-old i.p. B5 (clinical isolates) 108PFU limb paralysis & death ND [4]
stat-1 KO 7-day-old i.p. B5 (clinical isolates) 108PFU limb paralysis & death CNS [4]
pSCARB2x
stat-1 KO
younger than
2-week-old i.p. C2 (clinical isolates) 106–108PFU [5]
NOD/SCID
3-day-old oral B5 (clinical isolates) 108PFU hair loss, skin lesion,
limb paralysis & death muscle and spleen [4]
7-day-old i.p. 107–108PFU
3–4 week-old i.v./i.c. B1 (mouse adapted strain) 106.5 CCID50 skeletal muscle and heart [55]
IP10 KO 14-day-old i.p. C (mouse adapted strain M2) 3×105PFU The death rate was
higher than C57BL/6J ND [49]
* information not available. @The age that mice infected with EV71.
Viruses 2018,10, 674 6 of 20
2. In Vitro Propagation Systems
The most common cell line for EV71 infection in culture is the human RD cell line of
rhabdomyosarcoma origin [
23
]. This RD cell line can produce a high yield of virus particles and thus
is often used for amplification of EV71 in animal experiments [
4
] and studies on viral structure [
56
].
In addition to RD cells, various cell lines used in poliovirus infection, such as HeLa and HEK293,
are also susceptible to EV71 infection and transfection [
36
,
37
]. These cell lines were used as a platform
to investigate the function of viral protein. From clinical samples, EV71 protein can be identified in
brainstem [
6
,
57
], and thus EV71 is considered as a neurotropic virus [
1
]. Several neuronal cell lines,
including SK-N-SH, NSC-34 and SF268, had been used in a number of studies, such as dissecting the
viral life cycle, host response to EV71 [33], phylogenetic analysis [24], and anti-viral drug testing [34].
In natural infection, EV71 is transmitted through an oral-fecal route [
58
]. Some previous studies
therefore focused on the GI tract immune responses to viral infection and replication kinetics in the
intestinal epithelial cells [25,26].
Using the Jurkat cell line and an expression cloning strategy by panning, Nishimura et al.
identified human P-selectin glycoprotein ligand-1 (PSGL-1; CD162) as a functional receptor on
leukocytes for EV71 viral entry [
59
]. However, PSGL-1 cannot be the only receptor for EV71,
since PSGL-1-independent infection in non-leukocytes was also observed. Yamayoshi et al. reported
that human scavenger receptor class B, member 2 (SCARB2, also known as lysosomal integral
membrane protein II or CD36b like-2) is another receptor for EV71 [
60
]. The exogenous expression of
human SCARB2 converted non-permissive cell lines to instead support EV71 propagation and develop
cytopathic effects. EV71 infection can be interfered by the antibody to SCARB2 or by soluble SCARB2.
Several alternative receptors for EV71 have also been reported [6164].
In addition to Jurkat cells, THP-1 and human PBMC had been used to study the innate immunity
in EV71 infection [
37
]. Many other cell lines contributed to the studies on EV71 molecular virology,
virus-host interactions, or drug screening (Table 1). So far, the human rhabdomyosarcoma cell
line RD appeared to be the most commonly used gold standard in literature, in part due to
its high yield production of different EV71 genotypes [
65
]. As more and more laboratories are
using the same RD cell line (e.g., from ATCC), it would be easier to cross-compare results from
different laboratories. Since these cell line models are limited one way or the other in their resemblance
to human natural infection, an alternative approach is needed to validate the findings from the cell
culture system. In this regard, animal modeling can provide more complete and convincing evidence
for our understanding on EV71 in vivo infection and pathogenesis.
3. In Vivo Animal Models
Experimental results learned from the cell culture system need to be validated in animal models
and human clinical trials. Table 2summarizes different EV71 mouse models reported in literature
so far. Overall, one thing in common in these different models is their frequent use of limb paralysis
and mortality for scoring disease severity. This is in part due to the fact that paralysis and death
are more apparent, objective, and convenient phenotypes than other parameters, such as ataxia,
or pulmonary edema. Another common feature is the requirement of a neonatal host age for
productive
in vivo
infection, in particular with clinical isolates. Although the exact mechanism for this
young age requirement remains unclear, it is technically demanding and not user-friendly, when the
newborns are too small and fragile to handle experimentally. When tissue cytokine/chemokine
profiles were examined in these different EV71 mouse models, they were often found to share
some inflammatory cytokines in common, such as IL1
β
, IL6, IL10, IP10, MCP-1, IFN-
α
, and IFN-
γ
,
etc. [
4
,
52
,
53
]. For neuropathogenesis models, the distribution of viral antigens is also quite similar
among different models, which mimics human cases with fatal encephalomyelitis. For example,
inflammation and viral antigens in human cases were observed mainly in the spinal cord, brainstem,
hypothalamus, cerebellar dentate nucleus, and cerebrum [
66
]. In the stat-1 KO model, VP1 was strongly
expressed throughout the entire spinal cord, including the anterior horn and the gray matter [
4
].
Viruses 2018,10, 674 7 of 20
Furthermore, VP1 in the pons of the brainstem, but not in the cerebral cortex, supports the concept
of retrograde axonal transport from the peripheral nerves to the spinal cord ascending to the CNS.
This result is supported from the mouse models infected with mouse-adapted EV71 strains [
67
,
68
] and
poliovirus [69].
As summarized above, there is a high degree of similarity among EV71-infected human patients
and mouse models. However, there are also important differences among these mouse models,
which will need to be discussed individually.
3.1. Mouse Models Dependent on Mouse-Adapted EV71 Strains
In general, adult humans are resistant to EV71 infection and pathogenesis. Similarly, adult mice
cannot be easily infected with EV71 clinical isolates. To solve this problem, researchers developed
mouse-adapted strains of EV71, such as MP4, in an ICR mouse model [
21
,
27
,
44
46
]. MP4 was generated
through i.p. injection of the parental EV71 TW/4643/98 into one-day-old ICR mice. Adapted EV71
recovered from the brain tissue was re-used to repeat the serial injection-adaptation cycles.
EV71 recovered from the fourth passage was designated as MP4 [
27
]. Comparing to its parental
EV71 isolates, MP4 is more cytotoxic with a larger plaque size. Because EV71 is a RNA virus
with a quasi-species nature, mouse-adapted strains tend to accumulate many artificial mutations
from the serial passages. One legitimate concern is that these adaptive mutations cannot really be
found in human natural infection. In addition to MP4, mouse adapted strains were also generated
in the immunodeficient NOD/SCID mouse model [
55
]. Comparison of the amino acid sequences
between clinical isolates and mouse adapted strains revealed major differences in structural proteins,
such as VP1,
VP3, and protease 2A [
55
]. Therefore, it always remains uncertain whether results
obtained from using the adapted strain of EV71 can be automatically extended to human natural
infection with the clinical isolates.
3.2. Immunodeficient Mouse Models
Host immune system plays a critical role against viral infection [
70
]. Immunodeficient mice,
even without bearing any human receptors for viral entry, can support infection with EV71
clinical isolates, instead of relying on mouse-adapted strains. Innate immunity, such as IFNs, is required
for protection from EV71 infection and pathogenesis [
71
,
72
]. AG129 mice, which is deficient in
both
α
/
β
and
γ
interferon receptors, developed limb paralysis and death, when infected with a
non-mouse-adapted EV71 via i.p. or oral routes. Viral protein was detected in CNS in both i.p.
and oral infection [
53
]. Like mouse AG129, mouse A129 (deficient in
α
/
β
interferon receptor) can
also be infected with mouse-adapted strain of EV71 [
54
]. Moreover, mouse G129 deficient in
γ
interferon receptor developed limb paralysis via an i.p. route with clinical isolates of EV71 [
4
] (Table 2).
Another host factor stat-1 is a key transcription factor in IFN signaling. The stat-1 knockout (KO) mice
can be successfully infected with both genotype B and C clinical isolates of EV71 [4,5]. Infected stat-1
KO mice developed hindlimb paralysis with abundant viral protein in the CNS system, supporting an
important role of IFN signaling in protecting CNS from EV71 infection and pathogenesis.
In addition to innate immunity, humoral immunity also plays an important role in EV71 infection
and pathogenesis. For example, injection with an EV71-specific neutralizing antibody before or
after infection, can strongly reduce lethality and tissue viral loads of a mouse-adapted strain M2
in a B cell-deficient C57BL/6-derived mouse model [
44
]. In another example, NOD/SCID mice
deficient in T and B lymphocytes can be infected with a mouse-adapted strain of EV71 [
55
]. However,
since mouse-adapted EV71 can infect even immunocompetent mice [
27
], it was unclear if the missing
T and B lymphocytes in the NOD/SCID mouse model were really important for protection from
mouse-adapted EV71 [
55
]. This issue was addressed in one study when NOD/SCID mice were
inoculated with EV71 clinical isolates and developed limb paralysis via either an i.p. or oral route.
Although limb paralysis has been used as a common marker for scoring EV71 pathogenesis in
mouse modeling, acute flaccid paralysis does not occur as frequently as hand-foot-and-mouth
Viruses 2018,10, 674 8 of 20
disease (HFMD) in human natural infection. Interestingly, in the NOD/SCID model infected with
clinical isolates, an HFMD-like phenotype with skin rash was observed, for the first time in mouse
models [
4
]. Spleen atrophy was found in some EV-71 infected children [
57
,
73
], and was also noted
in this NOD/SCID model infected with clinical isolates [
4
]. Moreover, strong VP1 staining and
severe inflammation were apparent in the paralyzed muscle. On the other hand, no viral protein
and pathology was ever detected in the CNS in EV71-infected NOD/SCID mice. It is worth
mentioning here that in this NOD/SCID model, kinetic analysis in a time course experiment revealed
more delayed clearance of non-infectious viral RNA relative to the clearance of infectious viral
particles [
4
]. Furthermore, the inflammatory IL-23/IL-17 axis appeared to be activated in the infected
NOD/SCID model. It remains to be seen whether these intriguing phenomena are idiosyncratic to the
NOD/SCID model, or they can be generalized to other mouse models or human patients.
Interferon-gamma-inducible protein-10 (IP-10) is a highly expressed chemokine in
EV71-patients [
74
]. IP-10 knock-out mice infected with mouse-adapted EV71 exhibited a higher
mortality rate than the wild type control mice, suggesting a protective role of IP-10 in EV71 infection
and pathogenesis [49].
While the immunodeficient models are very useful for studying pathogenesis and evaluating
efficacies of drug and vaccine candidates, it is not always a good system for testing immune modulators
in therapeutic research. In addition, it is not as direct a platform for vaccine research either, since it can
only be used for comparing potencies of anti-EV71 neutralizing antibodies raised against different
vaccine candidates.
3.3. Transgenic Mouse Models
Recently, many cellular receptors have been proposed for EV71 viral entry
in vitro
, including
hSCARB2 (human scavenger receptor class B) [
60
], PSGL1 (P-selectin glycoprotein ligand 1 [
59
],
annexin II [
61
], and nucleolin [
62
]. These receptors facilitated EV71 infection in non-susceptible
cell lines, such as mouse fibroblast L929. However, transgenic (Tg) mice expressing human PSGL-1
failed to support infection with EV71 clinical isolates [
50
]. Among these reported cellular receptors,
only human SCARB2 (hSCARB2) has so far been shown to support EV71 infection in vivo.
There are two different hSCARB2 Tg mouse models in literature (Table 3) [
51
,
52
]. In the
EF-1a-hSCARB2 model, EF-1a promoter is used to drive hSCARB2 expression in the C57B/6 mouse
background [
52
]. In the SC2-hSCARB2 model, the native hSCARB2 promoter in a human SCARB2
BAC clone drives the transgene expression in the C57B/6 mouse background [
51
]. Both models
can be infected with EV71, develop limb paralysis, and detect viral protein in multiple tissues,
including muscle (muscle-tropism) and the CNS system (neurotropism). On the other hand, these two
models cannot be easily compared with each other due to a number of differences (Table 3): (1) virus
strains and genotypes (B4, B5, C2, C4 vs. Isehara); (2) host age at virus inoculation (1- or 7-day-old
vs. 3-wk-old); (3) virus dose (PFU vs. TCID
50
); (4) inoculation route (s.c. vs. i.c., i.v., i.p.); (5) assays
for readout (individual score of paralysis & death vs. combined score of ataxia, paralysis, and death).
In addition to the Isehara strain (gt C2), other genotypes of EV71 were tested in the SC2-hSCARB2
model [
51
]. However, since the non-Tg wild type mouse control was included only in the study using
the Isehara strain, we listed here in Table 3only the results based on the Isehara strain.
Viruses 2018,10, 674 9 of 20
Table 3. Comparisons between human SCARB2-transgenic and wild type non-transgenic mice in EV71 infection and pathogenesis.
Mouse Strains Promoter Virus Strain
(Genotype) Host Age Virus Dose Inoculation
Route
% Earlier symptom **
(Tg vs. non-Tg)
% Limb Paralysis
(Tg vs. non-Tg)
% Death
(Tg vs. non-Tg) References
pEF-1α- hSCARB2
ubiquitous
EF-1
Promoter
E59 (B4) 1-day-old 3×104PFU s.c. 100 vs. 57.1 18.8 vs. 14.2 0 vs. 0
[52]
N-2838 (B5) 100 vs.75 44.4 vs. 12.5 0 vs. 0
5476 (C2) 7-day-old 107PFU s.c. Nd @100 vs. 57.1 100 vs. 0
N3340 (C4) Nd @100 vs. 100 100 vs. 0
pSC2- hSCARB2 native
promoter Isehara (C2) 3-wk-old 106TCID50
i.c. N/A #100 vs. 0 *
[51]
i.v. N/A #89 vs. 0 *
i.p. N/A #39 vs. 0 *
oral N/A #5 vs. 0 *
@: No disease detected; #: No information available in the original paper; * combined score of ataxia, limb paralysis, and death; ** Earlier symptoms include hair loss and scurf.
Viruses 2018,10, 674 10 of 20
In the pEF-1a-hSCARB2 transgenic mouse model, limb paralysis rate cannot be differentiated
between Tg and non-Tg control mice, when EV71 gt B4 or gt C4 are used (Table 3) [
52
]. However,
mortality rates between Tg and non-Tg are different for gt C4; and the incidences of earlier symptoms
are different between Tg and non-Tg, when gt B4 was used. Despite the limitation in host age and
susceptibility to certain EV71 genotypes in this pEF-1a-hSCARB2 transgenic model, it had been used
in evaluating the efficacy of EV71 vaccine or adjuvant [
75
]. The Isehara strain behaved like a strain
with a high degree of virulence. For other viral isolates, such as BrCr, SK-EV006, and C7, much higher
viral doses were required for developing neurological symptoms in the hSCARB2-Tg10 mouse model.
In fact, even subcutaneous infection of the neonatal non-Tg control mice with the Isehara strain caused
paralysis [
51
]. In addition to the CNS, viral antigens were detected in skeletal muscle of both non-Tg
and Tg mice. In this regard, the hSCARB2 Tg mice are similar to the NOD/SCID mice in supporting
the replication of muscle-tropic EV71.
Neither model of these hSCARB2 Tg mice has so far been shown to support efficient oral
(intragastric) infection, which is a major route for transmission in children [
51
,
52
]. Before these
models are shown to be susceptible to a broader source of different clinical isolates and genotypes, it is
a caveat to carefully choose the best EV71 strains in the experimental design based on the hSCARB2
transgenic model.
3.4. A Hybrid Mouse Model
As discussed above, both hSCARB2 receptor and immune deficiency can contribute to efficient
EV71 infection and pathogenesis. Therefore, it is possible that a combination of a transgenic hSCARB2
and host immune deficiency might further enhance infection efficiency. Indeed, a new hybrid
mouse model was generated by cross-breeding hSCARB2 Tg and stat-1 KO mice. Comparing to
its parental hSCARB2 Tg or stat-1 KO mice, this hybrid mouse model is more user-friendly.
For example, at 2-week-old age, it can still be i.p. infected with different genotypes of EV71 at
1000-fold lower titer (pfu) than the parental mice [
5
]. EV71-infected hybrid mice exhibited high
density of viral protein in the CNS, such as mid-brain and spinal cord. Like the stat-1 KO model,
although EV71-infected hybrid mice developed limb paralysis, neither viral RNA nor protein was ever
detected in the muscle. It suggests that paralysis originated solely from the CNS injury, rather than
myositis and muscle destruction. This new hybrid (hSCARB2 Tg/stat-1 KO) model could serve as a
user-friendly platform for evaluating drug or vaccine efficacy [5].
Although this hybrid mouse model is a far more sensitive system for studying EV71-associated
neuropathogenesis and antiviral therapy, it is deficient in interferon signaling. Therefore, it is not a
good model for studying the therapeutic potential of immune modulators.
4. Pathogenesis Mechanism
Disease manifestations in EV-71 infected children are very diverse, ranging from subclinical infection,
common cold-like syndromes, hand-foot-and-mouth disease, to uncomplicated brainstem encephalitis,
severe dysregulated autonomic nerve system, fatal pulmonary edema, and cardiopulmonary collapse.
To date, while neurological disorders can be modeled in stat-1 KO mice, hybrid mice, and hSCARB2
Tg mice, pulmonary edema and cardiopulmonary failure remain to be recapitulated in EV71
mouse models. Because viral protein has never been detected in patients’ cardiomyocytes, it is generally
believed that cardiopulmonary failure is caused indirectly by the dysregulated autonomic nerve system
in the infected brainstem. Depending on the combination of both viral and host factors, current EV71
animal models could develop a wide variety of pathogenesis similar to human infection. The viral
factors include the infectious dose of EV71, genotypes, mutations, and possibly, sequence heterogeneity
(quasispecies). The host factors include the innate and adaptive immunity, routes of virus inoculation,
and the critical host age receiving viral infection.
Viruses 2018,10, 674 11 of 20
4.1. Viral Determinants of Virulence
In the case of viral factors, both viral load and sequence variations of different EV71 isolates
could significantly contribute to different degrees of virulence and clinical outcome. In the 1970s,
EV71 epidemics occurred in North America and Europe (genotype A, B1, and B2) and showed a
higher percentage of paralysis than the dermatological symptoms [
76
,
77
]. In contrast, clinical reports
in Asia-Pacific after the 1980s (other than genotype A, B1, and B2) observed both dermatological
and neurological symptoms [
3
,
21
,
45
]. Comparison of these results suggested that differences in
ethnic groups or/and genotypes of EV71 may have different sequelae. Sequence analysis identified
some nucleotide or amino acid differences between fatal and non-fatal cases [
78
80
]. For example,
it has been shown that point mutations at amino acid 145 of VP1 or amino acid 149 of VP2 could
determine whether neonatal mice can or cannot be successfully infected with a mouse-adapted EV71
strain [
55
,
81
]. In addition to the coding sequences, adaptive mutations at the IRES region also affected
the virulence in mice [
82
]. Finally, tissue tropism could also be associated with viral sequence variations.
Sequence comparisons of different EV71 isolated from respiratory, gastrointestinal, CNS, and blood
specimens, revealed nucleotide differences at three different positions. Further study showed that
amino acid 97 of VP1 was critical for neurotropism [
83
]. Both amino acid 97 and amino acid 145 of the
capsid protein VP1 were further investigated by two different research groups for their contribution
to pathogenesis in hSCARB2 Tg and non-Tg mouse models, respectively [
84
,
85
]. VP1 mutant L97R
was thought to be more virulent, because it acquired better binding to heparan sulfate on the target
cells [
85
]. Heparan sulfate is a common attachment receptor for many viruses, including EV71 [
63
].
In contrast, VP1 mutant 145G was thought to be less neurovirulent than VP1 mutant 145E, due to
its better binding to heparan sulfate than mutant 145E [
84
]. It was proposed that VP1-145G virus is
preferentially adsorbed by heparan sulfate attachment receptors on an excessive number of non-target
cells during circulation
in vivo
, leading to trapping and attenuation of mutant VP1-145G. In summary,
it would have to be resolved in the future, whether heparan sulfate binding property of EV71 capsid
protein VP1 and in vivo viral virulence, are positively or negatively correlated.
4.2. Host Factors
In this section, we will focus our scope on only those host factors which were investigated in
animal models. In one previous study [
4
], mouse models with different immunogenetic backgrounds
were i.p. infected with the same clinical isolates of EV71, and different disease outcomes and viral
tropisms were observed. For example, in the NOD/SCID mouse model infected with an EV71 clinical
isolate (gt B5), EV71 was predominantly muscle tropic, with severe atrophy and inflammation in the
paralyzed hindlimb muscle. In contrast, in the stat-1 KO and the hybrid mouse models, EV71 behaved
neurotropic with VP1 signals in both brain and spinal cord [4,5]. In the stat-1 KO model, VP1 protein
was often enriched in the Purkinje layer of cerebellar cortex, pons, brain stem,
and spinal cord.
IBA-1+ microglia cells were found to be amplified, indicating a high degree of neuronal injury.
In the hybrid mouse model, Nissl staining of spinal cord sections revealed abnormal neurons with
extensive vaccuoles. Unlike the NOD/SCID model, neither VP1 signal nor inflammation was ever
detected in the paralyzed muscle in stat-1 KO or hybrid mouse models. Since stat-1 is an upstream
transcription factor important for interferon signaling, stat-1 or interferon deficient CNS appeared
to favor neurotropic EV71. By contrast, limb muscles deficient in T and B cells appeared to favor
muscle-tropic EV71.
In human clinical cases, since EV71 infection in Europe and Asia-Pacific had different clinical features,
the difference in their respective ethnic (and immunogenetic) backgrounds (Caucasian vs. Asian)
could be responsible for their different disease manifestations [
6
,
76
,
86
]. Cytotoxic T-lymphocyte
antigen-4 (CTLA-4) is an important regulator of T-cell cytotoxicity and tolerance. The polymorphism
of CTLA-4 was found to be associated with encephalitis in EV71-infected children [
87
]. Other genetic
polymorphisms, such as human leukocyte antigen-A33 (HLA-A33) [
88
], tumor necrosis factor-
α
(TNF-
α
), and interferon-
α
receptor 1 (IFNAR1) [
89
,
90
], were also found to be associated with
Viruses 2018,10, 674 12 of 20
disease severity in EV71 patients. In summary, both viral and host factors are important for
EV71 immunopathogenesis.
5. Therapy
EV71 vaccination should be an effective way to protect children from infection. While inactivated
whole virus of EV71 appears to be a good vaccine candidate [
91
94
], it is unclear if it can be used
for all age groups of children. Therapeutics against EV71 remains an urgent need. We summarized
here in Table 4a list of candidate therapeutics, which had been tested in patients or animal models.
We categorized these candidate compounds into four different groups according to their approaches.
Group 1 relied on passive immunization with anti-EV71 neutralizing sera raised against
recombinant VP1 protein, virus-like particles (VLP), formaldehyde-inactivated whole virus, and IVIG
from human source [ref cited in Table 4]. Although IVIG were used in clinical practice [
17
],
the possibility of antibody-dependent enhancement of EV71 must be seriously considered [
19
21
].
Another important issue is whether these immune sera or antibodies can efficiently and broadly
cross protect against EV71 of different genotypes. Mouse monoclonal antibodies (mAb) need to be
humanized and re-tested in animal models in the future. Immune escape variants could be selected
during mAb treatment.
Group 2 includes immune modulators, such as interferon (IFN-
α
) or TLR agonists. Pre-treatment
with Poly (I:C) or co-treatment with mouse IFN and EV71, can reduce the death rate in EV71-infected
mice [
45
]. However, some of these studies in Group 2 are prophylactic rather than therapeutic.
Oral intake of GS-9620 (a TLR7 agonist) inhibited viral replication and increased the survival rate of
ICR mice infected with a mouse-adapted strain MP10 [
95
]. In clinical trials, IFN-
α
can decrease the
fever and viral load in mild HFMD patients [
72
,
96
]. Altogether, activation of IFN signaling could be a
potential therapy for EV71 infection.
Group 3 includes anti-inflammation treatments, such as anti-IL6 Ab, and complement
inhibitors [
38
,
97
]. Although compounds in Group 3 may not have any direct antiviral effect, they can
help reduce inflammatory injury by excessive cytokines.
Group 4 includes various compounds, whose mechanisms either remain unclear at present, or do
not belong to the above Group 1–3 [
22
,
98
101
]. For example, Milrinone is supposed to help maintain
the level of catecholamine [
22
]. As a side note, it is worth mentioning that disinfectants against EV71
have also been investigated. While disinfectants have no therapeutic value, solutions of chlorine
dioxide (ClO2) [102] and methylene blue (photosensitizer) [103] can inactivate EV71.
Viruses 2018,10, 674 13 of 20
Table 4. Therapeutic compounds tested in animal models and human patients.
Therapeutic
Approaches Drug Candidates Drug Delivery Methods Human or
Animal Model
Host Age at Virus
Inoculation
Virus
Inoculation
EV71 Clinical Isolates *
or Adapted Strains #
Virus
Genotype Ref.
GROUP 1
Passive
immunization
bispecific anti-EV71 &
CA16 Ab Bs(scFv) i.p. injection at 24 h after inoculation BALB/c 1-day-old i.c. EV71/pSVA-MP4 #B3 [104]
VP1-specific mAb i.p. injection at 3, 24, and 48 h after
inoculation SCARB2-Tg 1-day & 7-day-old s.c. E59 & 5746-TW98 * B4 & C2 [105]
human plasma or IVIG in vitro
pre-mix with EV71 at 37
C for 1 h
ICR 2-day-old i.p. KM593929 * C4 [106]
human IVIG 3 i.p. injections at 4 h, 1- and 2-days after
inoculation Kunming mice 7-day-old i.p. AH08/06 * C4 [107]
EV71-specific Ab
i.p. injection 1 day before and 1 day after
(ICR), or 2 and 4 days after inoculation
(C57BL/6)
ICR & C57BL/6 7-day & 9-day-old oral M2 #C2 [44]
adult immune sera i.p. injection at 1hr before and 24 h after
inoculation young gerbils 21-day-old i.p. or i.m. strain 58301 * C4 [108]
human IVIG
i.v. infusion at a dosage of 1 g/kg/day for
2 days human patients <2-yr-old
natural infection
N/A N/A [17]
GROUP 2
Immune
modulators
Poly (I:C) i.p. injection at 12 h before inoculation ICR 3-day-old i.p. or oral MP4 #C2 [45]
Ad-IFN-a one intranasal shot within 12 h
post-inoculation BALB/c 6-day-old i.p. strain 41 * B4 [71]
antagomir -146a i.p. injection before or after virus
inoculation C57BL/6 7-day-old i.p. mEV71 #B2 [109]
GS-9620 oral uptake at 2, 26, 50 h post-inoculation ICR 10-day-old i.p. MP10 #C4 [95]
GROUP 3
Anti-inflammation
anti-IL6 Ab i.p. co-injection of Ab and virus (day 0) BALB/c 1-day-old i.p. strain 41 * B4 [38]
complement inhibitor
(CR2-crry) treatment post-inoculation ICR 7-day-old i.c. BrCr * A [97]
GROUP 4
Others
Retro-2cycl i.p. injection following EV71 inoculation BALB/c <1-day-old i.c. KJ508817 * C4 [98]
Pleconaril daily i.p. injection for 5 days ICR 1-day-old i.p. BrCr * A [99]
andrographolide
sulfonate
i.p. injection once daily until 10 days
post-inoculation ICR 7-day-old i.p. BJ09/07 #C4 [100]
adoptive transfer of
macrophage
i.p. injection with adult macrophage at 1
day post-inoculation ICR 10-day-old i.p. MP10 #C4 [101]
Milrinone i.v. injection within 2–6 h after admission
at dosage 0.35–0.55 mg/kg/min for 72 h. human patients <2-yr-old
natural infection
N/A N/A [22]
i.p.: intraperitoneal; i.v.: intravenous; i.c.: intracranial; i.m.: intra-muscular; s.c.: subcutaneous; N/A: information not available in the original paper; * clinical isolates;
#
mouse-adapted strain.
Viruses 2018,10, 674 14 of 20
6. Conclusions
In 2018, there are many issues that need to be solved in the modeling of EV71 infection and
pathogenesis. First, while the immune deficient NOD/SCID mice offer an efficient model for oral
infection with EV71 clinical isolates, one remaining challenge is to establish an oral infection system in
an immune-competent mouse model. Second, another mystery is the requirement of a neonatal age
for productive EV71 infection and pathogenesis. It is widely speculated that some kind of age-related
immune maturation could confer the resistance to pediatric infections, such as EV71. However, if so,
the exact nature of such an age-dependent arm of the immune system remains elusive. It cannot be
excluded that a putative EV71 receptor could be expressed in muscle or nerve only transiently at the
neonatal age, but rapidly turned off at an older age during mouse development. Third, there is an
urgent need for FDA-approved antivirals against the frequent EV71 epidemics in Asia and worldwide.
Lessons that we learned from EV71 should be useful for other picornaviruses closely related to EV71,
such as the most recent D68 virus that emerged in North America a few years ago.
Funding:
The authors were supported by Institute of Biomedical Sciences, Academia Sinica and the EID Program,
Ministry of Science and Technology, Taiwan (MOST 106-2320-B-001- 010, MOST 106-0210-01-15-02, MOST
107-2321-B-001-028, and MOST 107-0210-01-19-01).
Acknowledgments:
We thank the collaborations from Ya-Yen Yu, Chi-Yung Lin, and Jen-Shiou Lin at Changhua
Christian Hospital, Changhua, Taiwan.
Conflicts of Interest: We declare no competing financial interests.
References
1. Schmidt, N.J.; Lennette, E.H.; Ho, H.H. An apparently new enterovirus isolated from patients with disease
of the central nervous system. J. Infect. Dis. 1974,129, 304–309. [CrossRef] [PubMed]
2.
Brown, B.A.; Pallansch, M.A. Complete nucleotide sequence of enterovirus 71 is distinct from poliovirus.
Virus Res. 1995,39, 195–205. [CrossRef]
3.
Cox, J.A.; Hiscox, J.A.; Solomon, T.; Ooi, M.H.; Ng, L.F.P. Immunopathogenesis and virus-host interactions
of enterovirus 71 in patients with hand, foot and mouth disease. Front. Microbiol.
2017
,8, 2249. [CrossRef]
[PubMed]
4.
Liao, C.C.; Liou, A.T.; Chang, Y.S.; Wu, S.Y.; Chang, C.S.; Lee, C.K.; Kung, J.T.; Tu, P.H.; Yu, Y.Y.;
Lin, C.Y.; et al.
Immunodeficient mouse models with different disease profiles by
in vivo
infection with the same clinical
isolate of enterovirus 71. J. Virol. 2014,88, 12485–12499. [CrossRef] [PubMed]
5.
Liou, A.-T.; Wu, S.-Y.; Liao, C.-C.; Chang, Y.-S.; Chang, C.-S.; Shih, C. A new animal model containing human
SCARB2 and lacking stat-1 is highly susceptible to EV71. Sci. Rep. 2016,6, 31151. [CrossRef] [PubMed]
6.
Ho, M.; Chen, E.R.; Hsu, K.H.; Twu, S.J.; Chen, K.T.; Tsai, S.F.; Wang, J.R.; Shih, S.R. An epidemic of
enterovirus 71 infection in Taiwan. N. Engl. J. Med. 1999,341, 929–935. [CrossRef] [PubMed]
7. Chang, L.-Y.; Huang, L.-M.; Gau, S.S.-F.; Wu, Y.-Y.; Hsia, S.-H.; Fan, T.-Y.; Lin, K.-L.; Huang, Y.-C.; Lu, C.-Y.;
Lin, T.-Y. Neurodevelopment and cognition in children after enterovirus 71 infection. N. Engl. J. Med.
2007
,
356, 1226–1231. [CrossRef] [PubMed]
8.
Wu, J.-M.; Wang, J.-N.; Tsai, Y.-C.; Liu, C.-C.; Huang, C.-C.; Chen, Y.-J.; Yeh, T.-F. Cardiopulmonary
manifestations of fulminant enterovirus 71 infection. Pediatrics 2002,109, e26. [CrossRef] [PubMed]
9.
Shimizu, H.; Utama, A.; Onnimala, N.; Li, C.; Zhang, L.-B.; Ma, Y.-J.; Pongsuwanna, Y.; Miyamura, T.
Molecular epidemiology of enterovirus 71 infection in the western pacific region. Pediatr. Int.
2004
,46,
231–235. [CrossRef] [PubMed]
10.
Mizuta, K.; Aoki, Y.; Matoba, Y.; Yahagi, K.; Itagaki, T.; Katsushima, F.; Katsushima, Y.; Ito, S.; Hongo, S.;
Matsuzaki, Y. Molecular epidemiology of enterovirus 71 strains isolated from children in Yamagata, Japan,
between 1990 and 2013. J. Med. Microbiol. 2014,63, 1356–1362. [CrossRef] [PubMed]
11.
AbuBakar, S.; Chee, H.Y.; Al-Kobaisi, M.F.; Xiaoshan, J.; Chua, K.B.; Lam, S.K. Identification of enterovirus 71
isolates from an outbreak of hand, foot and mouth disease (HFMD) with fatal cases of encephalomyelitis in
malaysia. Virus Res. 1999,61, 1–9. [CrossRef]
Viruses 2018,10, 674 15 of 20
12.
Wang, Y.; Feng, Z.; Yang, Y.; Self, S.; Gao, Y.; Longini, I.M.; Wakefield, J.; Zhang, J.; Wang, L.;
Chen, X.; et al.
Hand, foot, and mouth disease in china: Patterns of spread and transmissibility. Epidemiology
2011
,22,
781–792. [CrossRef] [PubMed]
13.
Fischer, T.K.; Nielsen, A.Y.; Sydenham, T.V.; Andersen, P.H.; Andersen, B.; Midgley, S.E. Emergence of
enterovirus 71 C4a in Denmark, 2009 to 2013. Eurosurveillance 2014,19, 20911. [CrossRef] [PubMed]
14.
Medici, M.C.; Tummolo, F.; Arcangeletti, M.C.; De Conto, F.; Chezzi, C.; Dodi, I.; Calderaro, A. A cluster of
enterovirus 71 subgenogroup C2 in a nursery school, Italy, 2014. New Microbiol.
2016
,39, 295–298. [PubMed]
15.
Akhmadishina, L.V.; Eremeeva, T.P.; Trotsenko, O.E.; Ivanova, O.E.; Mikhailov, M.I.; Lukashev, A.N.
Seroepidemiology and molecular epidemiology of enterovirus 71 in Russia. PLoS ONE
2014
,9, e97404.
[CrossRef] [PubMed]
16.
Zander, A.; Britton, P.N.; Navin, T.; Horsley, E.; Tobin, S.; McAnulty, J.M. An outbreak of enterovirus 71 in
metropolitan Sydney: Enhanced surveillance and lessons learnt. Med. J. Aust.
2014
,201, 663–666. [CrossRef]
[PubMed]
17.
Wang, S.M.; Lei, H.Y.; Huang, M.C.; Su, L.Y.; Lin, H.C.; Yu, C.K.; Wang, J.L.; Liu, C.C. Modulation of
cytokine production by intravenous immunoglobulin in patients with enterovirus 71-associated brainstem
encephalitis. J. Clin. Virol. 2006,37, 47–52. [CrossRef] [PubMed]
18.
Cao, R.Y.; Dong, D.Y.; Liu, R.J.; Han, J.F.; Wang, G.C.; Zhao, H.; Li, X.F.; Deng, Y.Q.; Zhu, S.Y.; Wang, X.Y.; et al.
Human igg subclasses against enterovirus type 71: Neutralization versus antibody dependent enhancement
of infection. PLoS ONE 2013,8, e64024. [CrossRef] [PubMed]
19.
Wang, S.M.; Chen, I.C.; Su, L.Y.; Huang, K.J.; Lei, H.Y.; Liu, C.C. Enterovirus 71 infection of monocytes with
antibody-dependent enhancement. Clin. Vaccine Immunol. 2010,17, 1517–1523. [CrossRef] [PubMed]
20.
Han, J.F.; Cao, R.Y.; Deng, Y.Q.; Tian, X.; Jiang, T.; Qin, E.D.; Qin, C.F. Antibody dependent enhancement
infection of enterovirus 71 in vitro and in vivo. Virol. J. 2011,8, 106. [CrossRef] [PubMed]
21.
Chen, I.C.; Wang, S.-M.; Yu, C.-K.; Liu, C.-C. Subneutralizing antibodies to enterovirus 71 induce
antibody-dependent enhancement of infection in newborn mice. Med. Microbiol. Immunol.
2013
,202,
259–265. [CrossRef] [PubMed]
22.
Wang, S.M.; Lei, H.Y.; Huang, M.C.; Wu, J.M.; Chen, C.T.; Wang, J.N.; Wang, J.R.; Liu, C.C. Therapeutic
efficacy of milrinone in the management of enterovirus 71-induced pulmonary edema. Pediatr. Pulmonol.
2005,39, 219–223. [CrossRef] [PubMed]
23.
Han, J.F.; Cao, R.Y.; Tian, X.; Yu, M.; Qin, E.D.; Qin, C.F. Producing infectious enterovirus type 71 in a rapid
strategy. Virol. J. 2010,7, 116. [CrossRef] [PubMed]
24.
Yu, P.; Bao, L.; Xu, L.; Li, F.; Lv, Q.; Deng, W.; Xu, Y.; Qin, C. Neurotropism
in vitro
and mouse models of
severe and mild infection with clinical strains of enterovirus 71. Viruses 2017,9, 351. [CrossRef] [PubMed]
25.
Wang, C.; Ji, L.; Yuan, X.; Jin, Y.; Cardona, C.J.; Xing, Z. Differential regulation of tlr signaling on the induction
of antiviral interferons in human intestinal epithelial cells infected with enterovirus 71. PLoS ONE
2016
,11,
e0152177. [CrossRef] [PubMed]
26.
Lui, Y.L.E.; Timms, P.; Hafner, L.M.; Tan, T.L.; Tan, K.H.; Tan, E.L. Characterisation of enterovirus 71
replication kinetics in human colorectal cell line, HT29. SpringerPlus 2013,2, 267. [CrossRef] [PubMed]
27.
Wang, Y.F.; Chou, C.T.; Lei, H.Y.; Liu, C.C.; Wang, S.M.; Yan, J.J.; Su, I.J.; Wang, J.R.; Yeh, T.M.; Chen, S.H.; et al.
A mouse-adapted enterovirus 71 strain causes neurological disease in mice after oral infection. J. Virol.
2004
,
78, 7916–7924. [CrossRef] [PubMed]
28.
Xing, Z.; Huang, H.-I.; Chio, C.-C.; Lin, J.-Y. Inhibition of EV71 by curcumin in intestinal epithelial cells.
PLoS ONE 2018,13, e0191617.
29.
Ang, L.Y.E.; Too, H.K.I.; Tan, E.L.; Chow, T.-K.V.; Shek, P.-C.L.; Tham, E.; Alonso, S. Antiviral activity of
lactobacillus reuteri protectis against coxsackievirus A and enterovirus 71 infection in human skeletal muscle
and colon cell lines. Virol. J. 2016,13, 111. [CrossRef] [PubMed]
30.
Liu, Y.; Zheng, Z.; Shu, B.; Meng, J.; Zhang, Y.; Zheng, C.; Ke, X.; Gong, P.; Hu, Q.; Wang, H.; et al.
Sumo modification stabilizes enterovirus 71 polymerase 3D to facilitate viral replication. J. Virol.
2016
,90,
10472–10485. [CrossRef] [PubMed]
31.
Du, X.; Wang, H.; Xu, F.; Huang, Y.; Liu, Z.; Liu, T.E. Enterovirus 71 induces apoptosis of SH-SY5Y human
neuroblastoma cells through stimulation of endogenous microRNA let-7b expression. Mol. Med. Rep. 2015,
12, 953–959. [CrossRef] [PubMed]
Viruses 2018,10, 674 16 of 20
32.
Xu, L.J.; Jiang, T.; Zhang, F.J.; Han, J.F.; Liu, J.; Zhao, H.; Li, X.F.; Liu, R.J.; Deng, Y.Q.; Wu, X.Y.; et al. Global
transcriptomic analysis of human neuroblastoma cells in response to enterovirus type 71 infection. PLoS ONE
2013,8, e65948. [CrossRef] [PubMed]
33.
Too, I.H.K.; Yeo, H.; Sessions, O.M.; Yan, B.; Libau, E.A.; Howe, J.L.C.; Lim, Z.Q.; Suku-Maran, S.; Ong, W.-Y.;
Chua, K.B.; et al. Enterovirus 71 infection of motor neuron-like NSC-34 cells undergoes a non-lytic exit
pathway. Sci. Rep. 2016,6, 36983. [CrossRef] [PubMed]
34.
Shih, S.-R.; Weng, K.-F.; Stollar, V.; Li, M.-L. Viral protein synthesis is required for enterovirus 71 to induce
apoptosis in human glioblastoma cells. J. Neurovirol. 2008,14, 53–61. [CrossRef] [PubMed]
35.
Tse, H.; Victorio, C.B.L.; Xu, Y.; Ng, Q.; Chow, V.T.K.; Chua, K.B. Phenotypic and genotypic characteristics of
novel mouse cell line (NIH/3T3)-adapted human enterovirus 71 strains (EV71:TLLm and EV71:TLLmv).
PLoS ONE 2014,9, e92719.
36.
Wang, B.; Xi, X.; Lei, X.; Zhang, X.; Cui, S.; Wang, J.; Jin, Q.; Zhao, Z. Enterovirus 71 protease 2Apro targets
MAVS to inhibit anti-viral type I interferon responses. PLoS Pathog.
2013
,9, e1003231. [CrossRef] [PubMed]
37.
Wang, W.; Xiao, F.; Wan, P.; Pan, P.; Zhang, Y.; Liu, F.; Wu, K.; Liu, Y.; Wu, J. Ev71 3d protein binds with
nlrp3 and enhances the assembly of inflammasome complex. PLoS Pathog.
2017
,13, e1006123. [CrossRef]
[PubMed]
38.
Khong, W.X.; Foo, D.G.; Trasti, S.L.; Tan, E.L.; Alonso, S. Sustained high levels of interleukin-6 contribute
to the pathogenesis of enterovirus 71 in a neonate mouse model. J. Virol.
2011
,85, 3067–3076. [CrossRef]
[PubMed]
39.
Chang, G.H.; Lin, L.; Luo, Y.J.; Cai, L.J.; Wu, X.Y.; Xu, H.M.; Zhu, Q.Y. Sequence analysis of six enterovirus 71
strains with different virulences in humans. Virus Res. 2010,151, 66–73. [CrossRef] [PubMed]
40.
Zaini, Z.; McMinn, P. A single mutation in capsid protein VP1 (Q145E) of a genogroup C4 strain of human
enterovirus 71 generates a mouse-virulent phenotype. J. Gen. Virol.
2012
,93, 1935–1940. [CrossRef] [PubMed]
41.
Chua, B.H.; Phuektes, P.; Sanders, S.A.; Nicholls, P.K.; McMinn, P.C. The molecular basis of mouse adaptation
by human enterovirus 71. J. Gen. Virol. 2008,89, 1622–1632. [CrossRef] [PubMed]
42.
Chen, Y.C.; Yu, C.-K.; Wang, Y.-F.; Liu, C.-C.; Su, I.-J.; Lei, H.-Y. A murine oral enterovirus 71 infection model
with central nervous system involvement. J. Gen. Virol. 2004,85, 69–77. [CrossRef] [PubMed]
43.
Wang, W.; Duo, J.; Liu, J.; Ma, C.; Zhang, L.; Wei, Q.; Qin, C. A mouse muscle-adapted enterovirus 71 strain
with increased virulence in mice. Microbes Infect. 2011,13, 862–870. [CrossRef] [PubMed]
44.
Lin, Y.W.; Chang, K.C.; Kao, C.M.; Chang, S.P.; Tung, Y.Y.; Chen, S.H. Lymphocyte and antibody responses
reduce enterovirus 71 lethality in mice by decreasing tissue viral loads. J. Virol.
2009
,83, 6477–6483.
[CrossRef] [PubMed]
45.
Liu, M.L.; Lee, Y.-P.; Liu, C.-C.; Yeh, T.-M.; Wang, S.-M.; Chen, S.-H.; Wang, Y.-F.; Su, I.-J.; Lei, H.-Y.;
Wang, J.-R.; et al. Type I interferons protect mice against enterovirus 71 infection. J. Gen. Virol.
2005
,86,
3263–3269. [CrossRef] [PubMed]
46.
Li, Z.H.; Li, C.M.; Ling, P.; Shen, F.H.; Chen, S.H.; Liu, C.C.; Yu, C.K.; Chen, S.H. Ribavirin reduces mortality
in enterovirus 71-infected mice by decreasing viral replication. J. Infect. Dis.
2008
,197, 854–857. [CrossRef]
[PubMed]
47.
Ong, K.C.; Devi, S.; Cardosa, M.J.; Wong, K.T. Formaldehyde-inactivated whole-virus vaccine protects a
murine model of enterovirus 71 encephalomyelitis against disease. J. Virol.
2010
,84, 661–665. [CrossRef]
[PubMed]
48.
Ong, K.C.; Badmanathan, M.; Devi, S.; Leong, K.L.; Cardosa, M.J.; Wong, K.T. Pathologic characterization of
a murine model of human enterovirus 71 encephalomyelitis. J. Neuropathol. Exp. Neurol.
2008
,67, 532–542.
[CrossRef] [PubMed]
49.
Shen, F.H.; Tsai, C.C.; Wang, L.C.; Chang, K.C.; Tung, Y.Y.; Su, I.J.; Chen, S.H. Enterovirus 71 infection
increases expression of interferon-gamma-inducible protein 10 which protects mice by reducing viral burden
in multiple tissues. J. Gen. Virol. 2013,94, 1019–1027. [CrossRef] [PubMed]
50.
Liu, J.; Dong, W.; Quan, X.; Ma, C.; Qin, C.; Zhang, L. Transgenic expression of human p-selectin glycoprotein
ligand-1 is not sufficient for enterovirus 71 infection in mice. Arch. Virol.
2011
,157, 539–543. [CrossRef]
[PubMed]
51.
Fujii, K.; Nagata, N.; Sato, Y.; Ong, K.C.; Wong, K.T.; Yamayoshi, S.; Shimanuki, M.; Shitara, H.; Taya, C.;
Koike, S. Transgenic mouse model for the study of enterovirus 71 neuropathogenesis. Proc. Natl. Acad.
Sci. USA 2013,110, 14753–14758. [CrossRef] [PubMed]
Viruses 2018,10, 674 17 of 20
52.
Lin, Y.-W.; Yu, S.-L.; Shao, H.-Y.; Lin, H.-Y.; Liu, C.-C.; Hsiao, K.-N.; Chitra, E.; Tsou, Y.-L.; Chang, H.-W.;
Sia, C.; et al. Human SCARB2 transgenic mice as an infectious animal model for enterovirus 71. PLoS ONE
2013,8, e57591. [CrossRef] [PubMed]
53.
Khong, W.X.; Yan, B.; Yeo, H.; Tan, E.L.; Lee, J.J.; Ng, J.K.; Chow, V.T.; Alonso, S. A non-mouse-adapted
enterovirus 71 (EV71) strain exhibits neurotropism, causing neurological manifestations in a novel mouse
model of EV71 infection. J. Virol. 2012,86, 2121–2131. [CrossRef] [PubMed]
54.
Caine, E.A.; Partidos, C.D.; Santangelo, J.D.; Osorio, J.E. Adaptation of enterovirus 71 to adult interferon
deficient mice. PLoS ONE 2013,8, e59501. [CrossRef] [PubMed]
55.
Arita, M.; Ami, Y.; Wakita, T.; Shimizu, H. Cooperative effect of the attenuation determinants derived from
poliovirus sabin 1 strain is essential for attenuation of enterovirus 71 in the NOD/SCID mouse infection
model. J. Virol. 2008,82, 1787–1797. [CrossRef] [PubMed]
56.
Lyu, K.; Wang, G.C.; He, Y.L.; Han, J.F.; Ye, Q.; Qin, C.F.; Chen, R. Crystal structures of enterovirus 71
(EV71) recombinant virus particles provide insights into vaccine design. J. Biol. Chem.
2015
,290, 3198–3208.
[CrossRef] [PubMed]
57.
Ooi, M.H.; Wong, S.C.; Lewthwaite, P.; Cardosa, M.J.; Solomon, T. Clinical features, diagnosis, and
management of enterovirus 71. Lancet Neurol. 2010,9, 1097–1105. [CrossRef]
58.
Chang, L.Y.; King, C.C.; Hsu, K.H.; Ning, H.C.; Tsao, K.C.; Li, C.C.; Huang, Y.C.; Shih, S.R.; Chiou, S.T.;
Chen, P.Y.; et al. Risk factors of enterovirus 71 infection and associated hand, foot, and mouth
disease/herpangina in children during an epidemic in taiwan. Pediatrics
2002
,109, e88. [CrossRef] [PubMed]
59.
Nishimura, Y.; Shimojima, M.; Tano, Y.; Miyamura, T.; Wakita, T.; Shimizu, H. Human p-selectin glycoprotein
ligand-1 is a functional receptor for enterovirus 71. Nat. Med. 2009,15, 794–797. [CrossRef] [PubMed]
60.
Yamayoshi, S.; Yamashita, Y.; Li, J.; Hanagata, N.; Minowa, T.; Takemura, T.; Koike, S. Scavenger receptor B2
is a cellular receptor for enterovirus 71. Nat. Med. 2009,15, 798–801. [CrossRef] [PubMed]
61.
Yang, S.L.; Chou, Y.T.; Wu, C.N.; Ho, M.S. Annexin ii binds to capsid protein vp1 of enterovirus 71 and
enhances viral infectivity. J. Virol. 2011,85, 11809–11820. [CrossRef] [PubMed]
62.
Su, P.Y.; Wang, Y.F.; Huang, S.W.; Lo, Y.C.; Wang, Y.H.; Wu, S.R.; Shieh, D.B.; Chen, S.H.; Wang, J.R.;
Lai, M.D.; et al.
Cell surface nucleolin facilitates enterovirus 71 binding and infection. J. Virol.
2015
,89,
4527–4538. [CrossRef] [PubMed]
63.
Tan, C.W.; Poh, C.L.; Sam, I.C.; Chan, Y.F. Enterovirus 71 uses cell surface heparan sulfate glycosaminoglycan
as an attachment receptor. J. Virol. 2013,87, 611–620. [CrossRef] [PubMed]
64.
Ren, X.X.; Ma, L.; Liu, Q.W.; Li, C.; Huang, Z.; Wu, L.; Xiong, S.D.; Wang, J.H.; Wang, H.B. The molecule of
DC-SIGN captures enterovirus 71 and confers dendritic cell-mediated viral trans-infection. Virol. J.
2014
,11,
47. [CrossRef] [PubMed]
65.
Fukuhara, M.; Iwami, S.; Sato, K.; Nishimura, Y.; Shimizu, H.; Aihara, K.; Koyanagi, Y. Quantification of the
dynamics of enterovirus 71 infection by experimental-mathematical investigation. J. Virol.
2013
,87, 701–705.
[CrossRef] [PubMed]
66.
Yu, P.; Gao, Z.; Zong, Y.; Bao, L.; Xu, L.; Deng, W.; Li, F.; Lv, Q.; Gao, Z.; Xu, Y.; et al. Histopathological features
and distribution of EV71 antigens and SCARB2 in human fatal cases and a mouse model of enterovirus 71
infection. Virus Res. 2014,189, 121–132. [CrossRef] [PubMed]
67.
Chen, C.S.; Yao, Y.C.; Lin, S.C.; Lee, Y.P.; Wang, Y.F.; Wang, J.R.; Liu, C.C.; Lei, H.Y.; Yu, C.K. Retrograde axonal
transport: A major transmission route of enterovirus 71 in mice. J. Virol.
2007
,81, 8996–9003. [CrossRef]
[PubMed]
68.
Wong, K.T.; Munisamy, B.; Ong, K.C.; Kojima, H.; Noriyo, N.; Chua, K.B.; Ong, B.B.; Nagashima, K. The
distribution of inflammation and virus in human enterovirus 71 encephalomyelitis suggests possible viral
spread by neural pathways. J. Neuropathol. Exp. Neurol. 2008,67, 162–169. [CrossRef] [PubMed]
69.
Ren, R.; Racaniello, V.R. Poliovirus spreads from muscle to the central nervous system by neural pathways.
J. Infect. Dis. 1992,166, 747–752. [CrossRef] [PubMed]
70.
Zhang, Y.; Li, J.; Li, Q. Immune evasion of enteroviruses under innate immune monitoring. Front. Microbiol.
2018,9, 1866. [CrossRef] [PubMed]
71.
Sun, J.; Ennis, J.; Turner, J.D.; Chu, J.J. Single dose of an adenovirus vectored mouse interferon-alpha protects
mice from lethal EV71 challenge. Antivir. Res. 2016,134, 207–215. [CrossRef] [PubMed]
Viruses 2018,10, 674 18 of 20
72.
Lin, H.; Huang, L.; Zhou, J.; Lin, K.; Wang, H.; Xue, X.; Xia, C. Efficacy and safety of interferon-
α
2b spray in
the treatment of hand, foot, and mouth disease: A multicenter, randomized, double-blind trial. Arch. Virol.
2016,161, 3073–3080. [CrossRef] [PubMed]
73.
Meng, G.; Li, M.; Li, Y.; Wang, X.; Chen, Q.; Wei, H.J.C.J.C.E.P. Severe hand-foot-mouth disease: A report of 3
autopsied cases. Chin. J. Clin. Exp. Pathol. 2011,27, 48–51.
74.
Wang, S.M.; Lei, H.Y.; Yu, C.K.; Wang, J.R.; Su, I.J.; Liu, C.C. Acute chemokine response in the blood and
cerebrospinal fluid of children with enterovirus 71-associated brainstem encephalitis. J. Infect. Dis.
2008
,198,
1002–1006. [CrossRef] [PubMed]
75.
Lin, Y.L.; Chow, Y.H.; Huang, L.M.; Hsieh, S.M.; Cheng, P.Y.; Hu, K.C.; Chiang, B.L. A CpG-adjuvanted
intranasal enterovirus 71 vaccine elicits mucosal and systemic immune responses and protects human
SCARB2-transgenic mice against lethal challenge. Sci. Rep. 2018,8, 10713. [CrossRef] [PubMed]
76.
Chumakov, M.; Voroshilova, M.; Shindarov, L.; Lavrova, I.; Gracheva, L.; Koroleva, G.; Vasilenko, S.;
Brodvarova, I.; Nikolova, M.; Gyurova, S.; et al. Enterovirus 71 isolated from cases of epidemic
poliomyelitis-like disease in Bulgaria. Arch. Virol. 1979,60, 329–340. [CrossRef] [PubMed]
77.
Nagy, G.; Takatsy, S.; Kukan, E.; Mihaly, I.; Domok, I. Virological diagnosis of enterovirus type 71 infections:
Experiences gained during an epidemic of acute CNS diseases in hungary in 1978. Arch. Virol.
1982
,71,
217–227. [CrossRef] [PubMed]
78.
Shih, S.R.; Ho, M.S.; Lin, K.H.; Wu, S.L.; Chen, Y.T.; Wu, C.N.; Lin, T.Y.; Chang, L.Y.; Tsao, K.C.;
Ning, H.C.; et al
. Genetic analysis of enterovirus 71 isolated from fatal and non-fatal cases of hand, foot and
mouth disease during an epidemic in taiwan, 1998. Virus Res. 2000,68, 127–136. [CrossRef]
79.
Yan, J.J.; Su, I.J.; Chen, P.F.; Liu, C.C.; Yu, C.K.; Wang, J.R. Complete genome analysis of enterovirus 71
isolated from an outbreak in taiwan and rapid identification of enterovirus 71 and coxsackievirus A16 by
RT-PCR. J. Med. Virol. 2001,65, 331–339. [CrossRef] [PubMed]
80.
Li, R.; Zou, Q.; Chen, L.; Zhang, H.; Wang, Y. Molecular analysis of virulent determinants of enterovirus 71.
PLoS ONE 2011,6, e26237. [CrossRef] [PubMed]
81.
Huang, S.W.; Wang, Y.F.; Yu, C.K.; Su, I.J.; Wang, J.R. Mutations in VP2 and VP1 capsid proteins increase
infectivity and mouse lethality of enterovirus 71 by virus binding and RNA accumulation enhancement.
Virology 2012,422, 132–143. [CrossRef] [PubMed]
82.
Yeh, M.T.; Wang, S.W.; Yu, C.K.; Lin, K.H.; Lei, H.Y.; Su, I.J.; Wang, J.R. A single nucleotide in stem loop
II of 5
0
-untranslated region contributes to virulence of enterovirus 71 in mice. PLoS ONE
2011
,6, e27082.
[CrossRef] [PubMed]
83.
Cordey, S.; Petty, T.J.; Schibler, M.; Martinez, Y.; Gerlach, D.; van Belle, S.; Turin, L.; Zdobnov, E.; Kaiser, L.;
Tapparel, C. Identification of site-specific adaptations conferring increased neural cell tropism during human
enterovirus 71 infection. PLoS Pathog. 2012,8, e1002826. [CrossRef] [PubMed]
84.
Kobayashi, K.; Sudaka, Y.; Takashino, A.; Imura, A.; Fujii, K.; Koike, S. Amino acid variation at VP1-145 of
enterovirus 71 determines attachment receptor usage and neurovirulence in human scavenger receptor B2
transgenic mice. J. Virol. 2018. [CrossRef] [PubMed]
85.
Tseligka, E.D.; Sobo, K.; Stoppini, L.; Cagno, V.; Abdul, F.; Piuz, I.; Meylan, P.; Huang, S.; Constant, S.;
Tapparel, C. A VP1 mutation acquired during an enterovirus 71 disseminated infection confers heparan
sulfate binding ability and modulates ex vivo tropism. PLoS Pathog.
2018
,14, e1007190. [CrossRef] [PubMed]
86.
McMinn, P.C. An overview of the evolution of enterovirus 71 and its clinical and public health significance.
FEMS Microbiol. Rev. 2002,26, 91–107. [CrossRef] [PubMed]
87.
Yang, K.D.; Yang, M.Y.; Li, C.C.; Lin, S.F.; Chong, M.C.; Wang, C.L.; Chen, R.F.; Lin, T.Y. Altered cellular but
not humoral reactions in children with complicated enterovirus 71 infections in Taiwan. J. Infect. Dis.
2001
,
183, 850–856. [CrossRef] [PubMed]
88.
Chang, L.Y.; Chang, I.S.; Chen, W.J.; Huang, Y.C.; Chen, G.W.; Shih, S.R.; Juang, J.L.; Shih, H.M.; Hsiung, C.A.;
Lin, T.Y.; et al. HLA-A33 is associated with susceptibility to enterovirus 71 infection. Pediatrics
2008
,122,
1271–1276. [CrossRef] [PubMed]
89.
Li, J.A.; Chen, Z.B.; Lv, T.G.; Han, Z.L.; Liu, P.P. Impact of endothelial nitric oxide synthase gene
polymorphism on severity of enterovirus 71-infection in Chinese children. Clin. Biochem.
2013
,46, 1842–1847.
[CrossRef] [PubMed]
Viruses 2018,10, 674 19 of 20
90.
Zou, R.; Zhang, G.; Li, S.; Wang, W.; Yuan, J.; Li, J.; Wang, Y.; Lin, Y.; Deng, Y.; Zhou, B.; et al. A functional
polymorphism in IFNAR1 gene is associated with susceptibility and severity of HFMD with EV71 infection.
Sci. Rep. 2015,5, 18541. [CrossRef] [PubMed]
91.
Zhu, F.; Xu, W.; Xia, J.; Liang, Z.; Liu, Y.; Zhang, X.; Tan, X.; Wang, L.; Mao, Q.; Wu, J.; et al. Efficacy, safety,
and immunogenicity of an enterovirus 71 vaccine in china. N. Engl. J. Med.
2014
,370, 818–828. [CrossRef]
[PubMed]
92.
Li, R.; Liu, L.; Mo, Z.; Wang, X.; Xia, J.; Liang, Z.; Zhang, Y.; Li, Y.; Mao, Q.; Wang, J.; et al. An inactivated
enterovirus 71 vaccine in healthy children. N. Engl. J. Med. 2014,370, 829–837. [CrossRef] [PubMed]
93.
Wei, M.; Meng, F.; Wang, S.; Li, J.; Zhang, Y.; Mao, Q.; Hu, Y.; Liu, P.; Shi, N.; Tao, H.; et al. 2-year efficacy,
immunogenicity, and safety of vigoo enterovirus 71 vaccine in healthy Chinese children: A randomized
open-label study. J. Infect. Dis. 2017,215, 56–63. [CrossRef] [PubMed]
94.
Yi, E.J.; Shin, Y.J.; Kim, J.H.; Kim, T.G.; Chang, S.Y. Enterovirus 71 infection and vaccines. Clin. Exp. Vaccine Res.
2017,6, 4–14. [CrossRef] [PubMed]
95.
Zhang, Q.; Zhao, B.; Chen, X.; Song, N.; Wu, J.; Li, G.; Yu, P.; Han, Y.; Liu, J.; Qin, C. GS-9620 inhibits
enterovirus 71 replication mainly through the NF-kB and PI3K-AKT signaling pathways. Antivir. Res.
2018
,
153, 39–48. [CrossRef] [PubMed]
96.
Huang, X.; Zhang, X.; Wang, F.; Wei, H.; Ma, H.; Sui, M.; Lu, J.; Wang, H.; Dumler, J.S.; Sheng, G.; et al.
Clinical efficacy of therapy with recombinant human interferon alpha1b in hand, foot, and mouth disease
with enterovirus 71 infection. PLoS ONE 2016,11, e0148907.
97.
Qiu, S.; Liu, N.; Jia, L.; Yang, G.; Su, W.; Li, J.; Song, L.; Yang, C.; Wang, J.; Zhang, C.; et al. A new treatment
for neurogenic inflammation caused by EV71 with CR2-targeted complement inhibitor. Virol. J.
2012
,9, 285.
[CrossRef] [PubMed]
98.
Dai, W.; Wu, Y.; Bi, J.; Lu, X.; Hou, A.; Zhou, Y.; Sun, B.; Kong, W.; Barbier, J.; Cintrat, J.-C.; et al. Antiviral
effects of Retro-2cycl and Retro-2.1 against Enterovirus 71
in vitro
and
in vivo
.Antivir. Res.
2017
,144,
311–321. [CrossRef] [PubMed]
99.
Zhang, G.; Zhou, F.; Gu, B.; Ding, C.; Feng, D.; Xie, F.; Wang, J.; Zhang, C.; Cao, Q.; Deng, Y.; et al.
In vitro
and
in vivo
evaluation of ribavirin and pleconaril antiviral activity against enterovirus 71 infection. Arch. Virol.
2012,157, 669–679. [CrossRef] [PubMed]
100.
Li, M.; Yang, X.; Guan, C.; Wen, T.; Duan, Y.; Zhang, W.; Li, X.; Wang, Y.; Zhao, Z.; Liu, S. Andrographolide
sulfonate reduces mortality in enterovirus 71 infected mice by modulating immunity. Int. Immunopharmacol.
2018,55, 142–150. [CrossRef] [PubMed]
101.
Liu, J.; Li, X.; Fan, X.; Ma, C.; Qin, C.; Zhang, L. Adoptive transfer of macrophages from adult mice reduces
mortality in mice infected with human enterovirus 71. Arch. Virol.
2013
,158, 387–397. [CrossRef] [PubMed]
102.
Ma, J.W.; Huang, B.S.; Hsu, C.W.; Peng, C.W.; Cheng, M.L.; Kao, J.Y.; Way, T.D.; Yin, H.C.; Wang, S.S. Efficacy
and safety evaluation of a chlorine dioxide solution. Int. J. Environ. Res. Public Health
2017
,14, 329. [CrossRef]
[PubMed]
103.
Wong, T.W.; Huang, H.J.; Wang, Y.F.; Lee, Y.P.; Huang, C.C.; Yu, C.K. Methylene blue-mediated photodynamic
inactivation as a novel disinfectant of enterovirus 71. J. Antimicrob. Chemother.
2010
,65, 2176–2182. [CrossRef]
[PubMed]
104.
Zhou, B.; Xu, L.; Zhu, R.; Tang, J.; Wu, Y.; Su, R.; Yin, Z.; Liu, D.; Jiang, Y.; Wen, C.; et al. A bispecific broadly
neutralizing antibody against enterovirus 71 and coxsackievirus A16 with therapeutic potential. Antivir. Res.
2019,161, 28–35. [CrossRef] [PubMed]
105.
Chang, H.-W.; Lin, Y.-W.; Ho, H.-M.; Lin, M.-H.; Liu, C.-C.; Shao, H.-Y.; Chong, P.; Sia, C.; Chow, Y.-H.
Protective efficacy of VP1-specific neutralizing antibody associated with a reduction of viral load and
pro-inflammatory cytokines in human SCARB2-transgenic mice. PLoS ONE
2013
,8, e69858. [CrossRef]
[PubMed]
106.
Wang, K.-T.; Lin, S.-J.; Wang, H.-C.; Chen, P.-C.; Lin, J.-J.; Chiang, J.-R.; Chang, C.-L.; Shih, D.Y.-C.; Lo, C.-F.;
Wang, D.-Y. Establishment of an animal challenge model as a potency assay for an inactivated enterovirus
type 71 vaccine. Biologicals 2016,44, 183–190. [CrossRef] [PubMed]
107.
Cao, R.-Y.; Han, J.-F.; Jiang, T.; Tian, X.; Yu, M.; Deng, Y.-Q.; Qin, E.D.; Qin, C.-F.
In vitro
and
in vivo
characterization of a new enterovirus type 71-specific human intravenous immunoglobulin manufactured
from selected plasma donors. J. Clin. Virol. 2011,51, 246–249. [CrossRef] [PubMed]
Viruses 2018,10, 674 20 of 20
108.
Xu, F.; Yao, P.-P.; Xia, Y.; Qian, L.; Yang, Z.-N.; Xie, R.-H.; Sun, Y.-S.; Lu, H.-J.; Miao, Z.-P.; Li, C.; et al.
Enterovirus 71 infection causes severe pulmonary lesions in gerbils, meriones unguiculatus, which can
be prevented by passive immunization with specific antisera. PLoS ONE
2015
,10, e0119173. [CrossRef]
[PubMed]
109.
Ho, B.-C.; Yu, I.S.; Lu, L.-F.; Rudensky, A.; Chen, H.-Y.; Tsai, C.-W.; Chang, Y.-L.; Wu, C.-T.; Chang, L.-Y.;
Shih, S.-R.; et al. Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of
type I interferon. Nat. Commun. 2014,5, 3344. [CrossRef] [PubMed]
©
2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... To establish an animal model, the main optimization factors are virus strains, species of mice, infection route, inoculation dose, and age of mice. Several mouse models of HFMD have been established, but the main infection routes are intraperitoneal (i.p.), intramuscular (i.m.), and intracranial (i.c.) [13]. Nevertheless, since those routes are not the natural infection route, the application will be limited if we establish the CVA19 mouse model imitating these infection routes. ...
... Researchers have tried to address this problem by using transgenic mice [14], immunodeficiency mouse [15], and mouse-adapted strains [16] or the combination of the above approaches. Transgenic mice are of producing high cost, and the immunodeficient mice hardly reflect the real infection due to defective immune systems [13]. In this study, we isolated an emerging CVA19 strain from a HFMD patient, naturally adapted to mice, and successfully established an orally infected CVA19 mouse model based on 7-day-old ICR mice, which manifested diarrhoea and encephalomyelitis similar to human infection. ...
... In our present study, we successfully simulated diarrhoea of human infection and the faeces exhibited yellow, soft, or unformed. Further studies provided sufficient evidences of small intestinal injury leading to diarrhoea, such as the colonization and proliferation of CVA19 in the intestinal tract, shortened intestinal villi, increased the number of secretory cells and apoptotic small intestinal cells, and inflammatory infiltration, which was similar with the findings of other enterovirus-associated models [13]. The GI tract is covered by epithelium comprising various cell types, including absorptive columnar epithelial cells, goblet cells, which may be permissive to CVA19 infection. ...
Article
Full-text available
Coxsackievirus A19 (CVA19) is a member of Enterovirus (EV) C group in the Picornaviridae family. Recently, we reported a case of CVA19-infected hand, foot, and mouth disease (HFMD) for the first time. However, the current body of knowledge on the CVA19 infection, particularly the pathogenesis of encephalomyelitis and diarrhea is still very limited, due to the lack of suitable animal models. Here, we successfully established a CVA19 mouse model via oral route based on 7-day old ICR mice. Our results found the virus strain could directly infect the neurons, astrocytes of brain and motor neurons of spinal cord causing neurological complications, such as acute flaccid paralysis (AFP). Importantly, viruses isolated from the spinal cords of infected mice caused severe illness in suckling mice, fulfilling Koch's postulates to some extent. CVA19 infection led to diarrhea with typical pathological features of shortened intestinal villi, increased number of secretory cells and apoptotic intestinal cells, and inflammatory cell infiltration. Much higher concentrations of serum cytokines and more peripheral blood inflammatory cells in CVA19-infected mice indicated a systematic inflammatory response induced by CVA19 infection. Finally, we found ribavirin and CVA19 VP1 monoclonal antibody could not prevent the disease progression, but higher concentrations of antisera and interferon alpha 2 (IFN-α2) could provide protective effects against CVA19. In conclusion, this study shows that a natural mouse-adapted CVA19 strain leads to diarrhea and encephalomyelitis in a mouse model via oral infection, which provides a useful tool for studying CVA19 pathogenesis and evaluating the efficacy of vaccines and antivirals.
... SPOP mutations in cells could affect the degradation of EV71-2A pro and therefore could exacerbate EV71 infection. However, a dearth of dependable animal models exists for the examination of disease manifestations in humans caused by EV71 infection (64,65). Notably, the induction of EV71 infection in mice under experimental conditions was contingent upon both the age of the mice and the dosage of the infection (66,67). ...
... Regrettably, none of the existing mouse models demonstrate pulmonary edema (72). Consequently, the utility of these murine models in investigating the disease mechanism of EV71 is limited (64). Consequently, we anticipate the development of a mouse model that mimics the infection mechanism observed in humans, which would enable the confirmation of SPOP function in vivo during EV71 infection using SPOP-deficient mice. ...
Article
Full-text available
The 2A protease (2A pro ) encoded by enterovirus 71 (EV71) serves as an accessory protein with significant involvement in the regulation of EV71 infection and viral replication. EV71-2A pro exhibits the ability to suppress various host factors, thereby disrupting the cellular antiviral immune response. However, whether host factors downregulate EV71-2A pro remains largely unknown. Here, we discovered that the speckle-type POZ protein (SPOP), functioning as a host E3 ubiquitin ligase, triggers ubiquitination modifications and subsequent degradation of EV71-2A pro . SPOP interacts with EV71-2A pro and exhibits a dose-dependent downregulation of EV71-2A pro levels. Conversely, knockdown of endogenous SPOP upregulated EV71-2A pro levels. Subsequent investigations revealed that the SPOP facilitates the lysosome-dependent degradation of EV71-2A pro by inducing K48-linked polyubiquitination of EV71-2A pro , which ultimately restricts EV71 replication. These findings shed light on the ubiquitination and lysosome-dependent regulation of the crucial EV71-encoded protease, offering a potential therapeutic target for the treatment of EV71 infection. IMPORTANCE EV71 poses a significant health threat to children aged 5 and below. The process of EV71 infection and replication is predominantly influenced by ubiquitination modifications. Our previous findings indicate that EV71 prompts the activation of host deubiquitinating enzymes, thereby impeding the host interferon signaling pathway as a means of evading the immune response. Nevertheless, the precise mechanisms by which the host employs ubiquitination modifications to hinder EV71 infection remain unclear. The present study demonstrated that the nonstructural protein 2A pro , which is encoded by EV71, exhibits ubiquitination and degradation mediated by the host E3 ubiquitin ligase SPOP. In addition, it is the first report, to our knowledge, that SPOP is involved in the host antiviral response.
... Animal models are excellent tools to study mechanisms of viral infection, as they often accurately simulate human pathogenesis. Humans are the only known natural hosts of EV-A71; however, a variety of animal models have been developed, including mouse, cynomolgus, rhesus, and green monkey (Shih et al., 2018;Wang and Yu, 2014). Of these models, mice have been widely used by researchers because of their low cost and ease of use. ...
... To date, a variety of murine EV-A71 models have been reported, including immunocompetent, immunodeficient, transgenic, hybrid, and humanized mice (Ke et al., 2019;Shih et al., 2018;Wang and Yu, 2014). Because neonatal mice are susceptible to EV-A71 clinical strains and develop objective phenotypes, such as limb paralysis and death, neonatal mice are widely used to study EV-A71. ...
Article
Full-text available
Enterovirus A71 (EV-A71) is neurotropic and one of the primary enteric pathogens responsible for severe central nervous system infection in infants and young children. Neonatal mice are ideal models for studying the pathogenesis of infection caused by EV-A71. In this study, we assessed the susceptibility of neonatal BALB/c, C57BL/6, ICR, Kunming, and NIH mice to a clinically isolated EV-A71 strain. One-day-old mice were challenged with a clinical isolate of EV-A71 via intraperitoneal injection, then observed for 13 days for mortality, body-weight changes, and limb paralysis. RT-qPCR was performed to quantify viral RNA in the brain, spinal cord, skeletal muscle, and lungs of BALB/c and C57BL/6 mice. The expression of murine scavenger receptor class B member 2 (mSCARB2) was measured by western blotting. Finally, lesions were assessed by histological examination. We found that neonatal BALB/c and C57BL/6 mice were both susceptible to EV-A71, leading to decreased survival rate, greater body weight loss, and prominent hind-limb paralysis. Tissue viral loads of C57BL/6J mice were markedly higher than those of BALB/c mice, indicating that EV-A71 replicated more efficiently in C57 mice. Increased expression of mSCARB2 was observed 5 days after infection in C57BL/6 mice, which coincided with the peak in EV-A71 replication. Histological examination indicated that infection caused obvious pathogenic lesions. In conclusion, C57BL/6 are most susceptible to infection caused by EV-A71 and can be used as a model for studying its pathogenesis and test therapeutic options.
... Studies have indicated that children infected with EV71 may experience suppression of innate immune, possibly due to interferon blockade (Coyne et al., 2013;Lei et al., 2013). Mice lacking interferon receptors and infected with EV71 exhibit symptoms such as limb paralysis and mortality, indicating the crucial role of interferons in preventing EV71 infection and disease progression (Liou et al., 2016;Shih et al., 2018). Furthermore, the immune-related protein TRAF3 interacting protein 3 (TRAF3IP3) has been shown to inhibit EV71 replication, while the non-structural protein 3C of EV71 can counteract this inhibition through protein cleavage . ...
Article
Full-text available
Hand, foot, and mouth disease (HFMD) is a common infectious disease caused by enterovirus 71 (EV71) that frequently affects children, leading to severe infections in some cases. In general, when infection occurs, the body upregulates inflammatory responses to eliminate pathogenic microorganisms to protect the host from infection. However, EV71 may inhibit host’s innate immunity to promote virus infection. At present, it is not fully understood how EV71 hijack the host cells for its own replication. Toll-like receptor 4 (TLR4), a natural immune receptor, historically associated with bacterial endotoxin-induced inflammatory responses. However, it is still unclear whether and how TLR4 is altered during EV71 infection. In this study, we observed a reduction in both TLR4 protein and gene transcript levels in RD, GES-1, and Vero cells following EV71 infection, as detected by RT-qPCR, immunofluorescence staining and western blot. Furthermore, we observed that the TLR4 downstream molecules of MYD88, p-NF-κB p65, p-TBK1 and related inflammatory cytokines were also reduced, suggesting that antiviral innate immune and inflammatory response were suppressed. To determine the impact of TLR4 changes on EV71 infection, we interfered EV71-infected RD cells with TLR4 agonist or inhibitor and the results showed that activation of TLR4 inhibited EV71 replication, while inhibition of TLR4 promote EV71 replication. Besides, EV71 replication was also promoted in TLR4 siRNA-transfected and EV71-infected RD cells. This suggests that down-regulation the expression of TLR4 by EV71 can inhibit host immune defense to promote EV71 self-replication. This novel mechanism may be a strategy for EV71 to evade host immunity.
... The lack of animal models is also a reason that limits the development of drugs. 172,173 Additionally, drug development is a cumbersome process, a long cycle, and closely related to the application of technology. Approved drugs already have extensive clinical trial data and experience. ...
Article
Full-text available
Enterovirus A71 (EV-A71) is one of the major causative agents of hand, foot, and mouth disease (HFMD) that majorly affects children. Most of the time, HFMD is a mild disease but can progress to severe complications, such as meningitis, brain stem encephalitis, acute flaccid paralysis, and even death. HFMD caused by EV-A71 has emerged as an acutely infectious disease of highly pathogenic potential in the Asia-Pacific region. In this review, we introduced the properties and life cycle of EV-A71, and the pathogenesis and the pathophysiology of EV-A71 infection, including tissue tropism and host range of virus infection, the diseases caused by the virus, as well as the genes and host cell immune mechanisms of major diseases caused by enterovirus 71 (EV-A71) infection, such as encephalitis and neurologic pulmonary edema. At the same time, clinicopathologic character�istics of EV-A71 infection were introduced. There is currently no specific medication for EV-A71 infection, highlighting the urgency and significance of developing suitable antieEV-A71 agents. This overview also summarizes the targets of existing antieEV-A71 agents, including virus entry, translation, polyprotein processing, replication, assembly and release; interferons; interleukins; the mitogen-activated protein kinase, phosphatidylinositol 3-kinase, and protein kinase B signaling pathways; the oxidative stress pathway; the ubiquitin-proteasome system; and so on. Furthermore, it overviews the effects of natural products, monoclonal antibodies, and RNA interference against EV-A71. It also discusses issues limiting the research of antiviral drugs. This review is a systematic and comprehensive summary of the mechanism and pathological characteristics of EV-A71 infection, the latest progress of existing antieEV-A71 agents. It would provide better understanding and guidance for the research and application of EV-A71 infection and antiviral inhibitors.
... EV71 is a small and non-enveloped virus with a positive-sense singlestranded RNA genome of 7500-8000 nucleotides that is composed of a long open reading frame (ORF) flanked by 5 ′ and 3 ′ nontranslated regions (NTR) (Lu et al., 2021;Shih et al., 2018;Yang et al., 2022). First, virus particle attaches and enters into host cells via specific receptors, and then the viral RNA genome is released into the cytoplasm. ...
Article
Full-text available
A series of 2-Benzoxyl-Phenylpyridine derivatives were evaluated for their potential antiviral activities against EV71. The preliminary assays indicated that some of these compounds exhibited excellent antiviral effects on EV71, they could effectively inhibit virus-induced cytopathic effects (CPEs), reduce progeny viral yields, and present similar or better antiviral activities compared to the positive control drug ribavirin. Among these derivatives, compounds WY7, WY13 and WY14 showed the most potency against EV71. Investigation of the underlying mechanism of action revealed that these compounds target EV71 replication in cells post infection, they could profoundly inhibit viral RNA replication and protein synthesis, and inhibit virus-induced cell apoptosis. Further experiments demonstrated that compound WY7 potently inhibited the activity of the EV71 3C protease (3Cpro), and to some extent, it affected the activity of 3D polymerase (3Dpol), thus blocking viral replication, but not the activity of the 2A proteinase (2Apro). Modeling of the molecular binding of the 3Cpro-WY7 complex revealed that compound WY7 was predicted to insert into the substrate-binding pocket of EV71 3Cpro, blocking substrate recognition and thereby inhibiting EV71 3Cpro activity. These results indicate that these compounds might be feasible therapeutic agents against EV71 infection and that these compounds may provide promising lead scaffolds for the further design and synthesis of potential antiviral agents.
... THP-1 cells could be infected by EV-A71. 49 We therefore investigated whether EV-A71 infection could prime STING mediated immune activation. As shown in Fig. 1e, f, EV-A71 infection triggered STING activation as indicated by phosphorylation at Ser366. ...
Article
Full-text available
Innate immunity represents one of the main host responses to viral infection.1–3 STING (Stimulator of interferon genes), a crucial immune adapter functioning in host cells, mediates cGAS (Cyclic GMP-AMP Synthase) sensing of exogenous and endogenous DNA fragments and generates innate immune responses.4 Whether STING activation was involved in infection and replication of enterovirus remains largely unknown. In the present study, we discovered that human enterovirus A71 (EV-A71) infection triggered STING activation in a cGAS dependent manner. EV-A71 infection caused mitochondrial damage and the discharge of mitochondrial DNA into the cytosol of infected cells. However, during EV-A71 infection, cGAS-STING activation was attenuated. EV-A71 proteins were screened and the viral protease 2Apro had the greatest capacity to inhibit cGAS-STING activation. We identified TRAF3 as an important factor during STING activation and as a target of 2Apro. Supplement of TRAF3 rescued cGAS-STING activation suppression by 2Apro. TRAF3 supported STING activation mediated TBK1 phosphorylation. Moreover, we found that 2Apro protease activity was essential for inhibiting STING activation. Furthermore, EV-D68 and CV-A16 infection also triggered STING activation. The viral protease 2Apro from EV-D68 and CV-A16 also had the ability to inhibit STING activation. As STING activation prior to EV-A71 infection generated cellular resistance to EV-A71 replication, blocking EV-A71-mediated STING suppression represents a new anti-viral target.
... Low cellular toxicity of some synthetic molecule peptideshas been observed when used as antivirals against EV71. Rupintrivir is an important synthetic antiviral agent which can promote the irreversible peptidomimetic inhibitors based on arestrictionprocessand inhibit viral replication [108]. Moreover, the synthetic component Rupintrivir can inhibit the action of VP1 to inhibit the host-pathogen interaction and relative responses. ...
Article
Full-text available
Enteroviruses are members of the Picornaviridae family consisting of human enterovirus groups A, B, C, and D as well as nonhuman enteroviruses. Human enterovirus type 71 (EV71) has emerged as a major cause of viral encephalitis, known as hand, foot, and mouth disease (HFMD), in children worldwide, especially in the Asia-Pacific region. EV71 and coxsackievirus A16 are the two viruses responsible for HFMD which are members of group A enteroviruses. The identified EV71 receptors provide useful information for understanding viral replication and tissue tropism. Host factors interact with the internal ribosome entry site (IRES) of EV71 to regulate viral translation. However, the specific molecular features of the respective viral genome that determine virulence remain unclear. Although a vaccine is currently approved, there is no effective therapy for treating EV71-infected patients. Therefore, understanding the host-pathogen interaction could provide knowledge in viral pathogenesis and further benefits to anti-viral therapy development. The aim of this study was to investigate the latest findings about the interaction of viral ligands with the host receptors as well as the activation of immunerelated signaling pathways for innate immunity and the involvement of different cytokines and chemokines during host-pathogen interaction. The study also examined the roles of viral proteins, mainly 2A and 3C protease, interferons production and their inhibitory effects.
Article
Enterovirus 71 (EV71) is a major causative agent of hand, foot, and mouth disease (HFMD) in children. Nowadays, there are still no effective antiviral drugs for EV71 infection. High mobility group box 1 (HMGB1) is reported to be highly expressed in HFMD patients. However, the role and underlying mechanism of HMGB1 in EV71‐associated HFMD are still unclear. HMGB1 expression was detected using RT‐qPCR and western blot assays. Loss‐ and gain‐function experiments were performed to evaluate the effects of HMGB1 on EV71‐infected cells. The virus titer was examined by TCID50. CCK‐8 and flow cytometry assays were applied to detect the cell viability and cell cycle. Oxidative stress was determined by relative commercial kits. HMGB1 level was elevated in the serum of EV71‐infected patients with HFMD and EV71‐induced RD cells. EV71 infection induced the transfer of HMGB1 from the nucleus into the cytoplasm. HMGB1 knockdown inhibited virus replication, viral protein (VP1) expression and promoted antiviral factor expression. In addition, the inhibition of HMGB1 improved cell viability, protected against S phase arrest, and inhibited EV71‐induced cell injury and oxidative stress, whereas HMGB1 overexpression showed the opposite effects. In terms of mechanism, HMGB1 overexpression activated the TLR4/NF‐κB/NLRP3 signaling pathway and promoted cell pyroptosis. The inhibition of TLR4 and NF‐κB reversed the effects of HMGB1 overexpression on virus replication, oxidative stress, and pyroptosis. In conclusion, HMGB1 knockdown inhibits EV71 replication and attenuates pyroptosis through TLR4/NF‐κB/NLRP3 axis.
Article
Full-text available
As a major component of immunological defense against a great variety of pathogens, innate immunity is capable of activating the adaptive immune system. Viruses are a type of pathogen that proliferate parasitically in cells and have multiple strategies to escape from host immune pressure. Here, we review recent studies of the strategies and mechanisms by which enteroviruses evade innate immune monitoring.
Article
Full-text available
Enterovirus 71 (EV71) causes hand, foot and mouth disease, a mild and self-limited illness that is sometimes associated with severe neurological complications. EV71 neurotropic determinants remain ill-defined to date. We previously identified a mutation in the VP1 capsid protein (L97R) that was acquired over the course of a disseminated infection in an immunocompromised host. The mutation was absent in the respiratory tract but was present in the gut (as a mixed population) and in blood and cerebrospinal fluid (as a dominant species). In this study, we demonstrated that this mutation does not alter the dependence of EV71 on the human scavenger receptor class B2 (SCARB2), while it enables the virus to bind to the heparan sulfate (HS) attachment receptor and modifies viral tropism in cell lines and in respiratory, intestinal and neural tissues. Variants with VP197L or VP197R were able to replicate to high levels in intestinal and neural tissues and, to a lesser extent, in respiratory tissues, but their preferred entry site (from the luminal or basal tissue side) differed in respiratory and intestinal tissues and correlated with HS expression levels. These data account for the viral populations sequenced from the patient’s respiratory and intestinal samples and suggest that improved dissemination, resulting from an acquired ability to bind HS, rather than specific neurotropism determinants, enabled the virus to reach and infect the central nervous system. Finally, we showed that iota-carrageenan, a highly sulfated polysaccharide, efficiently blocks the replication of HS-dependent variants in cells and 2D neural cultures. Overall, the results of this study emphasize the importance of HS binding in EV71 pathogenesis and open new avenues for the development of antiviral molecules that may prevent this virus’s dissemination.
Article
Full-text available
Enterovirus 71 (EV71) is an aetiological agent responsible for seasonal epidemics of hand-foot-and-mouth disease, which causes considerable mortality among young children. Mucosal vaccines can efficiently induce secretory IgA at mucosal surfaces and thereby prevent or limit infection at the site of virus entry. CpG oligodeoxynucleotides (ODNs), which resemble bacterial DNA, can induce the innate immune response through activation of Toll-like receptor 9. Here, we used CpG ODNs as adjuvants to investigate an EV71 mucosal vaccine in mice. In the EV71 + CpG group, the EV71-specific IgG and IgA titres in the serum, nasal wash, bronchoalveolar lavage fluid, and faeces were substantially higher than those in the EV71- and phosphate-buffered saline-treated groups. Moreover, the number of EV71-specific IgG- and IgA-producing cells was also higher in the EV71 + CpG group. Furthermore, T-cell proliferative responses and interleukin-17 secretion were markedly increased when CpG-adjuvanted EV71 was delivered intranasally. More importantly, the induced antibodies neutralised infection by EV71 of the C2 genotype and crossneutralised infection by EV71 of the B4 and B5 genotypes. Lastly, human scavenger receptor class B, member 2-transgenic mice intranasally immunised with the CpG-adjuvanted EV71 vaccine resisted a subsequent lethal challenge with EV71, indicating that CpG was an effective intranasal adjuvant for EV71 mucosal-vaccine development.
Article
Full-text available
Attachment receptors expressed on the host cell surface are thought to enhance EV71 infection by increasing the chance of encountering true receptors. Although this has been confirmed using cell culture for some viruses, the importance of attachment receptors in vivo is unknown. This report provides an unexpected answer to this question. We demonstrated that the VP1-145G virus binds to HS and shows an attenuated phenotype in an hSCARB2-dependent animal infection model. HS is highly expressed in cells that express hSCARB2 at low or undetectable levels. Our data indicate that HS binding directs VP1-145G virus toward abortive infection and keeps virus away from hSCARB2-positive cells. Thus, although the ability of VP1-145G virus to use HS might be an advantage in replication in certain cultured cells, it becomes a serious disadvantage in replication in vivo . This adsorption is thought to be a major mechanism of attenuation associated with attachment receptor usage.
Article
Full-text available
EV71 is a positive-sense single-stranded RNA virus that belongs to the Picornaviridae family. EV71 infection may cause various symptoms ranging from hand-foot-and-mouth disease to neurological pathological conditions such as aseptic meningitis, ataxia, and acute transverse myelitis. There is currently no effective treatment or vaccine available. Various compounds have been examined for their ability to restrict EV71 replication. However, most experiments have been performed in rhabdomyosarcoma or Vero cells. Since the gastrointestinal tract is the entry site for this pathogen, we anticipated that orally ingested agents may exert beneficial effects by decreasing virus replication in intestinal epithelial cells. In this study, curcumin (diferuloylmethane, C21H20O6), an active ingredient of turmeric (Curcuma longa Linn) with anti-cancer properties, was investigated for its anti-enterovirus activity. We demonstrate that curcumin treatment inhibits viral translation and increases host cell viability. Curcumin does not exert its anti-EV71 effects by modulating virus attachment or virus internal ribosome entry site (IRES) activity. Furthermore, curcumin-mediated regulation of mitogen-activated protein kinase (MAPK) signaling pathways is not involved. We found that protein kinase C delta (PKCδ) plays a role in virus translation in EV71-infected intestinal epithelial cells and that curcumin treatment decreases the phosphorylation of this enzyme. In addition, we show evidence that curcumin also limits viral translation in differentiated human intestinal epithelial cells. In summary, our data demonstrate the anti-EV71 properties of curcumin, suggesting that ingestion of this phytochemical may protect against enteroviral infections.
Article
Full-text available
Enterovirus 71 (EV71) is a global infectious disease that affects millions of people. The virus is the main etiological agent for hand, foot, and mouth disease with outbreaks and epidemics being reported globally. Infection can cause severe neurological, cardiac, and respiratory problems in children under the age of 5. Despite on-going efforts, little is known about the pathogenesis of EV71, how the host immune system responds to the virus and the molecular mechanisms behind these responses. Moreover, current animal models remain limited, because they do not recapitulate similar disease patterns and symptoms observed in humans. In this review the role of the host–viral interactions of EV71 are discussed together with the various models available to examine: how EV71 utilizes its proteins to cleave host factors and proteins, aiding virus replication; how EV71 uses its own viral proteins to disrupt host immune responses and aid in its immune evasion. These discoveries along with others, such as the EV71 crystal structure, have provided possible targets for treatment and drug interventions.
Article
Full-text available
Enterovirus 71 (EV71) is a common etiological agent of hand, foot, and mouth disease and fatal neurological diseases in children. The neuropathogenicity of severe EV71 infection has been documented, but studies comparing mouse models of severe and mild EV71 infection are lacking. The aim of the study was to investigate the neurovirulence of EV71 strains and the differences in serum cytokine and chemokine levels in mouse models of severe and mild EV71 infection. Nine EV71 isolates belonging to the C4 subgenogroup (proposed as genotype D) displayed infectivity in human neuroblastoma SK-N-SH cells; moreover, ultrastructural observation confirmed viral particle replication. The survival rate of the severe model was 71.43% (5/7), and 60% (3/5) of the surviving severe model mice displayed sequelae of paralysis, whereas the only symptom in mild model mice was ruffled fur. Dynamic detection of serum cytokine and chemokine levels demonstrated that interleukin (IL)-5, IL-13, IL-6, monocyte chemotactic protein 1 (MCP-1), and chemokine (C-C motif) ligand 5 (also called Regulated upon Activation, Normal T-cell Expressed, and Secreted (CCL5/RANTES) were significantly up-regulated at the early period of infection, indicating that these factors might herald a severe outcome. Our findings suggest that elevated cytokines and chemokines may have potential value as prognostic markers in mouse models.
Article
Enterovirus 71 (EV71) and coxsackievirus A16 (CA16) are the major pathogens of hand, foot and mouth disease (HFMD), which affects children worldwide and is often associated with neurological complications. At present, there is no vaccine or cure available for simultaneous EV71 and CA16 infection, posing a great need to develop novel strategies for the treatment of this disease. Here, we engineered four bispecific antibodies using variable fragments of monoclonal antibodies (mAbs) from EV71- and CA16-specific neutralizing antibodies. The engineered bispecific antibody Bs(scFv)4-IgG-1 exhibits remarkable cross-reactivity against EV71 and CA16 and has a more potent cross-neutralization than its parental antibodies. Furthermore, we showed that Bs(scFv)4-IgG-1 conferred 100% therapeutic efficacy against single or mixed EV71 and CA16 infections in mice. Our study provides important insights into bispecific antibody engineering against enterovirus and will inform new curative treatment options for HFMD.
Article
Human enterovirus 71 (EV71) is the second most common cause of hand, foot, and mouth disease (HFMD), which can occur as a severe epidemic especially among children under 5-years old. New and improved treatment strategies to control EV71 infection are therefore urgently required. The heterocyclic compound GS-9620, a potent and selective agonist of Toll-like receptor 7 (TLR7), has been reported to activate plasmacytoid dendritic cells (pDCs), and suppress HBV as well as HIV replication. In this study, we indicated that GS-9620 also could inhibit EV71 replication in the mouse model of EV71 infection. With three-days treatment after EV71 infection, the levels of proinflammatory cytokines/chemokines, like IFN-α, IFN-γ and MCP-1, were sharply reduced in serum compared to those without treatment. Furthermore, GS-9620 activated TLR7 in the limb muscle cells, which stimulated the NF-κB and PI3K/AKT signaling pathways. When NF-κB or PI3K/AKT inhibitors were used, the antiviral effect of the GS-9620 was impacted. Overall, our data implied GS-9620 probably activates NF-κB and PI3K/AKT signaling pathways to clear the virus.
Article
Outbreaks of hand, foot and mouth disease (HFMD), which is caused by Enterovirus 71 (EV71), have erupted in recent years. Andrographolide sulfonate (Trade name: Xiyanping injection) has been recommended to treat severe HFMD in China because of its conventional antithermic and antitoxic activities, but its actual mechanism has not been revealed clearly until now. To explore its therapeutic efficacy and mechanism, a Xiyanping injection treatment mouse model was established. Based on the therapeutic model, routine clinical parameters and histopathologic changes were investigated, in the same time, viral loads, immune cells, inflammatory molecules and cell signaling pathways were determined. Xiyanping injection treatment protected mice from lethal EV71 challenge in a therapeutic regimen-dependent manner, which may mostly depend on its direct immunomodulatory activities on neutrophil and T lymphocyte. Reduced inflammatory molecular production of neutrophil and elevated T lymphocyte activity may result from its marked inhibition of some signaling pathways. Taken together, Xiyanping injection was an effective treatment for severe HFMD by improving hosts' immunity.