ArticlePDF Available

Evaluation of luciferase and GFP-expressing Nipah viruses for rapid quantitative antiviral screening

Authors:

Abstract and Figures

Nipah virus (NiV) outbreaks have occurred in Malaysia, India, and Bangladesh, and the virus continues to cause annual outbreaks of fatal human encephalitis in Bangladesh due to spillover from its bat host reservoir. Due to its high pathogenicity, its potential use for bio/agro-terrorism, and to the current lack of approved therapeutics, NiV is designated as an overlap select agent requiring biosafety level-4 containment. Although the development of therapeutic monoclonal antibodies and soluble protein subunit vaccines have shown great promise, the paucity of effective antiviral drugs against NiV merits further exploration of compound libraries using rapid quantitative antiviral assays. As a proof-of-concept study, we evaluated the use of fluorescent and luminescent reporter NiVs for antiviral screening. We constructed and rescued NiVs expressing either Renilla luciferase or green fluorescent protein, and characterized their reporter signal kinetics in different cell types as well as in the presence of several inhibitors. The 50 percent effective concentrations (EC50s) derived for inhibitors against both reporter viruses are within range of EC50s derived from virus yield-based dose-response assays against wild-type NiV (within 1 Log10), thus demonstrating that both reporter NiVs can serve as robust antiviral screening tools. Utilizing these live NiV-based reporter assays requires modest instrumentation, and circumvents the time and labor-intensive steps associated with cytopathic effect or viral antigen-based assays. These reporter NiVs will not only facilitate antiviral screening, but also the study of host cell components that influence the virus life cycle.
No caption available
… 
Content may be subject to copyright.
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/authorsrights
Author's personal copy
Evaluation of luciferase and GFP-expressing Nipah viruses for rapid
quantitative antiviral screening
Michael K. Lo
, Stuart T. Nichol, Christina F. Spiropoulou
Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
article info
Article history:
Received 8 February 2014
Revised 6 March 2014
Accepted 18 March 2014
Available online 27 March 2014
Keywords:
Henipavirus
Nipah virus
Antiviral screening
Luciferase
GFP
High-throughput screening
abstract
Nipah virus (NiV) outbreaks have occurred in Malaysia, India, and Bangladesh, and the virus continues to
cause annual outbreaks of fatal human encephalitis in Bangladesh due to spillover from its bat host res-
ervoir. Due to its high pathogenicity, its potential use for bio/agro-terrorism, and to the current lack of
approved therapeutics, NiV is designated as an overlap select agent requiring biosafety level-4 contain-
ment. Although the development of therapeutic monoclonal antibodies and soluble protein subunit vac-
cines have shown great promise, the paucity of effective antiviral drugs against NiV merits further
exploration of compound libraries using rapid quantitative antiviral assays. As a proof-of-concept study,
we evaluated the use of fluorescent and luminescent reporter NiVs for antiviral screening. We
constructed and rescued NiVs expressing either Renilla luciferase or green fluorescent protein, and
characterized their reporter signal kinetics in different cell types as well as in the presence of several
inhibitors. The 50% effective concentrations (EC
50
s) derived for inhibitors against both reporter viruses
are within range of EC
50
s derived from virus yield-based dose–response assays against wild-type NiV
(within 1Log
10
), thus demonstrating that both reporter NiVs can serve as robust antiviral screening tools.
Utilizing these live NiV-based reporter assays requires modest instrumentation, and circumvents the
time and labor-intensive steps associated with cytopathic effect or viral antigen-based assays. These
reporter NiVs will not only facilitate antiviral screening, but also the study of host cell components that
influence the virus life cycle.
Published by Elsevier B.V.
1. Introduction
Nipah virus (NiV) is a highly pathogenic paramyxovirus in the
genus Henipavirus of the subfamily Paramyxovirinae within the
family Paramyxoviridae (Rota and Lo, 2012). Humans are infected
upon spillover of NiV from its bat reservoir host or by person-to-
person transmission (Luby, 2013). Due to its high pathogenicity,
its potential use for bio/agro-terrorism, and to the current lack of
approved therapeutics, NiV is designated as an overlap select agent
requiring biosafety level-4 containment. Although several preven-
tative measures against exposure to bat saliva or urine have been
studied (Nahar et al., 2013), NiV continues to cause near-annual
outbreaks of fatal encephalitis in Bangladesh. The development
of therapeutic monoclonal antibodies and soluble subunit vaccines
against henipaviruses have shown great promise (Broder et al.,
2013), but screening available compound libraries for potentially
efficacious therapeutics against NiV remains a high priority for
investigation. Most high-throughput antiviral assays against NiV
were developed to screen for inhibitors of virus entry and/or cell-
to-cell fusion (Bossart et al., 2005; Porotto et al., 2009; Talekar
et al., 2012; Tamin et al., 2009), while the NiV minigenome assay
was used to screen for inhibitors of NiV replication (Freiberg
et al., 2008). A chemiluminescent immunodetection assay for live
henipavirus infection significantly streamlined antiviral screening
when compared with cytopathic effect (CPE) or immune plaque-
based assays, but had relatively limited signal-to-noise ratios
(<100), and also required cell fixation and multiple incubation
steps which are time and labor-intensive (Aljofan et al., 2008).
Reverse genetic systems for henipaviruses have enabled the gener-
ation of recombinant viruses expressing fluorescent or luminescent
proteins which allow for rapid detection of infection in vitro and
in vivo (Lo et al., 2012; Marsh et al., 2013; Yoneda et al., 2006).
We sought to determine whether NiVs expressing fluorescent or
luminescent reporters could facilitate rapid quantitative antiviral
screening. As a proof-of-concept study, we constructed and res-
cued NiVs expressing either Renilla luciferase (LUC2AM) or green
fluorescent protein (GFP2AM), characterized their reporter signal
http://dx.doi.org/10.1016/j.antiviral.2014.03.011
0166-3542/Published by Elsevier B.V.
Corresponding authors. Address: Centers for Disease Control and Prevention,
Viral Special Pathogens Branch, 1600 Clifton Road, Mailstop G-14, Atlanta, GA
30333, United States. Tel.:+1 4046390327 (M.K. Lo).
E-mail addresses: mko2@cdc.gov (M.K. Lo), ccs8@cdc.gov (C.F. Spiropoulou).
Antiviral Research 106 (2014) 53–60
Contents lists available at ScienceDirect
Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral
Author's personal copy
kinetics in different human cell types, and tested them in parallel
against cellular inhibitors of pyrimidine biosynthesis as well as in-
nate immune agonists. We show that both reporter viruses can be
used in the 96-well format and have excellent signal-to-noise ra-
tios and Z
0
-factors. Furthermore, the 50% effective concentrations
(EC
50
s) derived for inhibitors tested against both reporter viruses
were not only within close range of one another (within 4-fold),
but also of EC
50
s derived from virus yield-based dose–response as-
says against wild-type NiV (within 1 Log
10
), thus demonstrating
that both reporter NiVs can serve as robust antiviral screening
tools.
2. Materials and methods
2.1. Cells
Vero (African green monkey kidney), A549 (human lung epithe-
lial carcinoma), HeLa (human cervical carcinoma), and HEK 293T
(human embryonic kidney) cells were maintained at 37 °C and
5% CO
2
in Dulbecco’s modified Eagle’s medium supplemented with
7.5% fetal bovine serum (FBS), 100 U/mL penicillin, 100
l
g/mL
streptomycin, and 2 mM
L
-glutamine. Baby hamster kidney cells
stably expressing T7 polymerase (BSRT7/5) (Buchholz et al.,
1999) were incubated at every third passage in the presence of
1 mg/mL G-418 (Geneticin).
2.2. Cloning and rescue of recombinant viruses
For LUC2AM, a modified NiV M gene fragment expressing the
Renilla luciferase (Promega) open reading frame (ORF) in-frame
with the NiV Matrix (M) gene ORF separated by the FMDV 2A pro-
tease cleavage peptide was constructed using a 4-step overlapping
PCR strategy as described for a DsRed-Express reporter NiV (RE-
D2AM) and inserted into a plasmid containing a full-length cDNA
copy of wild-type recombinant NiV genome using engineered SbfI
and SacII restriction sites (Lo et al., 2012). The total genome length
of LUC2AM was 19,254 nt.
The GFP2AM expressing the green fluorescence protein
Zsgreen1 (Clontech) ORF in-frame with the NiV M gene ORF sepa-
rated by the porcine teschovirus 2A (p2A) protease cleavage pep-
tide was constructed using a second-generation wild-type
recombinant NiV (rNiV) (Genbank sequence AF212302) devoid of
engineered restriction sites, assembled via 4 fragments into the
same plasmid backbone used for LUC2AM (Genscript). A synthe-
sized fragment containing the GFP, p2A, and NiV M coding se-
quence in-frame was cloned into WT using PacI restriction sites.
An additional 3 nucleotides ‘‘GGA’’ encoding the amino acid glycine
were inserted between the end of the Zsgreen1 ORF and the start of
the p2A cleavage peptide for the final genome length to obey the
‘‘Rule of six’’ at 19,014 nt (Kolakofsky et al., 1998). Both LUC2AM
and GFP2AM were rescued as previously described (Lo et al.,
2012). The genomes of the rescued viruses were fully sequenced
with no undesired mutations. Virus stock titers were determined
by median tissue culture infectious dose (TCID
50
) assay (see
below).
2.3. Infections with recombinant reporter viruses, reporter activity
measurements, cell type optimization, TCID
50
All infections in this study were performed in 96-well plates.
Cells were seeded in opaque black or white plates (Costar 3916,
3917) for fluorescence and luminescence-based assays respec-
tively. Initial time course experiments for rescued viruses were
performed by infecting 2 10
4
Vero cells/well at multiplicity of
infection (MOI) = 0.01 TCID
50
/cell of either virus for 2 h. Virus
inoculum was removed and replaced with 100
l
L Fluorobrite med-
ium (Life Technologies) supplemented with 2 mM
L
-glutamine and
10% FBS. At 24, 48, and 72 h post-infection (hpi), supernatants
were harvested for TCID
50
determination and replaced with
Fluorobrite medium. Fluorescence activity (gain = 65) was mea-
sured in a H1 Synergy plate reader (Biotek), while luminescence
activity (gain = 107) was measured in the same reader 10 min after
addition of Renilla-Glo substrate (Promega). Subsequent infections
were performed at MOI = 0.2 TCID
50
/cell unless stated otherwise.
Cytopathic effect-based TCID
50
assays were performed by infecting
Vero cells in 10-fold dilution series of samples (10
1
-10
8
) in a 96-
well plate format, infecting 9 wells per sample and dilution step.
Plates were read after 7 days to ensure a clear distinction between
infected and uninfected wells at the highest dilutions.
2.4. Fluorescence microscopy
210
4
HeLa cells were infected with LUC2AM or GFP2AM, and
at 24 hpi were fixed in 10% formalin for 15 min, permeablized with
PBS + 0.4% Triton-X for 5 min, washed with PBS + 0.1% Tween
(PBS-T), and blocked with PBS-T + 5% milk (PBS-T-M) for 30 min.
LUC2AM-infected cells were incubated with a mouse anti-NiV-N
monoclonal antibody 1A11C1 (Chiang et al., 2010) and a rabbit
anti-Renilla luciferase antibody (MBL) diluted 1:1000 and 1:200
respectively in PBS-T-M for 1 h at 37 °C, while GFP2AM-infected
cells were incubated with only 1A11C1 antibody. After 3 washes
with PBS-T, cells were incubated with anti-mouse DyLight 550
and anti-rabbit DyLight 488 antibodies (Bethyl Laboratories, Inc.)
diluted 1:1000 in PBS-T-M for 30 min at 37 °C. After 3 washes, fluo-
rescence images were taken on a Zeiss Axiovert 200 microscope at
40X magnification.
2.5. Antiviral compounds, assessment of cell viability (CC
50
) and 50%
effective concentrations (EC
50
)
The following compounds used in this study were dissolved in
dimethyl sulfoxide (DMSO): 6-azauridine acetate (Sigma), Pyra-
zofurin (TRC), and compound 09167 (Vitas-M Laboratory) (Yan
et al., 2013). The final DMSO concentration in Fluorobrite media
was 0.1%. Universal interferon (U-IFN) (Sigma) was dissolved in
sterile distilled H
2
O. The CellTiter-Glo assay (Promega) was used
to determine viability of uninfected 293T cells exposed to 3-fold
serial dilutions of the compounds for 48 h. Values were normalized
to DMSO controls according to % viability as follows: % viabil-
ity = [(specific valuereference value)/(DMSO control value ref-
erence value)] 100. Reference values were derived from control
wells without cells. DMSO control values (after subtraction of ref-
erence values) were set at 100% viability. 50% viability/cytotoxicity
(CC
50
) and 50% effective concentrations (EC
50
) were calculated
using four-parameter variable slope non-linear regression fitting
of mean values for three experiments. Therapeutic indices (TI)
were represented as CC
50
/EC
50
. In instances which CC
50
could not
be properly calculated due to non-convergence of dose response
curves, we designated the highest concentration of compound used
as the CC
50
.
2.6. Statistical analysis
Statistical analysis was performed using Prism 6 software
(GraphPad Software). Z
0
(separation band/dynamic range of the as-
say) values were calculated based on the following formula:
Z
0
=1[(3SD
C
+ 3SD
B
)/(mean
C
mean
B
)], where SD is the stan-
dard deviation, Cis the control, and Bis the background (Zhang
et al., 1999). The percent coefficient of variation (%CV) was calcu-
lated as follows: %CV = SD
C
/mean
C
100.
54 M.K. Lo et al. / Antiviral Research 106 (2014) 53–60
Author's personal copy
3. Results and discussion
3.1. Construction and characterization of luciferase and GFP expressing
NiVs
To evaluate the use of luminescent reporters for antiviral
screening against NiV, we constructed a Renilla luciferase-
expressing NiV (LUC2AM) by employing previous methods used
to generate a red fluorescent protein-expressing NiV (RED2AM)
(Lo et al., 2012). We inserted the Renilla luciferase (LUC) gene into
the NiV matrix (M) gene 5
0
of the M ORF in the same reading frame,
separated by the coding sequence of the Foot and Mouth Disease
virus protease cleavage peptide (F2A) (Fig. 1A). We used the same
strategy to construct a second generation recombinant green fluo-
rescent protein(GFP)-expressing NiV (GFP2AM), except that we
used the more efficiently-cleaved porcine Teschovirus 2A cleavage
Fig. 1. Design and characterization of reporter Nipah (NiV) viruses. (A) Schematic of wild-type NiV genome alongside reporter NiVs expressing either luciferase (LUC2AM) or
green-fluorescent protein (GFP2AM) respective nucleotide (nt) lengths indicated. Black segments represent non-coding regions, while white segments indicate open reading
frames. Modified M gene coding regions for LUC2AM and GFP2AM are highlighted in yellow and green respectively. (B) Microscopic examination of HeLa cells infected with
LUC2AM and GFP2AM. LUC2AM-infected were stained with antibodies against NiV nucleoprotein (N) and anti-Renilla luciferase antibodies, while GFP2AM-infected cells
were stained only with N antibodies. (C) Time course analysis of growth kinetics and reporter activities of LUC2AM and GFP2AM alongside the growth kinetics of their
corresponding wild-type recombinant NiVs (WT, rNiV respectively). Vero cells were infected with each virus at an MOI of 0.01. At 1, 2, and 3 days post-infection, supernatants
titers were determined by TCID
50
, (left Y-axis) while relative light units (RLU) or relative fluorescence units (RFU) in the cells infected with reporter viruses were measured
(right Y-axis). Reporter activity levels at day 0 post-infection indicate background noise of the luminometer/fluorometer. Values represent means and standard deviations for
3 experiments.
M.K. Lo et al. / Antiviral Research 106 (2014) 53–60 55
Author's personal copy
peptide (p2A) coding sequence instead of F2A (Kim et al., 2011)
(see Section 2). We rescued both LUC2AM and GFP2AM and con-
firmed expression of their respective reporter proteins by immuno-
fluorescence microscopy. The distribution of both LUC and GFP
were distinct from NiV nucleoprotein, with the luciferase localizing
exclusively to the cytoplasm, and with the GFP occupying both nu-
clear and cytoplasmic compartments (Fig. 1B). While GFP2AM
grew similarly in Vero cells (MOI = 0.01 TCID
50
/cell) compared
with the non-reporter recombinant (rNiV) (Fig. 1C, right) LUC2AM
had an attenuated growth phenotype when compared with its cor-
responding first generation non-reporter recombinant NiV (WT)
(Fig. 1C, left). This is consistent with previous observations that
the RED2AM virus was moderately attenuated compared with
WT (Lo et al., 2012). Furthermore our result is consistent with
Fig. 2. Assay optimization. (A) 293T cells yielded the highest reporter activity levels for both LUC2AM and GFP2AM reporter viruses. (B) 293T cells were infected at the
indicated multiplicity of infection (MOI) with LUC2AM or GFP2AM, and respective reporter values were measured at 48 hpi. (C) Time course analysis of growth kinetics and
reporter activities of LUC2AM and GFP2AM in 293T cells (MOI = 0.2 TCID
50
/cell). At 1, 2, and 3 days post-infection, supernatant titers were determined by TCID
50
(left Y-axis),
while respective reporter activities in the cells were measured (right Y-axis). Values represent means and standard deviations for 3 experiments.
56 M.K. Lo et al. / Antiviral Research 106 (2014) 53–60
Author's personal copy
previous work showing that NiV M requires ubiquitination inside
the nucleus to become functional for budding (Wang et al.,
2010), since the comparatively less-efficient cleavage of F2A would
result in decreased amounts of functional NiV M for budding
new virus particles. In spite of this phenotype however, LUC2AM
generated increasing levels of LUC activity from 0 to 48 h post-
infection (hpi), and reached viral titers of over 10
6
TCID
50
/mL by
72 hpi (Fig. 1C, left panel). The fluorescence signal generated by
GFP2AM was detectable at 24 hpi, but increased by greater than
20-fold at 48 hpi, and continued to increase through 72 hpi. The
Fig. 3. Reporter NiVs as tools for antiviral testing. (A) Testing of 2 known inhibitors (6-azauridine acetate and pyrazofurin) of pyrimidine biosynthesis and (B) 2 innate
immune agonists (Universal interferon and compound 09167) against LUC2AM (left panels) and GFP2AM (right panels). 293T cells were infected with reporter NiVs
(MOI = 0.2 TCID
50
/cell) for 2 h, and then incubated with indicated compounds in 3-fold serial dilutions. Each concentration was assessed in three replicates. Reporter virus
activity was measured on the left Y-axis; % cell viability was measured on the right Y-axis. Values represent means and standard deviations of 3 experiments.
M.K. Lo et al. / Antiviral Research 106 (2014) 53–60 57
Author's personal copy
accumulation of GFP signal over the course of infection corre-
sponded with the increase in GFP2AM viral titer (Fig. 1C, right
panel).
3.2. Optimization of cell type and infection conditions for LUC2AM and
GFP2AM-based assays
We then optimized both assays for antiviral testing. We tested
each virus against 3 common human cell lines (A549, HeLa, 293T)
alongside Vero cells to determine which would yield the strongest
reporter signal over the course of infection. For negative controls,
we assayed for reporter activity in each cell line infected with
wild-type recombinant NiV (rNiV). Infection with LUC2AM over
the course of 72 h (MOI = 0.1 TCID
50
/cell) showed that HeLa and
293T cells supported peak LUC activity to approximately equal or
greater levels than in Vero cells, while A549 only marginally sup-
ported LUC activity (Fig. 2A, left panel). In both Vero and HeLa cells,
LUC signal peaked at 24 hpi (4–5 10
4
relative light units) and
steadily decreased by over 10-fold by 72 hpi. In 293T cells how-
ever, the LUC signal was approximately equal to those of Vero
and HeLa cells at 24 hpi, but proceeded to double by 48 hpi
(810
4
RLU) before decreasing at 72 hpi to levels similar to
24 hpi. Infection with GFP2AM yielded similar results, showing
minimal NiV replication in A549 cells, and indicating 293T cells
as the optimal cell type which generated the highest GFP signal on-
ward from 48 hpi to 72 hpi (Fig. 2A right panel). In contrast to the
LUC2AM infections however, all 4 infected cell lines attained their
highest levels of GFP expression at 72 hpi. The different reporter
signal kinetics observed when comparing LUC2AM and GFP2AM
infections are likely a function of respective reporter protein mat-
uration rates and half-lives. Whereas LUC matures rapidly and has
a protein half-life of approximately 14 h in the cell cytoplasm
(Loening et al., 2006), GFP requires 8–12 h for maturation, and
has a protein half-life of approximately 26 h (Corish and
Tyler-Smith, 1999). Since the peak LUC signal in infected 293T cells
occurred at 48 hpi, and because of the minimal difference in GFP
signal between 48 hpi and 72 hpi, we chose 48 hpi as our assay
time point for both LUC2AM and GFP2AM.
To further optimize these reporter assays, we infected 293T
cells with either reporter virus at MOIs ranging from 0.05 to
0.4 TCID
50
/cell. Increasing the MOI for both LUC2AM and GFP2AM
led to corresponding increases in reporter activity (Fig. 2B). Since
the increase in MOI from 0.2 to 0.4 for both LUC2AM and GFP2AM
only marginally increased their respective reporter activity (less
than 2-fold), we used MOI = 0.2 TCID
50
/cell for both reporter
viruses for our antiviral assays. We performed growth curves for
both viruses in 293T cells by infecting at MOI = 0.2 TCID
50
/cell,
and observed increases in reporter signals corresponding with in-
creases in virus titers (Fig. 2C).
3.3. Testing antiviral inhibitors against LUC2AM and GFP2AM
Having optimized both fluorescent and luminescent reporter
assays in 293T cells, we assayed LUC2AM and GFP2AM against
two known inhibitors of NiV, 6-azauridine acetate and pyrazofurin.
Both of these compounds inhibit cellular pyrimidine biosynthesis
via blocking orotidine 5
0
-monophosphate decarboxylase (Bono
et al., 1964; Georges-Courbot et al., 2006; Gutowski et al., 1975).
Consistent with a prior study utilizing a CPE-based assay
(Georges-Courbot et al., 2006), 6-azauridine acetate and pyrazofu-
rin showed dose-dependent inhibition of LUC2AM and GFP2AM,
reducing reporter activity to background levels at the highest doses
(25–50
l
M) while having mild to minimal effects on cell viability
(Fig. 3A). The respective 50% effective concentrations (EC
50
) deter-
mined for each compound using the LUC2AM and GFP2AM were
within 3-fold of each other (Table 1).
Since several studies have demonstrated the sensitivity of
henipaviruses to type-1 interferon (IFN) signaling (Dhondt et al.,
2013; Escaffre et al., 2013; Virtue et al., 2011), we evaluated the
antiviral effect of universal interferon (U-IFN) against our reporter
viruses. Although increasing concentrations of U-IFN led to corre-
sponding decreases in respective reporter activity, even the highest
concentration (1000 IU) of U-IFN only reduced LUC and GFP activity
by approximately 4-fold and 3-fold respectively (Fig. 3B, top pan-
els). These results are consistent with the early expression of multi-
ple antagonists of IFN signaling by NiV (Kulkarni et al., 2009;
Rodriguez et al., 2002; Shaw et al., 2005). EC
50
values for U-IFN
against LUC2AM and GFP2AM were nearly identical (Table 1). In
addition to U-IFN we also tested compound 09167, a recently char-
acterized agonist of the innate antiviral response (Yan et al., 2013)
(Fig. 3B, lower panels). In contrast to U-IFN, treatment with com-
pound 09167 was able to completely ablate both LUC and GFP activ-
ity at the highest dose tested (12.5
l
M), indicating that IFN
signaling-independent components have crucial roles in blocking
NiV replication. Dose response curves for compound 09167 against
both LUC2AM and GFP2AM confirmed sub-micro molar EC
50
s,
which were consistent with EC
50
s previously obtained for 09167
against other paramyxoviruses (Yan et al., 2013)(Table 1). Further
studies of this compound’s effect on cell viability are warranted to
determine its suitability for antiviral testing in vivo.
3.4. Counter screening against wild-type NiV, evaluating reporter
assays for high-throughput screening
To confirm whether the dose–response curves and EC
50
values
obtained using these reporter viruses reflected actual inhibition
of NiV replication, we tested 6-azauridine acetate and compound
09167 against wild-type non-recombinant NiV via virus yield-
based dose–response assays (Fig. 4A). We observed dose-
dependent reductions in virus titers to below initial levels of input
virus at the two highest doses for each compound (>4 Log
10
TCID
50
/
mL reduction), suggesting complete ablation of viral replication at
these concentrations. EC
50
values calculated for 6-azauridine and
09167 against wild-type NiV were within 1 Log
10
of EC
50
values de-
rived for each compound against LUC2AM and GFP2AM, indicating
that inhibition of the reporter viruses paralleled inhibition of NiV
replication (Fig. 4B, Table 1). The moderately higher EC
50
s calcu-
lated from the reporter viruses suggest a lesser likelihood to detect
false positives in the context of high-throughput screening (HTS).
To quantitatively assess the suitability of these two viruses for
Table 1
50% effective concentrations (EC
50
) of compounds against reporter and wild-type NiVs (therapeutic indices in parentheses).
Inhibitor ID Units NiV-LUC2AM NiV-GFP2AM NiV-MY-99
6-Azauridine acetate
l
M 0.274 ± 0.0357 (P364) 0.553 ± 0.151 (P180) 0.120 ± 0.0445 (P833)
Pyrazofurin
l
M 0.432 ± 0.0871 (P115) 0.281 ± 0.0448 (P177) ND
09167
l
M 0.0723 ± 0.0123 (877) 0.254 ± 0.0905 (249) 0.0289 ± 0.0102 (2196)
U-IFN IU 10.96 ± 4.90 (P91) 10.2 ± 8.49 (P98) ND
ND - Not done.
58 M.K. Lo et al. / Antiviral Research 106 (2014) 53–60
Author's personal copy
HTS, we used compound 09167 as a positive control for inhibition
and calculated Z
0
factor values, signal-to-noise ratios, and coeffi-
cients of variation for the 96-well plate format used throughout
this study. For both assays, these values were robust and well
within the acceptable range for HTS (Zhang et al., 1999)(Table 2).
4. Conclusions
The primary advantages to using viral reporter assays over viral
antigen-based assays are the significant reduction in assay turn-
around time and the minimal reagents required. Whereas a previ-
ously described henipavirus immunodetection assay required at
least 2 h of post-infection processing (fixing, blocking, antibody
staining) before the plate reading step, the LUC2AM assay required
just 10 min of reagent incubation before plate reading, while the
GFP2AM assay required even less time since no additional reagents
were required. The LUC2AM assay has several advantages over the
GFP2AM assay including better signal-to-noise ratios and less var-
iability between samples/wells. Although we established the LU-
C2AM assay to be read alongside the GFP2AM assay at 48 hpi,
the robust signal-to-noise ratio and Z
0
values for LUC2AM at even
24 hpi would allow for adaptation of the assay to span a shorter
infection period, whereas this would not be possible for GFP2AM
due to its longer maturation time (data not shown). The GFP2AM
assay has some advantages over LUC2AM assay in that GFP activity
can be measured multiple times, whereas LUC measurement is an
endpoint measurement. Furthermore, GFP2AM can be easily
adapted for high content imaging to simultaneously assess cell via-
bility and reporter activity, as well as for flow-cytometry-based
applications. In sum, we established reporter NiVs that will not
only facilitate antiviral screening, but also the study of cellular
components that influence viral replication.
Acknowledgements
We thank Mike Flint for helpful discussions and suggestions.
The findings and conclusions in this report are those of the authors
and do not necessarily represent those of the Centers for Disease
Control and Prevention.
References
Aljofan, M., Porotto, M., Moscona, A., Mungall, B.A., 2008. Development and
validation of a chemiluminescent immunodetection assay amenable to high
throughput screening of antiviral drugs for Nipah and Hendra virus. J. Virol.
Methods 149, 12–19.
Bono Jr., V.H., Weissman, S.M., Frei 3rd, E., 1964. The effect of 6-azauridine
administration on De Novo pyrimidine production in chronic myelogenous
leukemia. J. Clin. Investig. 43, 1486–1494.
Fig. 4. Confirmatory counter-screening against wild-type NiV. (A) Virus titer-reduction assays. 293T cells were infected with wild-type NiV (MOI = 0.2 TCID
50
/cell) for 2 h,
and then incubated with 3-fold dilutions of 6-azauridine acetate (left panel) or compound 09167 (right panel) for 48 h. Supernatant virus titers were determined by TCID
50
titrations and plotted against compound concentration. Values represent means and standard deviations of 3 experiments. Virus titers were then (B) normalized to vehicle
(DMSO) controls. EC
50
s were calculated based on four-parameter variable-slope nonlinear regression models. Normalized data points represent means for three experiments.
Table 2
Comparison of Nipah LUC2AM and GFP2AM assays used in this study.
Plate format Target virus
a
Z
0
value
b
S/N ratio
c
%CV
96 wells NiV-LUC2AM 0.85 1.33E + 04 5%
96 wells NiV-GFP2AM 0.67 2.16E + 03 11%
a
293T cells were infected with either LUC2AM or GFP2AM viruses for 2 h before
treatment with host antiviral agonist compound 09167 (final concentration,
12.5 mM) or an equivalent amount of vehicle (DMSO). Relative luciferase/fluores-
cence units were determined at 48 hpi.
b
Z
0
factor (Zhang et al., 1999). Statistical analyses were based on means of four
independent experiments.
c
Ratio of signal to background noise.
M.K. Lo et al. / Antiviral Research 106 (2014) 53–60 59
Author's personal copy
Bossart, K.N., Mungall, B.A., Crameri, G., Wang, L.F., Eaton, B.T., Broder, C.C., 2005.
Inhibition of Henipavirus fusion and infection by heptad-derived peptides of
the Nipah virus fusion glycoprotein. Virol. J. 2, 57.
Broder, C.C., Xu, K., Nikolov, D.B., Zhu, Z., Dimitrov, D.S., Middleton, D., Pallister, J.,
Geisbert, T.W., Bossart, K.N., Wang, L.F., 2013. A treatment for and vaccine
against the deadly Hendra and Nipah viruses. Antiviral Res. 100, 8–13.
Buchholz, U.J., Finke, S., Conzelmann, K.K., 1999. Generation of bovine respiratory
syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus
replication in tissue culture, and the human RSV leader region acts as a
functional BRSV genome promoter. J. Virol. 73, 251–259.
Chiang, C.F., Lo, M.K., Rota, P.A., Spiropoulou, C.F., Rollin, P.E., 2010. Use of
monoclonal antibodies against Hendra and Nipah viruses in an antigen capture
ELISA. Virol. J. 7, 115.
Corish, P., Tyler-Smith, C., 1999. Attenuation of green fluorescent protein half-life in
mammalian cells. Protein Eng. 12, 1035–1040.
Dhondt, K.P., Mathieu, C., Chalons, M., Reynaud, J.M., Vallve, A., Raoul, H., Horvat, B.,
2013. Type I interferon signaling protects mice from lethal henipavirus
infection. J. Infect. Dis. 207, 142–151.
Escaffre, O., Borisevich, V., Carmical, J.R., Prusak, D., Prescott, J., Feldmann, H., Rockx,
B., 2013. Henipavirus pathogenesis in human respiratory epithelial cells. J. Virol.
87, 3284–3294.
Freiberg, A., Dolores, L.K., Enterlein, S., Flick, R., 2008. Establishment and
characterization of plasmid-driven minigenome rescue systems for Nipah
virus: RNA polymerase I- and T7-catalyzed generation of functional
paramyxoviral RNA. Virology 370, 33–44.
Georges-Courbot, M.C., Contamin, H., Faure, C., Loth, P., Baize, S., Leyssen, P., Neyts,
J., Deubel, V., 2006. Poly(I)-poly(C12U) but not ribavirin prevents death in a
hamster model of Nipah virus infection. Antimicrob. Agents Chemother. 50,
1768–1772.
Gutowski, G.E., Sweeney, M.J., DeLong, D.C., Hamill, R.L., Gerzon, K., Dyke, R.W.,
1975. Biochemistry and biological effects of the pyrazofurins (pyrazomycins):
initial clinical trial. Ann. N. Y. Acad. Sci. 255, 544–551.
Kim, J.H., Lee, S.R., Li, L.H., Park, H.J., Park, J.H., Lee, K.Y., Kim, M.K., Shin, B.A.,
Choi, S.Y., 2011. High cleavage efficiency of a 2A peptide derived from
porcine teschovirus-1 in human cell lines, zebrafish and mice. PLoS ONE 6,
e18556.
Kolakofsky, D., Pelet, T., Garcin, D., Hausmann, S., Curran, J., Roux, L., 1998.
Paramyxovirus RNA synthesis and the requirement for hexamer genome
length: the rule of six revisited. J. Virol. 72, 891–899.
Kulkarni, S., Volchkova, V., Basler, C.F., Palese, P., Volchkov, V.E., Shaw, M.L., 2009.
Nipah virus edits its P gene at high frequency to express the V and W proteins. J.
Virol. 83, 3982–3987.
Lo, M.K., Peeples, M.E., Bellini, W.J., Nichol, S.T., Rota, P.A., Spiropoulou, C.F., 2012.
Distinct and overlapping roles of Nipah virus P gene products in modulating the
human endothelial cell antiviral response. PLoS ONE 7, e47790.
Loening, A.M., Fenn, T.D., Wu, A.M., Gambhir, S.S., 2006. Consensus guided
mutagenesis of Renilla luciferase yields enhanced stability and light output.
Protein Eng. Des. Sel. 19, 391–400.
Luby, S.P., 2013. The pandemic potential of Nipah virus. Antiviral Res. 100, 38–43.
Marsh, G.A., Virtue, E.R., Smith, I., Todd, S., Arkinstall, R., Frazer, L., Monaghan, P.,
Smith, G.A., Broder, C.C., Middleton, D., Wang, L.F., 2013. Recombinant Hendra
viruses expressing a reporter gene retain pathogenicity in ferrets. Virol. J. 10, 95.
Nahar, N., Mondal, U.K., Sultana, R., Hossain, M.J., Khan, M.S., Gurley, E.S., Oliveras,
E., Luby, S.P., 2013. Piloting the use of indigenous methods to prevent Nipah
virus infection by interrupting bats’ access to date palm sap in Bangladesh.
Health Promot. Int. 28, 378–386.
Porotto, M., Orefice, G., Yokoyama, C.C., Mungall, B.A., Realubit, R., Sganga, M.L.,
Aljofan, M., Whitt, M., Glickman, F., Moscona, A., 2009. Simulating henipavirus
multicycle replication in a screening assay leads to identification of a promising
candidate for therapy. J. Virol. 83, 5148–5155.
Rodriguez, J.J., Parisien, J.P., Horvath, C.M., 2002. Nipah virus V protein evades alpha
and gamma interferons by preventing STAT1 and STAT2 activation and nuclear
accumulation. J. Virol. 76, 11476–11483.
Rota, P.A., Lo, M.K., 2012. Molecular virology of the henipaviruses. Curr. Top.
Microbiol. Immunol. 359, 41–58.
Shaw, M.L., Cardenas, W.B., Zamarin, D., Palese, P., Basler, C.F., 2005. Nuclear
localization of the Nipah virus W protein allows for inhibition of both virus- and
toll-like receptor 3-triggered signaling pathways. J. Virol. 79, 6078–6088.
Talekar, A., Pessi, A., Glickman, F., Sengupta, U., Briese, T., Whitt, M.A., Mathieu, C.,
Horvat, B., Moscona, A., Porotto, M., 2012. Rapid screening for entry inhibitors of
highly pathogenic viruses under low-level biocontainment. PLoS ONE 7,
e30538.
Tamin, A., Harcourt, B.H., Lo, M.K., Roth, J.A., Wolf, M.C., Lee, B., Weingartl, H.,
Audonnet, J.C., Bellini, W.J., Rota, P.A., 2009. Development of a neutralization
assay for Nipah virus using pseudotype particles. J. Virol. Methods 160, 1–6.
Virtue, E.R., Marsh, G.A., Wang, L.F., 2011. Interferon signaling remains functional
during henipavirus infection of human cell lines. J. Virol. 85, 4031–4034.
Wang, Y.E., Park, A., Lake, M., Pentecost, M., Torres, B., Yun, T.E., Wolf, M.C.,
Holbrook, M.R., Freiberg, A.N., Lee, B., 2010. Ubiquitin-regulated nuclear-
cytoplasmic trafficking of the Nipah virus matrix protein is important for
viral budding. PLoS Pathog. 6, e1001186.
Yan, D., Krumm, S.A., Sun, A., Steinhauer, D.A., Luo, M., Moore, M.L., Plemper, R.K.,
2013. Dual myxovirus screen identifies a small-molecule agonist of the host
antiviral response. J. Virol. 87, 11076–11087.
Yoneda, M., Guillaume, V., Ikeda, F., Sakuma, Y., Sato, H., Wild, T.F., Kai, C., 2006.
Establishment of a Nipah virus rescue system. Proc. Nat. Acad. Sci. U.S.A. 103,
16508–16513.
Zhang, J.H., Chung, T.D., Oldenburg, K.R., 1999. A simple statistical parameter for use
in evaluation and validation of high throughput screening assays. J. Biomol.
Screen. 4, 67–73.
60 M.K. Lo et al. / Antiviral Research 106 (2014) 53–60
... Plasma was obtained from terminal intracardiac blood collected in EDTA from naïve hamsters (male and female; 5-10 week old HsdHan:AURA Syrian hamsters; Envigo no. 8903F or 8903 M) or hamsters inoculated intranasally with 10 3 to 10 7 TCID 50 (100 μL total, divided bilaterally between the nares) of NiV strain Malaysia, recombinant NiV Malaysia, or recombinant reporterexpressing NiV Malaysia (rNIV-M-ZsG), as previously reported (Lo et al., 2014b;Welch et al., 2020). Antibodies against NiV-F (12B2) and henipavirus-G (7B7 and 5B1) were obtained from Absolute Antibody. ...
... In order to find whether the neutralization activity observed on the rVSVΔG-NiV-G assay was comparable with that using fully infectious virus, we tested representative samples using recombinant NiV-ZsG (Lo et al., 2014b) in our BSL4 lab. As expected, representative samples that exhibited neutralization activity against rVSVΔG-NiV-G also neutralized fully infectious NiV (Supplementary figure). ...
Article
Full-text available
Nipah virus (NiV) is a highly pathogenic paramyxovirus with a high case fatality rate. Due to its high pathogenicity, pandemic potential, and lack of therapeutics or approved vaccines, its study requires biosafety level 4 (BSL4) containment. In this report, we developed a novel neutralization assay for use in biosafety level 2 laboratories. The assay uses a recombinant vesicular stomatitis virus expressing NiV glycoprotein and a fluorescent protein. The recombinant virus propagates as a replication-competent virus in a cell line constitutively expressing NiV fusion protein, but it is restricted to a single round of replication in wild-type cells. We used this system to evaluate the neutralization activity of monoclonal and polyclonal antibodies, plasma from NiV-infected hamsters, and serum from human patients. Therefore, this recombinant virus could be used as a surrogate for using pathogenic NiV and may constitute a powerful tool to develop therapeutics in low containment laboratories.
... The use of mCherry as a readout of TBEV replication can be performed with a fluorescence microscope or microplate fluorescence reader. This is the primary advantage of the mCherry-TBEV system over classical CPE, plaque-or antigen-based assays, or luciferase-based reporter viruses, leading to significant reduction in assay turn-around time and reduction of the use of reagents (Lo et al., 2014;Li et al., 2016). In summary, mCherry-TBEV provides a powerful quantitative tool to facilitate the identification of potential antiviral agents and to measure levels of neutralizing antibodies in human and animal sera. ...
Article
The flavivirus, tick-borne encephalitis virus (TBEV) is transmitted by Ixodes spp. ticks and may cause severe and potentially lethal neurological tick-borne encephalitis (TBE) in humans. Studying TBEV requires the use of secondary methodologies to detect the virus in infected cells. To overcome this problem, we rationally designed and constructed a recombinant reporter TBEV that stably expressed the mCherry reporter protein. The resulting TBEV reporter virus (named mCherry-TBEV) and wild-type parental TBEV exhibited similar growth kinetics in cultured cells; however, the mCherry-TBEV virus produced smaller plaques. The magnitude of mCherry expression correlated well with progeny virus production but remained stable over < 4 passages in cell culture. Using well-characterized antiviral compounds known to inhibit TBEV, 2’-C-methyladenosine and 2′-deoxy-2′-β-hydroxy-4′-azidocytidine (RO-9187), we demonstrated that mCherry-TBEV is suitable for high-throughput screening of antiviral drugs. Serum samples from a TBEV-vaccinated human and a TBEV-infected dog were used to evaluate the mCherry-based neutralization test (mCherry-NT). Collectively, recombinant mCherry-TBEV reporter virus described here provides a powerful tool to facilitate the identification of potential antiviral agents, and to measure levels of neutralizing antibodies in human and animal sera.
Article
Full-text available
We present a novel small molecule antiviral chemotype that was identified by an unconventional cell-free protein synthesis and assembly-based phenotypic screen for modulation of viral capsid assembly. Activity of PAV-431, a representative compound from the series, has been validated against infectious viruses in multiple cell culture models for all six families of viruses causing most respiratory diseases in humans. In animals, this chemotype has been demonstrated efficacious for porcine epidemic diarrhoea virus (a coronavirus) and respiratory syncytial virus (a paramyxovirus). PAV-431 is shown to bind to the protein 14-3-3, a known allosteric modulator. However, it only appears to target the small subset of 14-3-3 which is present in a dynamic multi-protein complex whose components include proteins implicated in viral life cycles and in innate immunity. The composition of this target multi-protein complex appears to be modified upon viral infection and largely restored by PAV-431 treatment. An advanced analog, PAV-104, is shown to be selective for the virally modified target, thereby avoiding host toxicity. Our findings suggest a new paradigm for understanding, and drugging, the host–virus interface, which leads to a new clinical therapeutic strategy for treatment of respiratory viral disease.
Article
Broad spectrum oral antivirals are urgently needed for the early treatment of many RNA viruses of clinical concern. We previously described the synthesis of 1-O-octadecyl-2-O-benzyl-glycero-3-phospho-RVn (V2043), an orally bioavailable lipid prodrug of remdesivir nucleoside (RVn, GS-441524) with broad spectrum antiviral activity against viruses with pandemic potential. Here we compared the relative activity of V2043 with new RVn lipid prodrugs containing sn-1 alkyl ether or sn-2 glycerol modifications. We found that 3-F-4-MeO-Bn, 3-CN-Bn, and 4-CN-Bn sn-2 glycerol modifications improved antiviral activity compared to V2043 when tested in vitro against clinically important RNA viruses from 5 virus families. These results support the continued development of V2043 and sn-2 glycerol modified RVn lipid prodrugs for the treatment of a broad range of RNA viruses for which there are limited therapies.
Article
In vivo imaging system (IVIS) is a powerful tool for the study of infectious diseases, providing the ability to non-invasively follow viral infection in an individual animal over time. Recombinant henipaviruses expressing bioluminescent or fluorescent reporter proteins can be used both to monitor the spatial and temporal progression of Nipah virus (NiV) infection in vivo as well as in ex vivo tissues. Virally produced luciferases react with systemically administered substrate to produce bioluminescence that can then be detected via IVIS imaging, while fluorescent reporters inherently generate detectable fluorescence without a substrate. Here we describe protocols applying bioluminescent or fluorescent reporter expressing recombinant viruses to in vivo or ex vivo imaging of NiV infection.
Article
Spillovers of Nipah virus (NiV) from its pteropid bat reservoir into the human population continue to cause near-annual outbreaks of fatal encephalitis and respiratory disease in Bangladesh and India since its emergence in Malaysia over 20 years ago. The current lack of effective antiviral therapeutics against NiV merits further testing of compound libraries against NiV using rapid quantitative antiviral assays. The development of recombinant henipaviruses expressing reporter fluorescence and/or luminescence proteins has facilitated the screening of such libraries. In this chapter, we provide a basic protocol for both types of reporter viruses. Utilizing these live NiV-based reporter assays requires modest instrumentation and sidesteps the labor-intensive steps associated with traditional cytopathic effect or viral antigen-based assays.
Article
Full-text available
Background The COVID-19 pandemic resulted in a worldwide shortage of N95 respirators, prompting the development of decontamination methods to enable limited reuse. Countries lacking reliable supply chains would also benefit from the ability to safely reuse PPE. Methylene blue (MB) is a light-activated dye with demonstrated antimicrobial activity used to sterilize blood plasma. Decontamination of respirators using photoactivated MB requires no specialized equipment, making it attractive for use in the field during outbreaks. Methods We examined decontamination of N95 and KN95 respirators using photoactivated MB and 3 variants of SARS-CoV-2, the virus that causes COVID-19; and 4 World Health Organization priority pathogens: Ebola virus, Middle East respiratory syndrome coronavirus, Nipah virus, and Lassa virus. Virus inactivation by pretreating respirator material was also tested. Results Photoactivated MB inactivated all tested viruses on respirator material, albeit with varying efficiency. Virus applied to respirator material pre-treated with MB was also inactivated, thus MB pretreatment may potentially protect respirator wearers from virus exposure in real-time. Conclusions These results demonstrate that photoactivated MB represents a cost-effective, rapid, and widely deployable method to decontaminate N95 respirators for reuse during supply shortages.
Chapter
New antiviral drugs are urgently needed. Recent outbreaks caused by viruses with great epidemiological impact such as Zika, or extraordinary virulence such as Ebola, Nipah, Lassa, Crimean-Congo haemorrhagic fever highlight the current lack of clinically proven vaccines and treatments for these potentially catastrophic agents. Antiviral Discovery for Highly Pathogenic Emerging Viruses comprehensively outlines the state of the art in antiviral drug discovery including identification of targets, screening strategies and the current pipeline of antiviral candidates including regulatory issues. The book also addresses the challenges faced in proceeding from pre-clinical studies to animal models and clinical trials with these highly pathogenic agents. Ideal for drug discovery scientists and medicinal chemists with an interest in antiviral drug discovery and development, this book provides a complete overview of the latest progress in the field, recent advances and the challenges that remain in developing these highly pathogenic agents. Illustrated throughout with case studies this book is a valuable resource in this complex and multidisciplinary field.
Preprint
Full-text available
The intravenous administration of remdesivir for COVID-19 confines its utility to hospitalized patients. We evaluated the broad-spectrum antiviral activity of ODBG-P-RVn, an orally available, lipid-modified monophosphate prodrug of the remdesivir parent nucleoside (GS-441524) against viruses that cause diseases of human public health concern, including SARS-CoV-2. ODBG-P-RVn showed 20-fold greater antiviral activity than GS-441524 and had near-equivalent activity to remdesivir in primary-like human small airway epithelial cells. Our results warrant investigation of ODBG-P-RVn efficacy in vivo.
Article
Full-text available
Confronted with an increasing number of emerging and re-emerging viral pathogens, the identification of novel pathogen-specific and broad-spectrum antivirals has become a major developmental objective. Targeting host factors required for virus replication presents a tangible approach towards novel hits with broadened indication range. However, the identification of developable host-directed antiviral candidates remains challenging. We describe a novel screening protocol that interrogates the myxovirus host-pathogen interactome for broad-spectrum drug candidates and simultaneously probes for conventional, pathogen-directed hits. With resource-efficiency and pan-myxovirus activity as the central developmental parameters, we explored co-screening against two distinct, independently traceable myxoviruses in a single-well setting. Having identified a pair of unrelated pathogenic myxoviruses (influenza A virus and measles virus) with comparable replication kinetics, we observed unimpaired co-replication of both viruses, generated suitable firefly and renilla luciferase reporter constructs, respectively, and validated the protocol up to 384-well plate format. Combined with an independent counterscreen using a recombinant respiratory syncytial virus luciferase reporter, implementation of the protocol identified candidates with broadened anti-myxovirus profile in addition to pathogen-specific hits. Mechanistic characterization revealed a newly discovered broad-spectrum lead that does not block viral entry, but stimulates effector pathways of the innate cellular antiviral response. In summary, we provide proof-of-concept for the efficient discovery of broad-spectrum myxovirus inhibitors in parallel to para- and orthomyxovirus-specific hit candidates in a single screening campaign. The newly identified compound provides a basis for the development of a novel broad-spectrum small-molecule antiviral class.
Article
Full-text available
Hendra virus and Nipah virus are bat-borne paramyxoviruses that are the prototypic members of the genus Henipavirus. The henipaviruses emerged in the 1990s, spilling over from their natural bat hosts and causing serious disease outbreaks in humans and livestock. Hendra virus emerged in Australia and since 1994 there have been 7 human infections with 4 case fatalities. Nipah virus first appeared in Malaysia and subsequent outbreaks have occurred in Bangladesh and India. In total, there have been an estimated 582 human cases of Nipah virus and of these, 54% were fatal. Their broad species tropism and ability to cause fatal respiratory and/or neurologic disease in humans and animals make them important transboundary biological threats. Recent experimental findings in animals have demonstrated that a human monoclonal antibody targeting the viral G glycoprotein is an effective post-exposure treatment against Hendra and Nipah virus infection. In addition, a subunit vaccine based on the G glycoprotein of Hendra virus affords protection against Hendra and Nipah virus challenge. The vaccine has been developed for use in horses in Australia and is the first vaccine against a Biosafety Level-4 (BSL-4) agent to be licensed and commercially deployed. Together, these advances offer viable approaches to address Hendra and Nipah virus infection of livestock and people.
Article
Full-text available
Background Hendra virus (HeV) is an Australian bat-borne zoonotic paramyxovirus that repeatedly spills-over to horses causing fatal disease. Human cases have all been associated with close contact with infected horses. Methods A full-length antigenome clone of HeV was assembled, a reporter gene (GFP or luciferase) inserted between the P and M genes and transfected to 293T cells to generate infectious reporter gene-encoding recombinant viruses. These viruses were then assessed in vitro for expression of the reporter genes. The GFP expressing recombinant HeV was used to challenge ferrets to assess the virulence and tissue distribution by monitoring GFP expression in infected cells. Results Three recombinant HeV constructs were successfully cloned and rescued; a wild-type virus, a GFP-expressing virus and a firefly luciferase-expressing virus. In vitro characterisation demonstrated expression of the reporter genes, with levels proportional to the initial inoculum levels. Challenge of ferrets with the GFP virus demonstrated maintenance of the fatal phenotype with disease progressing to death consistent with that observed previously with the parental wild-type isolate of HeV. GFP expression could be observed in infected tissues collected from animals at euthanasia. Conclusions Here, we report on the first successful rescue of recombinant HeV, including wild-type virus and viruses expressing two different reporter genes encoded as an additional gene cassette inserted between the P and M genes. We further demonstrate that the GFP virus retained the ability to cause fatal disease in a well-characterized ferret model of henipavirus infection despite the genome being an extra 1290 nucleotides in length.
Article
Full-text available
Hendra virus (HeV) and Nipah virus (NiV) are deadly zoonotic viruses for which no vaccines or therapeutics are licensed for human use. Henipavirus infection causes severe respiratory illness and encephalitis. Although the exact route of transmission in human is unknown, epidemiological studies and in vivo studies suggest the respiratory tract is important for virus replication. However, the target cells in the respiratory tract are unknown as well as the mechanisms by which henipaviruses can cause disease.Here we characterize henipavirus pathogenesis using primary cells derived from the human respiratory tract. The growth kinetics of NiV-Malaysia, NiV-Bangladesh and HeV was determined in Bronchial/Tracheal Epithelial Cells (NHBE) and Small Airway Epithelial Cells (SAEC). In addition, host responses to infection were assessed by gene expression analysis and immunoassays.Viruses replicated efficiently in both cell types and induced large syncytia. The host response to henipavirus infection in NHBE and SAEC highlighted a difference in the inflammatory response between HeV and NiV strains as well as intrinsic differences in the ability to mount an inflammatory response between NHBE and SAEC. These responses were highest during HeV infection in SAEC as characterized by the levels of key cytokines (IL-6, IL-8, IL-1α, MCP-1, CSFs) responsible for immune cell recruitment. Finally, we identified virus strain-dependant variability in type I interferon antagonism in NHBE/SAEC where NiV-Malaysia counteracted this pathway more efficiently than NiV-Bangladesh or HeV. These results provide crucial new information in the understanding of henipavirus pathogenesis in the human respiratory tract at an early stage of infection.
Article
Full-text available
Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that causes fatal encephalitis in up to 75% of infected humans. Like other paramyxoviruses, NiV employs co-transcriptional mRNA editing during transcription of the phosphoprotein (P) gene to generate additional mRNAs encoding the V and W proteins. The C protein is translated from the P mRNA, but in an alternative reading frame. There is evidence from both in vitro and in vivo studies to show that the P gene products play a role in NiV pathogenesis. We have developed a reverse genetic system to dissect the individual roles of the NiV P gene products in limiting the antiviral response in primary human microvascular lung endothelial cells, which represent important targets in human NiV infection. By characterizing growth curves and early antiviral responses against a number of recombinant NiVs with genetic modifications altering expression of the proteins encoded by the P gene, we observed that multiple elements encoded by the P gene have both distinct and overlapping roles in modulating virus replication as well as in limiting expression of antiviral mediators such as IFN-β, CXCL10, and CCL5. Our findings corroborate observations from in vivo hamster infection studies, and provide molecular insights into the attenuation and the histopathology observed in hamsters infected with C, V, and W-deficient NiVs. The results of this study also provide an opportunity to verify the results of earlier artificial plasmid expression studies in the context of authentic viral infection.
Article
Full-text available
Hendra virus (HeV) and Nipah virus (NiV) are closely related, recently emerged paramyxoviruses that form Henipavirus genus and are capable of causing considerable morbidity and mortality in a number of mammalian species, including humans. However, in contrast to many other species and despite expression of functional virus entry receptors, mice are resistant to henipavirus infection. We report here the susceptibility of mice deleted for the type I interferon receptor (IFNAR-KO) to both HeV and NiV. Intraperitoneally infected mice developed fatal encephalitis, with pathology and immunohistochemical features similar to what was found in humans. Viral RNA was found in the majority of analyzed organs, and sublethally infected animals developed virus-specific neutralizing antibodies. Altogether, these results reveal IFNAR-KO mice as a new small animal model to study HeV and NiV pathogenesis, prophylaxis, and treatment and suggest the critical role of type I interferon signaling in the control of henipavirus infection.
Article
Full-text available
People in Bangladesh frequently drink fresh date palm sap. Fruit bats (Pteropus giganteus) also drink raw sap and may contaminate the sap by shedding Nipah virus through saliva and urine. In a previous study we identified two indigenous methods to prevent bats accessing the sap, bamboo skirts and lime (calcium carbonate). We conducted a pilot study to assess the acceptability of these two methods among sap harvesters. We used interactive community meetings and group discussions to encourage all the sap harvesters (n = 12) from a village to use either bamboo skirts or lime smear that some of them (n = 4) prepared and applied. We measured the preparation and application time and calculated the cost of bamboo skirts. We conducted interviews after the use of each method. The sap harvesters found skirts effective in preventing bats from accessing sap. They were sceptical that lime would be effective as the lime was washed away by the sap flow. Preparation of the skirt took ∼105 min. The application of each method took ∼1 min. The cost of the bamboo skirt is minimal because bamboo is widely available and they made the skirts with pieces of used bamboo. The bamboo skirt method appeared practical and affordable to the sap harvesters. Further studies should explore its ability to prevent bats from accessing date palm sap and assess if its use produces more or better quality sap, which would provide further incentives to make it more acceptable for its regular use.
Article
Full-text available
Nipah (NiV) and Hendra (HeV) viruses comprise the genus Henipavirus and are highly pathogenic paramyxoviruses, which cause fatal encephalitis and respiratory disease in humans. Since their respective initial outbreaks in 1998 and 1994, they have continued to cause sporadic outbreaks resulting in fatal disease. Due to their designation as Biosafety Level 4 pathogens, the level of containment required to work with live henipaviruses is available only to select laboratories around the world. This chapter provides an overview of the molecular virology of NiV and HeV including comparisons to other, well-characterized paramyxoviruses. This chapter also describes the sequence diversity present among the henipaviruses.
Article
Full-text available
Emerging viruses including Nipah, Hendra, Lujo, and Junin viruses have enormous potential to spread rapidly. Nipah virus, after emerging as a zoonosis, has also evolved the capacity for human-to-human transmission. Most of the diseases caused by these pathogens are untreatable and require high biocontainment conditions. Universal methods for rapidly identifying and screening candidate antivirals are urgently needed. We have developed a modular antiviral platform strategy that relies on simple bioinformatic and genetic information about each pathogen. Central to this platform is the use of envelope glycoprotein cDNAs to establish multi-cycle replication systems under BSL2 conditions for viral pathogens that normally require BSL3 and BSL4 facilities. We generated monoclonal antibodies against Nipah G by cDNA immunization in rats, and we showed that these antibodies neutralize both Nipah and Hendra live viruses. We then used these effective Henipavirus inhibitors to validate our screening strategy. Our proposed strategy should contribute to the response capability for emerging infectious diseases, providing a way to initiate antiviral development immediately upon identifying novel viruses.
Article
Nipah virus, a paramyxovirus whose wildlife reservoir is Pteropus bats, was first discovered in a large outbreak of acute encephalitis in Malaysia in 1998 among persons who had contact with sick pigs. Apparently, one or more pigs was infected from bats, and the virus then spread efficiently from pig to pig, then from pigs to people. Nipah virus outbreaks have been recognized nearly every year in Bangladesh since 2001 and occasionally in neighboring India. Outbreaks in Bangladesh and India have been characterized by frequent person-to-person transmission and the death of over 70% of infected people. Characteristics of Nipah virus that increase its risk of becoming a global pandemic include: humans are already susceptible; many strains are capable of limited person-to-person transmission; as an RNA virus, it has an exceptionally high rate of mutation: and that if a human-adapted strain were to infect communities in South Asia, high population densities and global interconnectedness would rapidly spread the infection. Appropriate steps to estimate and manage this risk include studies to explore the molecular and genetic basis of respiratory transmission of henipaviruses, improved surveillance for human infections, support from high-income countries to reduce the risk of person-to-person transmission of infectious agents in low-income health care settings, and consideration of vaccination in communities at ongoing risk of exposure to the secretions and excretions of Pteropus bats.