ArticlePDF Available

Development of a reverse genetics system to generate a recombinant Ebola virus Makona expressing a green fluorescent protein

Authors:

Abstract and Figures

Previous studies have demonstrated the potential application of reverse genetics technology in studying a broad range of aspects of viral biology, including gene regulation, protein function, cell entry, and pathogenesis. Here, we describe a highly efficient reverse genetics system used to generate recombinant Ebola virus (EBOV) based on a recent isolate from a human patient infected during the 2014-2015 outbreak in Western Africa. We also rescued a recombinant EBOV expressing a fluorescent reporter protein from a cleaved VP40 protein fusion. Using this virus and an inexpensive method to quantitate the expression of the foreign gene, we demonstrate its potential usefulness as a tool for screening antiviral compounds and measuring neutralizing antibodies. Published by Elsevier Inc.
(A) VP40 fusion protein. Schematics of plasmids expressing VP40 or ZsG/P2A/VP40 fusion protein are shown. (B) ZsGreen (ZsG) expression. Huh7 cells were transfected with pC-VP40 or pC-ZsG/VP40. Images shown were taken 3 days post transfection on an inverted fluorescent microscope using the GFP channel. (C) VP40 expression. Huh7 cells were transfected as above and harvested 3 days post transfection. Protein lysates were analyzed by western blotting using a Mab against EBOV-VP40 (lower panel). (D) Schematic representation of rEBOV genome expressing ZsG. Genome of rEBOV expressing ZsG fusion protein (rEBOV/ZsG) is shown in the viral complementary sense. (E) Rescue of rEBOV/ZsG. rEBOV/ZsG was rescued using the same conditions as wild-type virus (see Fig. 1B). Fresh monolayers of Huh7 cells were infected with transfection supernatants. ZsG expression was visualized on an inverted fluorescent microscope using the GFP channel (left panel), or via IFA as in Fig. 1D (middle panel, and merged images panel at right). (F) Rescue efficiency. Efficiency of rescue and average titer of rEBOV/ZsG were assessed as in Fig. 1. (G) Growth kinetics of wild-type and recombinant viruses. Huh7 cells were infected with wild-type EBOV Mayinga (EBOV-Z1976), or wild-type human isolate of the Makona variant (EBOV-L2014), or with the 2 recombinant viruses, rEBOV or rEBOV/ZsG, with an MOI ¼0.1. Growth kinetics were assessed by determining viral titers in cell supernatants using a standard TCID 50 assay.
… 
Content may be subject to copyright.
Brief Communication
Development of a reverse genetics system to generate a recombinant
Ebola virus Makona expressing a green uorescent protein
$
César G. Albariño
n
, Lisa Wiggleton Guerrero, Michael K. Lo, Stuart T. Nichol,
Jonathan S. Towner
Centers for Disease Control and Prevention, Atlanta, GA, USA
article info
Article history:
Received 2 April 2015
Returned to author for revisions
21 April 2015
Accepted 10 June 2015
Available online 27 June 2015
abstract
Previous studies have demonstrated the potential application of reverse genetics technology in studying
a broad range of aspects of viral biology, including gene regulation, protein function, cell entry, and
pathogenesis. Here, we describe a highly efcient reverse genetics system used to generate recombinant
Ebola virus (EBOV) based on a recent isolate from a human patient infected during the 20142015
outbreak in Western Africa. We also rescued a recombinant EBOV expressing a uorescent reporter
protein from a cleaved VP40 protein fusion. Using this virus and an inexpensive method to quantitate the
expression of the foreign gene, we demonstrate its potential usefulness as a tool for screening antiviral
compounds and measuring neutralizing antibodies.
Published by Elsevier Inc.
Introduction
The rst outbreak of Ebola virus disease (EVD) caused by Ebola
virus (EBOV) was detected in 1976 in Zaire (now the Democratic
Republic of the Congo), and received signicant international
attention due to its high case fatality rate (Johnson, 1978;
Johnson et al., 1977). During the early months of 2014, a large
EVD outbreak was detected in the Republic of Guinea (Baize et al.,
2014; Gatherer, 2014) and spread quickly to the neighboring
countries of Liberia and Sierra Leone (Dixon et al., 2014). To date,
this continuing outbreak has been the largest EVD outbreak ever
recorded, with over 23,000 cases and over 10,000 deaths in
Western Africa as of March 2015. A limited number of cases were
also recorded in Mali, Nigeria, Senegal, USA, Spain, Germany, and
the United Kingdom (Baize, 2015; CDC, 2015).
Several viruses of the Filoviridae family, including EBOV, Sudan
virus, Bundibugyo virus, Tai Forest virus, Marburg virus (MARV),
and Ravn virus, cause sporadic outbreaks of viral hemorrhagic
fevers (VHFs) with high case fatality rates in sub-Saharan Africa
(Albariño et al., 2013a; Feldmann et al., 2013; Hartman et al., 2010;
Leroy et al., 2011). Filoviruses are enveloped viruses that carry a
single-strand RNA genome with negative-sense polarity (Feldmann
et al., 2013). The loviral genome is approximately 19 kb, and
encodes 7 genes, NP, VP35, VP40, VP30, VP24, and L, which are
transcribed in sequential order from the 3
0
end of the viral genome
and are separated by intergenic untranslated regions (Fig. 1A).
For more than a decade, reverse genetics technology has been a
useful tool for screening antiviral compounds and for studying
different aspects of lovirus biology, including virulence factors in
the viral genome, cell entry, mechanisms of transcription and
replication, and pathogenesis (Ebihara et al., 2005; Falzarano and
Feldmann, 2014; Hoenen and Feldmann, 2014; Hoenen et al., 2011;
Neumann et al., 2002; Theriault et al., 2005). Several reports also
describe modifying the genomes of EBOV and MARV by inserting
reporter genes, such as those coding for uorescent proteins and
luciferase (Albariño et al., 2013b; Ebihara et al., 2007; Hoenen
et al., 2013; Schmidt et al., 2011; Schudt et al., 2013; Towner et al.,
2005; Uebelhoer et al., 2014).
In order to start studies on the biological characteristics of the
EBOV variant responsible for the current Western Africa outbreak,
we developed a reverse genetics system for an isolate (Makona)
that was obtained from a patient in 2014. Here, we report the
generation of recombinant viruses that would constitute an
appropriate tool for future studies on this particular EBOV variant.
Results and discussion
Comparisons of full-length genomes from EBOV samples iso-
lated from 1976 to 2014 show a high degree of sequence identity
(Fig. 1A). As expected, representative isolates of the Makona
variant from the 2014 outbreak in Western Africa, obtained from
Contents lists available at ScienceDirect
journal homepage: www.elsevier.com/locate/yviro
Virology
http://dx.doi.org/10.1016/j.virol.2015.06.013
0042-6822/Published by Elsevier Inc.
The ndings and conclusions in this report are those of the author(s) and do not
necessarily represent the ofcial position of the Centers for Disease Control and
Prevention.
n
Corresponding author.
E-mail address: calbarino@cdc.gov (C.G. Albariño).
Virology 484 (2015) 259264
patients in Liberia, Guinea, and Sierra Leone, showed over 99%
sequence identity with each other across the full length of the
genome (Baize et al., 2014; Gire et al., 2014). In contrast, these
isolates showed 97% sequence identity with the Mayinga isolate
of the Yambuku variant collected during the 1976 outbreak, and
with representative isolates of the Kikwit variant from 1995. Until
now, all published studies using reverse genetics technology with
EBOV have used only the historic Mayinga isolate (Groseth et al.,
2012; Hartman et al., 2008, 2006; Martinez et al., 2011; Neumann
et al., 2002; Volchkov et al., 2001). Despite the limited divergence
between the Makona and Yambuku variant isolates (Fig.1A: 97%
identities, 561 different nucleotides), concerns have been raised
about differences in pathogenicity, virulence, or transmission
associated with the 2014 variant (Dowall et al., 2014; Gire et al.,
2014; Stadler et al., 2014). For this reason, we developed a new
reverse genetics system based on an isolate from the current EVD
outbreak so that detailed studies on this 2014 variant or compar-
ison studies with previous variants could be performed.
After obtaining conrmation of the sequence identity of the
viral RNA termini by standard 5
0
and 3
0
RACE methods (data not
shown), we constructed a full-length recombinant clone (Fig. 1B)
based on the Makona variant isolate (Ebola virus/H.sapiens-wt/
LBR/2014/Makona-201403007; an isolate of the Makona variant
(GenBank accession #KP178538). Similarly to previously reported
constructs for EBOV and MARV (Albariño et al., 2013b; Enterlein
et al., 2006; Neumann et al., 2002; Volchkov et al., 2001), the viral
complementary (anti-genomic) sense genome was cloned into a
standard T7 transcription vector, anked at the 3
0
end by the HDV
ribozyme and the T7 terminator.
Different cells lines, including 293T, BSRT7 and BHK, have been
traditionally used to rescue EBOV and MARV (Albariño et al.,
2013b; Neumann et al., 2002; Towner et al., 2005; Volchkov
et al., 2001). Since human liver cells are an important target of
EBOV infection, we decided to use Huh7 cells, a fully differentiated
human liver cell line, for viral rescue due to their high transfection
efciency and the high titers obtained by propagating wild-type
loviruses in these cells. In addition, Huh7 cells minimize selection
of variant viruses generated by reverse genetics (Tsuda et al.,
2015). To rescue the wild-type recombinant EBOV (rEBOV), we
co-transfected Huh7 cells with the full-length genomic plasmid,
with codon-optimized support plasmids expressing EBOV NP, L (or
inactive L; see below), VP35, and VP30, and with another plasmid
expressing a codon-optimized version of the T7 RNA polymerase
(Uebelhoer et al., 2014). For the negative control, we built a new
plasmid expressing an inactive form of the EBOV L polymerase
(L-inac), in which the GDN motif was removed from the L ORF.
Four days post transfection, we applied the claried supernatants
from this transfection onto fresh monolayers of Huh7. We assessed
viral rescue 5 days later by performing an immunouorescence
assay to detect EBOV antigens (Fig. 1C). As shown in Fig. 1D, the
rescue of rEBOV was successful in 11 of 11 replica wells, which
demonstrated the high efciency of our new approach to
rescue rEBOV.
After conrming the successful rescue of rEBOV, we developed
a new recombinant virus expressing a reporter gene that could be
used to detect viral infection in live cells or to facilitate screening
antiviral agents against the 2014 EBOV variant. Historically, the
most common approach for introducing a reporter gene into the
loviral genome has been to insert an additional transcription unit
into any of the 6 intergenic regions (Albariño et al., 2013b; Ebihara
et al., 2007; Hoenen et al., 2013; Schmidt et al., 2011; Schudt et al.,
2013; Towner et al., 2005; Uebelhoer et al., 2014). Unfortunately,
recombinant viruses generated using this approach exhibited
attenuated phenotypes in animal models (Ebihara et al., 2007)or
Fig. 1. (A) Sequence comparison. Full-length genomes of representative Ebola virus (EBOV) isolates compared at the nucleotide level. Sequence identity (%) is indicated in the
lower diagonal half, while the number of differing residues is indicated in the upper diagonal half. (B) Schematic representation of recombinant EBOV (rEBOV) genome.
Genome of recombinant EBOV in the viral complementary sense, with the 7 ORFs depicted in the 5
0
to 3
0
orientation. (C) Rescue of rEBOV. Wild-type rEBOV was rescued in
Huh7 cells using the full-length clone shown in B, T7 polymerase expression plasmid, and codon-optimized support plasmids that express NP, VP35, VP30, and L or inactive L
(L-inac). Supernatants from transfected cells were used to infect fresh monolayers of Huh7 cells. The infected cells were xed 4 days post infection, and stained for
immunouorescence assays (IFA) using a polyclonal rabbit anti-EBOV antibody followed by anti-rabbit Alexa-Fluor 594. (D) Efciency of rescue. Rescue efciency is shown as
the number of positive wells (pos)/total wells; average titer was assessed by standard TCID
50
assay.
C.G. Albariño et al. / Virology 484 (2015) 259264260
reduced growth in interferon-competent cells (Albariño et al.,
2013b). An alternative strategy, used for non-segmented negative
strand viruses, has been to fuse the foreign gene to one of the viral
genes (Chambers and Takimoto, 2010; Duprex et al., 2002; Hoenen
et al., 2012; Lo et al., 2014; Schudt et al., 2013; Silin et al., 2007). A
particularly interesting report by Lo and colleagues describes the
successful generation of a fully functional recombinant Nipah virus
in which the reporter gene, green uorescent protein (GFP), had
been fused to the matrix protein, M (Lo et al., 2014). In this
particular construct, M and GFP are released through the action of
self-cleaving peptides, such as F2A derived from foot and mouth
disease virus, or P2A derived from porcine teschovirus 2A (Kim
et al., 2011). We therefore decided to test this latest strategy by
fusing the modied green uorescent protein, ZsGreen1 (ZsG), to
the self-cleaving peptide P2A and EBOV VP40.
To verify that VP40 is released from the protein fusion, ZsG/
P2A/VP40 was initially cloned into a standard expression vector
(Fig. 2A) and used to transfect Huh7 cells. As shown in Fig. 2B, the
fusion protein yields bright green uorescence in transfected cells.
Moreover, protein analysis by western blotting showed that EBOV
VP40 is released from the fusion protein, although at lower
expression levels than wild-type VP40 (Fig. 2C).
Fig. 2. (A) VP40 fusion protein. Schematics of plasmids expressing VP40 or ZsG/P2A/VP40 fusion protein are shown. (B) ZsGreen (ZsG) expression. Huh7 cells were
transfected with pC-VP40 or pC-ZsG/VP40. Images shown were taken 3 days post transfection on an inverted uorescent microscope using the GFP channel. (C) VP40
expression. Huh7 cells were transfected as above and harvested 3 days post transfection. Protein lysates were analyzed by western blotting using a Mab against EBOV-VP40
(lower panel). (D) Schematic representation of rEBOV genome expressing ZsG. Genome of rEBOV expressing ZsG fusion protein (rEBOV/ZsG) is shown in the viral
complementary sense. (E) Rescue of rEBOV/ZsG. rEBOV/ZsG was rescued using the same conditions as wild-type virus (see Fig. 1B). Fresh monolayers of Huh7 cells were
infected with transfection supernatants. ZsG expression was visualized on an inverted uorescent microscope using the GFP channel (left panel), or via IFA as in Fig. 1D
(middle panel, and merged images panel at right). (F) Rescue efciency. Efciency of rescue and average titer of rEBOV/ZsG were assessed as in Fig. 1. (G) Growth kinetics of
wild-type and recombinant viruses. Huh7 cells were infected with wild-type EBOV Mayinga (EBOV-Z1976), or wild-type human isolate of the Makona variant (EBOV-L2014),
or with the 2 recombinant viruses, rEBOV or rEBOV/ZsG, with an MOI¼0.1. Growth kinetics were assessed by determining viral titers in cell supernatants using a standard
TCID
50
assay.
C.G. Albariño et al. / Virology 484 (2015) 259264 261
After conrming that ZsG and EBOV VP40 can be expressed
from the fusion cassette, we modied our new full-length clone to
carry this cassette in the same locus as wild-type VP40 (Fig. 2D).
Using the conditions described above to rescue wild-type rEBOV,
we successfully generated rEBOV/ZsG in all 11 replica wells (Fig. 2E
and F). Both rEBOV and rEBOV/ZsG were successfully rescued with
100% efciency (11 of 11 wells) in 2 successive rescue attempts.
As shown in Fig. 2E, rEBOV/ZsG recombinant virus expressed
ZsG and spread through the cell monolayer in a pattern similar to
that of wild-type rEBOV.
We compared the growth kinetics of the most relevant wild-
type viruses, EBOV Mayinga isolate and the human isolate of the
Makona variant, and of the 2 recombinant viruses, rEBOV and
rEBOV/ZsG. Interestingly, the wild-type Mayinga virus (EBOV-
Z1976) exhibited a slight growth advantage over the Makona
isolate (EBOV-L2014), while EBOV-L2014 and rEBOV exhibited
very similar growth characteristics (Fig. 2G). Moreover, rEBOV
grew slightly faster than rEBOV/ZsG (Fig.2G).
Quantifying the uorescence signal from GFP- or RFP-
expressing viruses has an intrinsic limitation because it requi-
res expensive, high-content, sophisticated imaging techniques
(Panchal et al., 2010, 2012). To avoid this need, we sought to
validate an alternative method described by Lo et al. (2014), which
quanties viral growth by measuring the uorescence signal from
live cells infected with rEBOV/ZsG. In this experiment, we infected
Huh7 cells with rEBOV/ZsG at a low multiplicity of infection (MOI),
and captured daily micrographs to record the progress of infection
(Fig. 3A). Concurrently, we also measured the ZsG-specic signal
on a multi-mode microplate reader and removed sample aliquots
to determine viral titers over 5 days of infection. As shown in
Fig. 3B, the uorescence signal of ZsG could be easily quantied in
a non-destructive way, and these data correlated with the spread
of virus shown in the captured images (Fig. 3A). Moreover, the
gradual increase of the ZsG signal also matched with increases in
viral titers (Fig. 3B).
Using this straightforward approach for quantifying the uor-
escence signal, we conducted a proof of principle experiment
using rEBOV/ZsG as a rapid tool for screening antiviral compounds
and measuring neutralizing antibodies in sera collected from
convalescent rhesus monkeys. As shown in Fig. 3C, we used
rEBOV/ZsG virus in Huh7 cells to measure the antiviral effect of
6azaU, a known inhibitor of viral replication (Crance et al., 2003;
Morrey et al., 2002; Pyrc et al., 2006; Smee et al., 1987; Uebelhoer
et al., 2014). Consistent with our previous report (Uebelhoer et al.,
2014), 6azaU used at the maximum concentration tolerated with-
out toxicity signicantly reduced ZsG signal: 94% and 97% signal
reduction at 62.5 μM and 125 μM concentrations of 6azaU,
respectively
As a second proof of principle experiment, we measured the
neutralizing effect of sera collected from convalescent rhesus
monkeys infected with EBOV Mayinga on rEBOV/ZsG (Fig. 3D). In
this experiment, we pre-incubated rEBOV/ZsG with different
dilutions of an IgGþor an IgGserum, and then used the treated
virus to infect Huh7 cells. As shown in Fig. 3D, the ZsG signal was
reduced by 98% after using a 1:400 dilution of the IgG þserum.
Conclusions
Here, we describe a highly efcient reverse genetics system to
generate a recombinant EBOV (rEBOV) based on a virus isolated
from a patient during the 20142015 Western Africa EVD out-
break. We also report the successful rescue of a recombinant EBOV
expressing a ZsG reporter protein that is initially fused to the EBOV
protein VP40, but is released by a cis-acting proteolytic cleavage
event. The fusion of this reporter gene did not adversely affect
Fig. 3. (A) Characterization of rEBOV/ZsG. Vero-E6 cells were infected with rEBOV/
ZsG viruses at MOI of 0.01, and pictures were taken as in Fig. 2C on indicated days
post infection (dpi). (B) Quantitation of ZsG expression and viral titers. Huh7 cells
were infected as indicated above, and uorescence resulting from ZsG expression
was measured daily for 5 days using a multi-mode microplate reader (BioTek
Synergy). Fluorescence is reported as relative uorescence units (RU). Viral titers
were determined as in Fig. 2G. (C) Effects of antiviral agent 6azaU. Huh7 cells
growing in 96-well plates were pre-treated with 6azaU at indicated concentrations
for 1 h, and then infected with rEBOV/ZsG at MOI of 0.1. Three days post infection,
ZsG expression (bars) was measured as indicated above. Mean and SEM of ZsG
expression from 4 wells are displayed (left Y-axis). Cytotoxicity was assayed by
measuring cellular ATP content (circles, right Y-axis) in uninfected cells treated
with 6azaU at indicated concentrations. (D) Serum neutralization assay. rEBOV/ZsG
was pre-incubated for 1 h with the indicated dilutions of an IgG-positive or control
rhesus monkey serum, and then used to infect Huh7 cells growing in 96-well
plates. Expression of ZsG was measured as indicated above; mean and SEM from
4 wells are depicted.
C.G. Albariño et al. / Virology 484 (2015) 259264262
viral replication in vitro. Moreover, we show a straightforward
approach to quantify virus growth of rEBOV/ZsG in live infected
cells, and demonstrate that this virus can be used to screen
antiviral drugs and to measure the neutralizing effect of antibodies
in sera from convalescent animals. Finally, we consider that the
tools described here could have other potential applications, such
as testing the effects of novel mutations that could arise in nature,
specically directed at the variant responsible for the largest
outbreak of EVD ever reported.
Materials and methods
Cell culture and biosafety
All work with recombinant viruses was performed in a biosaf-
ety level 4 (BSL-4) facility. Huh7 and Vero-E6 cells were propa-
gated in Dulbecco's modied Eagle's medium (DMEM, Life
Technologies, Grand Island, NY, USA) supplemented with 5% fetal
bovine serum (FBS) and penicillinstreptomycin (PenStrep).
Plasmid construction
(A) Support plasmids. The construction of EBOV support plas-
mids (pC-L, pC-NP, pC-V35, and pC-v30) has been described before
(Uebelhoer et al., 2014). Briey, these clones were designed to
express rodent codon-optimized synthetic genes corresponding to
those of the EBOV Mayinga isolate. The original pC-L plasmid was
also modied to express an inactive form of the EBOV L polymer-
ase (L-inac) by removing the GDN motif from the L ORF. An
expression cassette containing ZsG/P2A/VP40 was made using a
previously described strategy (Lo et al., 2014). Briey, ZsGreen1
(ZsG) ORF (Clontech, Mountain View, CA, USA) was fused to the
self-cleaving P2A peptide and to EBOV VP40, and cloned into the
standard Pol II expression vector pCAGGS (Niwa et al., 1991). (B)
Full-length clone. Viral RNA from Ebola virus/H.sapiens-wt/LBR/
2014/Makona-201403007 (GenBank accession KP178538) was
used as template to amplify by RT-PCR 2 overlapping fragments
of similar size spanning the full-length genome. These fragments
were gel-puried and used to assemble a full-length clone into the
T7 transcription vector. The nal plasmid contained the full-length
anti-genome (viral complementary sense) preceded by the T7 RNA
polymerase promoter and followed by the hepatitis delta virus
ribozyme and T7 polymerase terminator. A spurious nucleotide
change in the GP-VP30 intergenic region was kept to differentiate
the recombinant viruses rEBOV and rEBOV/ZsG from the wild-
type virus.
The full-length clone was later modied by replacing the VP40
ORF with the ZsG/P2A/VP40 cassette described above. Both full-
length clones were sequenced to completion. Details regarding
construction strategies of support expression plasmids and plas-
mids encoding the full-length clone are available upon request.
Rescue of infectious viruses
Rescue of recombinant viruses was performed in Huh7 cells as
described previously (Albariño et al., 2013b). Briey, a 70%
conuent monolayer of Huh7 cells grown in 12-well plates was
transfected with 1 μg pEBOV, 0.5 μg pC-L (wells #111) or pC-L-
inac (well #12), 0.5 μg pC-NP, 0.05 μg pC-VP35, 0.05 μg pC-VP30,
and 1 μg of codon-optimized pC-T7
Supernatants from transfected cells were harvested 4 days post
transfection, claried by low-speed centrifugation, and passaged
twice in fresh monolayers of Huh7 cells. The rescue events were
conrmed by immunostaining Huh7 cell monolayers (also grown
in 12-well plates) infected with the rst passage of the virus.
A 100% rescue efciency (positive detection in 11 of 11 replica
wells) was obtained in 2 successive rescue experiments.
Both recombinant viruses were sequenced to completion, and
complete genomic sequences were deposited in GenBank (acces-
sion KR781608 and KR781609 for rEBOV and rEBOV/ZsG, respec-
tively). The viral genomic sequences were identical to those in the
full-length plasmids.
Virus titration and growth curves
To characterize the growth kinetics of wild-type and recombi-
nant viruses, 210
6
Huh7 cells were infected at MOI¼0.1. After
1 h of adsorption, cell monolayers were washed with PBS. Aliquots
of the supernatant were taken daily, and viral titers were deter-
mined by tissue culture infective dose 50 (TCID
50
) assay, as
described previously (Uebelhoer et al., 2014).
Protein expression
Expression of ZsG in transfected or infected live cells was
determined by direct UV microscopy using the GFP channel.
Expression of cleaved VP40 was detected by western blotting
using a monoclonal anti-EBOV-VP40 antibody followed by anti-
mouse HRP secondary antibody. Staining of TCID
50
plates was
done using a polyclonal rabbit anti-EBOV antibody followed by
anti-rabbit Alexa-Fluor 594 antibody.
Quantitation of ZsG
The protocol described by Lo et al. (2014) was used to
quantitate ZsG uorescence in infected cells. Briey, 410
4
Huh7 cells were seeded in 96-well at-bottom black plates
(Corning) in 100 μL Fluorobrite medium (Life Technologies) per
well. The medium was removed on the following day, and cells
were pre-treated with 100 μL media containing various concen-
trations of 6azaU for 1 h. Virus was added in an additional 100 μL
media (nal MOI¼0.1). Three days post infection, ZsG uorescence
was measured in 4 replicates for each antiviral concentration using
a multi-mode microplate reader (BioTek Synergy) with a gain/
sensitivity preset of 90. Cell viability was determined by measur-
ing ATP content using CellTiter-Glo Luminescent Cell Viability
reagent (Promega) as previously described (Uebelhoer et al., 2014).
Serum neutralization
About 300 TCID
50
of rEBOV/ZsG were incubated for 1 h at 37 1C
with indicated dilutions of an IgGþor a control (IgG) serum
from rhesus monkeys, and then used to infect Huh7 cells growing
in 96-well at-bottom black plates with Fluorobrite medium.
Three days post infection, ZsG uorescence was measured in
4 replicates for each serum concentration, as described above.
Acknowledgments
We thank Marina L. Khristova for continuous excellent sequen-
cing support, and Tatyana Klimova for editing this manuscript. This
study was reviewed and approved by CDC's Institutional Biosafety
Committee.
References
Albariño, C.G., Shoemaker, T., Khristova, M.L., Wamala, J.F., Muyembe, J.J., Balinandi, S.,
Tumusiime, A., Campbell, S., Cannon, D., Gibbons, A., Bergeron, E., Bird, B., Dodd,
K., Spiropoulou, C., Erickson, B.R., Guerrero, L., Knust, B., Nichol, S.T., Rollin, P.E.,
Ströher, U., 2013a. Genomic analysis of loviruses associated with four viral
C.G. Albariño et al. / Virology 484 (2015) 259264 263
hemorrhagic fever outbreaks in Uganda and the Democratic Republic of the
Congo in 2012. Virology 442, 97100.
Albariño, C.G., Uebelhoer, L.S., Vincent, J.P., Khristova, M.L., Chakrabarti, A.K.,
McElroy, A., Nichol, S.T., Towner, J.S., 2013b. Development of a reverse genetics
system to generate recombinant Marburg virus derived from a bat isolate.
Virology 446, 230237.
Baize, S., 2015. Ebola virus in West Africa: new conquered territories and new risks-
or how I learned to stop worrying and (not) love Ebola virus. Curr. Opin. Virol.
10C, 7076.
Baize, S., Pannetier, D., Oestereich, L., Rieger, T., Koivogui, L., Magassouba, N., Soropogui, B.,
Sow,M.S.,Keita,S.,DeClerck,H.,Tiffany,A.,Dominguez,G.,Loua,M.,Traore,A.,Kolie,
M.,Malano,E.R.,Heleze,E.,Bocquin,A.,Mely,S.,Raoul,H.,Caro,V.,Cadar,D.,Gabriel,
M., Pahlmann, M., Tappe, D., Schmidt-Chanasit, J., Impouma, B., Diallo, A.K., Formenty,
P., Van Herp, M., Günther, S., 2014. Emergence of Zaire Ebola virus disease in Guinea.
N.Engl.J.Med.371,14181425.
CDC, 2015. Update: Ebola virus disease epidemic west Africa. MMWR Morb.
Mortal. Wkly. Rep. 64, 109110.
Chambers, R., Takimoto, T., 2010. Trafcking of Sendai virus nucleocapsids is
mediated by intracellular vesicles. PLoS One 5, e10994.
Crance, J.M., Scaramozzino, N., Jouan, A., Garin, D., 2003. Interferon, ribavirin, 6-
azauridine and glycyrrhizin: antiviral compounds active against pathogenic
aviviruses. Antiviral Res. 58, 7379.
Dixon, M.G., Schafer, I.J., Centers for Disease, C., Prevention, 2014. Ebola viral
disease outbreak West Africa. MMWR Morb. Mortal. Wkly. Rep. 63, 548551.
Dowall, S.D., Matthews, D.A., Garcia-Dorival, I., Taylor, I., Kenny, J., Hertz-Fowler, C.,
Hall, N., Corbin-Lickfett, K., Empig, C., Schlunegger, K., Barr, J.N., Carroll, M.W.,
Hewson, R., Hiscox, J.A., 2014. Elucidating variations in the nucleotide sequence
of Ebola virus associated with increasing pathogenicity. Genome Biol. 15, 540.
Duprex, W.P., Collins, F.M., Rima, B.K., 2002. Modulating the function of the measles
virus RNA-dependent RNA polymerase by insertion of green uorescent protein
into the open reading frame. J. Virol. 76, 73227328.
Ebihara, H., Groseth, A., Neumann, G., Kawaoka, Y., Feldmann, H., 2005. The role of
reverse genetics systems in studying viral hemorrhagic fevers. Thromb. Hae-
most. 94, 240253.
Ebihara, H., Theriault, S., Neumann, G., Alimonti, J.B., Geisbert, J.B., Hensley, L.E.,
Groseth, A., Jones, S.M., Geisbert, T.W., Kawaoka, Y., Feldmann, H., 2007. In vitro
and in vivo characterization of recombinant Ebola viruses expressing enhanced
green uorescent protein. J. Infect. Dis. 196 (Suppl. 2), S313S322.
Enterlein, S., Volchkov, V., Weik, M., Kolesnikova, L., Volchkova, V., Klenk, H.D.,
Muhlberger, E., 2006. Rescue of recombinant Marburg virus from cDNA is
dependent on nucleocapsid protein VP30. J. Virol. 80, 1038104 3.
Falzarano, D., Feldmann, H., 2014. Possible leap ahead in lovirus therapeutics. Cell
Res. 24, 647648.
Feldmann, H., Sanchez, A., Geisbert, T.W., 2013. Filoviridae: marburg and Ebola
viruses. In: Knipe, D.M.a.H., P.M (Ed.), Fields Virology, 6th ed. Lippincott,
Williams and Wilkins, Philadelphia, pp. 923956.
Gatherer, D., 2014. The 2014 Ebola virus disease outbreak in West Africa. J. Gen.
Virol. 95, 161916 24.
Gire, S.K., Goba, A., Andersen, K.G., Sealfon, R.S., Park, D.J., Kanneh, L., Jalloh, S.,
Momoh, M., Fullah, M., Dudas, G., Wohl, S., Moses, L.M., Yozwiak, N.L., Winnicki,
S., Matranga, C.B., Malboeuf, C.M., Qu, J., Gladden, A.D., Schaffner, S.F., Yang, X.,
Jiang, P.P., Nekoui, M., Colubri, A., Coomber, M.R., Fonnie, M., Moigboi, A.,
Gbakie, M., Kamara, F.K., Tucker, V., Konuwa, E., Saffa, S., Sellu, J., Jalloh, A.A.,
Kovoma, A., Koninga, J., Mustapha, I., Kargbo, K., Foday, M., Yillah, M., Kanneh, F.,
Robert, W., Massally, J.L., Chapman, S.B., Bochicchio, J., Murphy, C., Nusbaum, C.,
Young, S.,Birren, B.W., Grant, D.S., Scheiffelin, J.S., Lander, E.S., Happi, C., Gevao, S.
M., Gnirke, A., Rambaut, A., Garry, R.F., Khan, S.H., Sabeti, P.C., 2014. Genomic
surveillance elucidates Ebola virus origin and transmission during the 2014
outbreak. Science 345, 13691372.
Groseth, A., Marzi, A., Hoenen, T., Herwig, A., Gardner, D., Becker, S., Ebihara, H.,
Feldmann, H., 2012. The Ebola virus glycoprotein contributes to but is not
sufcient for virulence in vivo. PLoS Pathog. 8, e1002847.
Hartman, A.L., Bird, B.H., Towner, J.S., Antoniadou, Z.A., Zaki, S.R., Nichol, S.T., 2008.
Inhibition of IRF-3 activation by VP35 is critical for the high level of virulence of
Ebola virus. J. Virol. 82, 26992704.
Hartman, A.L., Dover, J.E., Towner, J.S., Nichol, S.T., 2006. Reverse genetic generation
of recombinant Zaire Ebola viruses containing disrupted IRF-3 inhibitory
domains results in attenuated virus growth in vitro and higher levels of IRF-3
activation without inhibiting viral transcription or replication. J. Virol. 80,
64306440.
Hartman, A.L., Towner, J.S., Nichol, S.T., 2010. Ebola and marburg hemorrhagic fever.
Clin. Lab. Med. 30, 161177.
Hoenen, T., Feldmann, H., 2014. Reverse genetics systems as tools for the develop-
ment of novel therapies against loviruses. Expert Rev. Anti-Infect. Ther. 12,
12531263.
Hoenen, T., Groseth, A., Callison, J., Takada, A., Feldmann, H., 2013. A novel Ebola
virus expressing luciferase allows for rapid and quantitative testing of anti-
virals. Antiviral Res. 99, 207213.
Hoenen, T., Groseth, A., de Kok-Mercado, F., Kuhn, J.H., Wahl-Jensen, V., 2011.
Minigenomes, transcription and replication competent virus-like particles and
beyond: reverse genetics systems for loviruses and other negative stranded
hemorrhagic fever viruses. Antiviral Res. 91, 195208.
Hoenen, T., Shabman, R.S., Groseth, A., Herwig, A., Weber, M., Schudt, G., Dolnik, O.,
Basler, C.F., Becker, S., Feldmann, H., 2012. Inclusion bodies are a site of
ebolavirus replication. J. Virol. 86, 1177911788.
Johnson, K.M., 1978. Ebola haemorrhagic fever in Zaire, 1976. Bull. World Health
Organ. 56, 271293.
Johnson, K.M., Lange, J.V., Webb, P.A., Murphy, F.A., 1977. Isolation and partial
characterisation of a new virus causing acute haemorrhagic fever in Zaire.
Lancet 1, 569571.
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 efciency of a 2A peptide derived from porcine teschovirus-
1 in human cell lines, zebrash and mice. PLoS One 6, e18556.
Leroy, E.M., Gonzalez, J.P., Baize, S., 2011. Ebola and Marburg haemorrhagic fever
viruses: major scientic advances, but a relatively minor public health threat
for Africa. Clin. Microbiol. Infect. 17, 964976.
Lo, M.K., Nichol, S.T., Spiropoulou, C.F., 2014. Evaluation of luciferase and GFP-
expressing Nipah viruses for rapid quantitative antiviral screening. Antiviral
Res. 106, 5360.
Martinez, M.J., Volchkova, V.A., Raoul, H., Alazard-Dany, N., Reynard, O., Volchkov,
V.E, 2011. Role of VP30 phosphorylation in the Ebola virus replication cycle.
J. Infect. Dis. 204 (Suppl. 3), S934S940.
Morrey, J.D., Smee, D.F., Sidwell, R.W., Tseng, C., 2002. Identication of active
antiviral compounds against a New York isolate of West Nile virus. Antiviral
Res. 55, 107116.
Neumann, G., Feldmann, H., Watanabe, S., Lukashevich, I., Kawaoka, Y., 2002.
Reverse genetics demonstrates that proteolytic processing of the Ebola virus
glycoprotein is not essential for replication in cell culture. J. Virol. 76, 406410.
Niwa, H., Yamamura, K., Miyazaki, J., 1991. Efcient selection for high-expression
transfectants with a novel eukaryotic vector. Gene 108, 193199.
Panchal, R.G., Kota, K.P., Spurgers, K.B., Ruthel, G., Tran, J.P., Boltz, R.C., Bavari, S.,
2010. Development of high-content imaging assays for lethal viral pathogens.
J. Biomol. Screen. 15, 755765.
Panchal, R.G., Reid, S.P., Tran, J.P., Bergeron, A.A., Wells, J., Kota, K.P., Aman, J., Bavari,
S., 2012. Identication of an antioxidant small-molecule with broad-spectrum
antiviral activity. Antiviral Res. 93, 2329.
Pyrc, K., Bosch, B.J., Berkhout, B., Jebbink, M.F., Dijkman, R., Rottier, P., van der Hoek,
L., 2006. Inhibition of human coronavirus NL63 infection at early stages of the
replication cycle. Antimicrob. Agents Chemother. 50, 20002008.
Schmidt, K.M., Schumann, M., Olejnik, J., Krahling, V., Muhlberger, E., 2011.
Recombinant Marburg virus expressing EGFP allows rapid screening of virus
growth and real-time visualization of virus spread. J. Infect. Dis. 204 (Suppl. 3),
S861S870.
Schudt, G., Kolesnikova, L., Dolnik, O., Sodeik, B., Becker, S., 2013. Live-cell imaging
of Marburg virus-infected cells uncovers actin-dependent transport of nucleo-
capsids over long distances. Proc. Natl. Acad. Sci. USA 110, 1440214407.
Silin, D., Lyubomska, O., Ludlow, M., Duprex, W.P., Rima, B.K., 2007. Development of
a challenge-protective vaccine concept by modication of the viral RNA-
dependent RNA polymerase of canine distemper virus. J. Virol. 81,
1364913658.
Smee, D.F., McKernan, P.A., Nord, L.D., Willis, R.C., Petrie, C.R., Riley, T.M., Revankar,
G.R., Robins, R.K., Smith, R.A., 1987. Novel pyrazolo[3,4-d]pyrimidine nucleoside
analog with broad-spectrum antiviral activity. Antimicrob. Agents Chemother.
31, 15351541.
Stadler, T., Kuhnert, D., Rasmussen, D.A., du Plessis, L., 2014. Insights into the early
epidemic spread of ebola in sierra leone provided by viral sequence data. PLoS Curr. 6,
Oct 6 . Edition 1. 10.1371/currents.outbreaks.02bc6d927ecee7bbd33532ec8ba6a25f.
Theriault, S., Groseth, A., Artsob, H., Feldmann, H., 2005. The role of reverse genetics
systems in determining lovirus pathogenicity. Arch. Virol. Suppl. 19, 157177 .
Towner, J.S., Paragas, J., Dover, J.E., Gupta, M., Goldsmith, C.S., Huggins, J.W., Nichol,
S.T., 2005. Generation of eGFP expressing recombinant Zaire Ebola virus for
analysis of early pathogenesis events and high-throughput antiviral drug
screening. Virology 332, 2027.
Tsuda, Y., Hoenen, T., Banadyga, L., Weisend, C., Ricklefs, S.M., Porcella, S.F., Ebihara,
H., 2015. An Improved reverse genetics system to overcome cell-type-
dependent Ebola virus genome plasticity. J. Infect. Dis., Mar 24. pii: jiu681.
[Epub ahead of print].
Uebelhoer, L.S., Albariño, C.G., McMullan, L.K., Chakrabarti, A.K., Vincent, J.P., Nichol,
S.T., Towner, J.S., 2014. High-throughput, luciferase-based reverse genetics
systems for identifying inhibitors of Marburg and Ebola viruses. Antiviral Res.
106, 8694.
Volchkov, V.E., Volchkova, V.A., Muhlberger, E., Kolesnikova, L.V., Weik, M., Dolnik,O.,
Klenk, H.D., 2001. Recovery of infectious Ebola virus from complementary DNA:
RNA editing of the GP gene and viral cytotoxicity. Science 291, 196519 69.
C.G. Albariño et al. / Virology 484 (2015) 259264264
... chosen because this minigenome performed best in the minigenome assay (Figs 1B and S1). Since there are no commercially available antibodies against LLOV and we detected no crossreactivity with LLOV proteins using antibodies against other filoviruses, we also constructed these rLLOV comp full-length clones with the addition of a ZsGreen reporter attached to the VP40 gene via a P2A sequence as previously described for EBOV [10]. Viral rescue was attempted by transfecting cocultures of African green monkey kidney (Vero E6) and human hepatocarcinoma (Huh7) cells with full-length clone rLLOV comp plasmids along with codonoptimized LLOV support plasmids as previously described for the LLOV minigenome system [9]. ...
... DNA fragments comprising a portion of the LLOV L open reading frame (nucleotides 18447-18820) followed by EBOV (nucleotides 18220-18958, EBOV UTR+tr ), or MARV sequences (nucleotides 18477-19111, MARV UTR+tr ) and ending with same portion of the HdV rizbozyme SuperCutII sequence as used in Fragment 1 were synthesized (Twist Biosciences). A DNA portion of Fragment 2 containing an insertion of the ZsGreen-P2A upstream of the VP40 ORF as described previously [10] was synthesized (Twist Biosciences). Finally, a DNA fragment introducing overlapping gene start and end signals into the intergenic region of the VP24-L IR (GAAGAATATTAAGAAAAA between nucleotides 12001 and 12002, IR ins ) was synthesized. ...
... For the construction of the EBOV-ZsGreen and RESTV-ZsGreen full-length plasmids, DNA fragments consisting of EBOV nucleotides 2148-5773 and RESTV nucleotides 2555-5287 each with an insert of the ZsGreen-P2A upstream of the VP40 ORF as described previously [10] were synthesized (Twist Biosciences). These fragments were cloned into the p15AK-EBOV [44] and p15AK-RESTV (kind gift of Thomas Hoenen, Friedrich Loeffler Institute) [35] via SphI or NcoI and SpeI, respectively. ...
Article
Full-text available
Next generation sequencing has revealed the presence of numerous RNA viruses in animal reservoir hosts, including many closely related to known human pathogens. Despite their zoonotic potential, most of these viruses remain understudied due to not yet being cultured. While reverse genetic systems can facilitate virus rescue, this is often hindered by missing viral genome ends. A prime example is Lloviu virus (LLOV), an uncultured filovirus that is closely related to the highly pathogenic Ebola virus. Using minigenome systems, we complemented the missing LLOV genomic ends and identified cis-acting elements required for LLOV replication that were lacking in the published sequence. We leveraged these data to generate recombinant full-length LLOV clones and rescue infectious virus. Similar to other filoviruses, recombinant LLOV (rLLOV) forms filamentous virions and induces the formation of characteristic inclusions in the cytoplasm of the infected cells, as shown by electron microscopy. Known target cells of Ebola virus, including macrophages and hepatocytes, are permissive to rLLOV infection, suggesting that humans could be potential hosts. However, inflammatory responses in human macrophages, a hallmark of Ebola virus disease, are not induced by rLLOV. Additional tropism testing identified pneumocytes as capable of robust rLLOV and Ebola virus infection. We also used rLLOV to test antivirals targeting multiple facets of the replication cycle. Rescue of uncultured viruses of pathogenic concern represents a valuable tool in our arsenal for pandemic preparedness.
... Fluorescent reporter proteins have also been demonstrably useful to study viral pathogenesis and tissue tropism [7,8,14,15]; however, most of these studies are limited to in situ or ex vivo approaches. Reporter EBOV and Marburg viruses [17][18][19][20][21] have been used to study filovirus pathogenesis [20,22,23] and in highthroughput in vitro drug screening [20][21][22][23][24][25][26][27]. However, thus far, reverse-genetics approaches to study filovirus infection using reporter proteins have been limited to wild-type, un-adapted viruses only, with no reporter-protein expressing species-adapted Ebolaviruses available. ...
... Single-reporter rMA-EBOVs were generated by inserting nLuc (rMA-EBOV/nLuc) or ZsG (rMA-EBOV/ZsG) coding sequence upstream of the VP40 coding sequence ( Figure 1A). While including ZsG alongside VP40 of WT-EBOV has been demonstrated to minimally deter viral growth [17], the impact of including two reporter proteins on EBOV fitness is unknown. Thus, dual-reporter rMA-EBOV was produced using two strategies: inserting ZsG and nLuc coding sequences as fusion proteins upstream of the VP40 coding sequence (rMA-EBOV/Zn) or inserting ZsG and nLuc coding sequences upstream of NP and VP40, respectively (rMA-EBOV/nZ). ...
Article
Ebola virus (EBOV) causes lethal disease in humans but not in mice. Here, we generated recombinant mouse-adapted (MA)-EBOVs, including one based on the previously reported serially adapted strain (rMA-EBOV), along with single-reporter rMA-EBOVs expressing either fluorescent (ZsGreen1 [ZsG]) or bioluminescent (nano-luciferase [nLuc]) reporters, and dual-reporter rMA-EBOVs expressing both ZsG and nLuc. No detriment to viral growth in vitro was seen with inclusion of MA-associated mutations or reporter proteins. In CD-1 mice, infection with MA-EBOV, rMA-EBOV, and single-reporter rMA-EBOVs conferred 100% lethality; infection with dual-reporter rMA-EBOV resulted in 80% lethality. Bioluminescent signal from rMA-EBOV expressing nLuc was detected in vivo and ex vivo using the IVIS Spectrum CT. Fluorescent signal from rMA-EBOV expressing ZsG was detected in situ using hand-held blue-light transillumination and ex vivo through epi-illumination with the IVIS Spectrum CT. These data support the use of reporter MA-EBOV for studies of Ebola virus in animal disease models.
... Recombinant Ebola viruses expressing a fluorescent reporter gene have been widely used as valuable tools in tropism studies, live-cell imaging, inactivation studies, and antiviral drug screening assays [17,[27][28][29]. Incorporating a reporter gene into recombinant EBOV currently has been achieved with a variety of methods including introducing additional transcriptional units, expressing reporters as fusion proteins with viral proteins, and using a 2A peptide system [17,27,30]. Drawbacks to these We first examined our ZsG-containing intein (ZsG-Int) for the ability to both produce a fluorescent signal and splice efficiently in Escherichia coli when using the maltose-binding protein (MBP) as the N-extein and a His-tag as the C-extein ( Figure 1B). ...
... Recombinant Ebola viruses expressing a fluorescent reporter gene have been widely used as valuable tools in tropism studies, live-cell imaging, inactivation studies, and antiviral drug screening assays [17,[27][28][29]. Incorporating a reporter gene into recombinant EBOV currently has been achieved with a variety of methods including introducing additional transcriptional units, expressing reporters as fusion proteins with viral proteins, and using a 2A peptide system [17,27,30]. Drawbacks to these systems exist; however, additional transcriptional units can interfere with ratios of viral mRNAs and proteins, fusion proteins can reduce the functionality of viral proteins, and the 2A system incorporates potentially undesired RNA secondary structures into viral genomes and leaves either a residual N-terminal proline or C-terminal 2A peptide on the viral protein [13]. ...
Article
Full-text available
Inteins (intervening proteins) are polypeptides that interrupt the sequence of other proteins and remove themselves through protein splicing. In this intein-catalyzed reaction, the two peptide bonds surrounding the intein are rearranged to release the intein from the flanking protein sequences, termed N- and C-exteins, which are concurrently joined by a peptide bond. Because of this unique functionality, inteins have proven exceptionally useful in protein engineering. Previous work has demonstrated that heterologous proteins can be inserted within an intein, with both the intein and inserted protein retaining function, allowing for intein-containing genes to coexpress additional coding sequences. Here, we show that a fluorescent protein (ZsGreen) can be inserted within the Pyrococcus horikoshii RadA intein, with the hybrid protein (ZsG-Int) maintaining fluorescence and splicing capability. We used this system to create a recombinant Ebola virus expressing a fluorescent protein. We first tested multiple potential insertion sites for ZsG-Int within individual Ebola virus proteins, identifying a site within the VP30 gene that facilitated efficient intein splicing in mammalian cells while also preserving VP30 function. Next, we successfully rescued a virus containing the ZsG-Int-VP30 fusion protein, which displayed fluorescence in the infected cells. We thus report a new intein-based application for adding reporters to systems without the need to add additional genes. Further, this work highlights a novel reporter design, whereby the reporter is only made if the protein of interest is translated and does not remain fused to the protein of interest.
... To facilitate high-throughput assays with authentic SUDV (variant Gulu), we rescued a recombinant virus that expresses the fluorescent reporter ZsGreen (ZSG) at the N-terminus of viral protein 40 (VP40). Separation of ZSG and VP40 is achieved by the P2A peptide [4], an insertion strategy shown to work well in rescue of EBOV [5]. In short, viral antigenomic RNA is produced from a rescue plasmid by transfected T7 RNA polymerase and replication is initiated by EBOV RNA-dependent RNA polymerase (L), nucleoprotein (NP), and VP30 and VP35 proteins provided in trans (Supplementary Materials and Methods, Supplementary Table 2). ...
Article
Full-text available
Ebola disease outbreaks are major public health events because of human-to-human transmission and high mortality. These outbreaks are most often caused by Ebola virus, but at least three related viruses can also cause the disease. In 2022, Sudan virus re-emerged causing more than 160 confirmed and probable cases. This report describes generation of a recombinant Sudan virus and demonstrates its utility by quantifying antibody cross-reactivity between Ebola and Sudan virus glycoproteins after human infection or vaccination with a licensed Ebola virus vaccine.
... It is therefore possible that antibodies targeted to a non-homologous region of the EBOV-GP could be missed when comparing binding and subsequent ADCD on the EBOV-sGP. A comparison of the full-length genomes of representative EBOV isolates shows an estimated 97% sequence identity between the Makona and Mayinga variants used in this study (55). ...
Article
Full-text available
The 2013–2016 Ebola virus (EBOV) epidemic in West Africa was unprecedented in case numbers and fatalities, and sporadic outbreaks continue to arise. Antibodies to the EBOV glycoprotein (GP) are strongly associated with survival and their use in immunotherapy is often initially based on their performance in neutralisation assays. Other immune effector functions also contribute to EBOV protection but are more complex to measure. Their interactions with the complement system in particular are comparatively under-researched and commonly excluded from cellular immunoassays. Using EBOV convalescent plasma samples from the 2013–2016 epidemic, we investigated antibody and complement-mediated neutralisation and how these interactions can influence immunity in response to EBOV-GP and its secreted form (EBOV-sGP). We defined two cohorts: one with low-neutralising titres in relation to EBOV-GP IgG titres (LN cohort) and the other with a direct linear relationship between neutralisation and EBOV-GP IgG titres (N cohort). Using flow cytometry antibody-dependent complement deposition (ADCD) assays, we found that the LN cohort was equally efficient at mediating ADCD in response to the EBOV-GP but was significantly lower in response to the EBOV-sGP, compared to the N cohort. Using wild-type EBOV neutralisation assays with a cohort of the LN plasma, we observed a significant increase in neutralisation associated with the addition of pooled human plasma as a source of complement. Flow cytometry ADCD was also applied using the GP of the highly virulent Sudan virus (SUDV) of the Sudan ebolavirus species. There are no licensed vaccines or therapeutics against SUDV and it overlaps in endemicity with EBOV. We found that the LN plasma was significantly less efficient at cross-reacting and mediating ADCD. Overall, we found a differential response in ADCD between LN and N plasma in response to various Ebolavirus glycoproteins, and that these interactions could significantly improve EBOV neutralisation for selected LN plasma samples. Preservation of the complement system in immunoassays could augment our understanding of neutralisation and thus protection against infection
... Cut banana, mango, and bat mix samples ( ∼ =113 g) contained within 12-well tissue culture plates (Corning Inc., Corning, NY, USA) were inoculated in triplicate with 250 µL of high (1.00 × 10 5 TCID 50 ), medium (1.00 × 10 3 TCID 50 ), and low (1.00 × 10 1 TCID 50 ) doses of replication-competent, infectious recombinant (r) MARV expressing the fluorescent ZsGreen1 (ZsG) (rMARV-ZsG; GenBank accession number: MK271062; Huh7+2 passages; [39,40]), gently mixed with a polyester-tipped applicator to distribute the virus, and then incubated at room temperature for 24 HPI. Each rMARV-ZsG-inoculated fruit specimen was sampled by gently swabbing the surface of each piece of fruit in a well using a polyester-tipped applicator at 0, 1, 6, and 24 HPI. ...
Article
Full-text available
Marburg virus (MARV), the causative agent of Marburg virus disease, emerges sporadically in sub-Saharan Africa and is often fatal in humas. The natural reservoir for this zoonotic virus is the frugivorous Egyptian rousette bat (Rousettus aegyptiacus) that when infected, sheds virus in the highest amounts in oral secretions and urine. Being fruit bats, these animals forage nightly for ripened fruit throughout the year, including those types often preferred by humans. During feeding, they continually discard partially eaten fruit on the ground that could then be consumed by other Marburg virus susceptible animals or humans. In this study, using qRT-PCR and virus isolation, we tested fruit discarded by Egyptian rousette bats experimentally infected with a natural bat isolate of Marburg virus. We then separately tested viral persistence on fruit varieties commonly cultivated in sub-Saharan Africa using a recombinant Marburg virus expressing the fluorescent ZsGreen1. Marburg virus RNA was repeatedly detected on fruit in the food bowls of the infected bats and viable MARV was recovered from inoculated fruit for up to 6 h.
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
Comparable to the related Ebola virus, Marburg virus is an emerging zoonotic pathogen that causes hemorrhagic fever with a high mortality rate. Therefore, handling of Ebola virus and Marburg virus is limited to biosafety level 4 facilities, of which only a limited number exists worldwide. However, researchers have developed several virus alternatives that are safe to handle in lower biosafety settings. One particularly interesting approach is the engineering of biologically contained Ebola virus by removing an essential gene from the virus genome and providing this missing gene in trans in a specific cell line. Because the virus is confined to this specific cell line, this results in a system that is safe to handle. So far, Ebola virus is the only virus for which biological containment has been reported. Here, we describe the first successful rescue of biologically contained Marburg virus and demonstrate that biological containment is also feasible for other filoviruses. Specifically, we describe the development of containment cell lines for Marburg virus through lentiviral transduction and show the growth and safety characteristics of eGFP-expressing, biologically contained Marburg virus in these cell lines. Additionally, we exploited this newly established Marburg virus system to screen over 500 compounds from available libraries. Lastly, we also validated the applicability of our biologically contained Marburg virus system in a 384-well format, to further illustrate the usefulness of this novel system as an alternative for high-throughput MARV screening of compound libraries.
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.
Article
Full-text available
CDC is assisting ministries of health and working with other organizations to end the ongoing epidemic of Ebola virus disease (Ebola) in West Africa. The updated data in this report were compiled from situation reports from the Guinea Interministerial Committee for Response Against the Ebola Virus, the Liberia Ministry of Health and Social Welfare, the Sierra Leone Ministry of Health and Sanitation, and the World Health Organization.
Article
Full-text available
On March 21, 2014, the Guinea Ministry of Health reported the outbreak of an illness characterized by fever, severe diarrhea, vomiting, and a high case-fatality rate (59%) among 49 persons. Specimens from 15 of 20 persons tested at Institut Pasteur in Lyon, France, were positive for an Ebola virus by polymerase chain reaction. Viral sequencing identified Ebola virus (species Zaïre ebolavirus), one of five viruses in the genus Ebolavirus, as the cause. Cases of Ebola viral disease (EVD) were initially reported in three southeastern districts (Gueckedou, Macenta, and Kissidougou) of Guinea and in the capital city of Conakry. By March 30, cases had been reported in Foya district in neighboring Liberia (1), and in May, the first cases identified in Sierra Leone were reported. As of June 18, the outbreak was the largest EVD outbreak ever documented, with a combined total of 528 cases (including laboratory-confirmed, probable, and suspected cases) and 337 deaths (case-fatality rate = 64%) reported in the three countries. The largest previous outbreak occurred in Uganda during 2000-2001, when 425 cases were reported with 224 deaths (case-fatality rate = 53%). The current outbreak also represents the first outbreak of EVD in West Africa (a single case caused by Taï Forest virus was reported in Côte d'Ivoire in 1994 [3]) and marks the first time that Ebola virus transmission has been reported in a capital city.
Article
Full-text available
Background Ebolaviruses causes a severe and often fatal hemorrhagic fever in humans, with some species such as Ebola virus having case fatality rates approaching 90%. Currently the worst Ebola virus outbreak since the disease was discovered is occurring in West Africa. Although thought to be a zoonotic infection, a concern is that with increasing numbers of humans being infected, Ebola virus variants could be selected which are better adapted for human-to-human transmission.ResultsTo investigate whether genetic changes in Ebola virus become established in response to adaptation in a different host, a guinea pig model of infection was used. In this experimental system, guinea pigs were infected with Ebola virus (EBOV), which initially did not cause disease. To simulate transmission to uninfected individuals, the virus was serially passaged five times in naive animals. As the virus was passaged, virulence increased and clinical effects were observed in the guinea pig. An RNAseq and consensus mapping approach was then used to evaluate potential nucleotide changes in the Ebola virus genome at each passage.Conclusions Upon passage in the guinea pig model, EBOV become more virulent, RNA editing and also coding changes in key proteins become established. The data suggest that the initial evolutionary trajectory of EBOV in a new host can lead to a gain in virulence. Given the circumstances of the sustained transmission of EBOV in the current outbreak in West Africa, increases in virulence may be associated with prolonged and uncontrolled epidemics of EBOV.
Article
Reverse genetics systems represent a key technique for studying replication and pathogenesis of viruses, including Ebola virus (EBOV). During the rescue of recombinant EBOV from Vero cells, a high frequency of mutations was observed throughout the genomes of rescued viruses, including at the RNA editing site of the glycoprotein gene. The influence that such genomic instability could have on downstream uses of rescued virus may be detrimental, and we therefore sought to improve the rescue system. Here we report an improved EBOV rescue system with higher efficiency and genome stability, using a modified full-length EBOV clone in Huh7 cells. Moreover, by evaluating a variety of cells lines, we revealed that EBOV genome instability is cell-type dependent, a fact that has significant implications for the preparation of standard virus stocks. Thus, our improved rescue system will have an impact on both basic and translational research in the filovirus field. Published by Oxford University Press on behalf of the Infectious Diseases Society of America 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.
Article
After being restricted to Central Africa for 35 years, the Ebola virus has suddenly emerged in Guinea in early 2014. The virus rapidly spread to Liberia and Sierra Leone and was imported to Nigeria, Senegal, Mali and even USA. The main features of this outbreak are a relentless dissemination among several countries, a dramatic number of cases including health-care workers and an inability to control the outbreak which grows exponentially. This conquest of new territories by Ebola virus implies that new risks have now to be taken into account. Filoviruses have been promoted from the status of neglected diseases to that of international public health and security concerns and we now have to live with that threat. A vaccine is probably the only efficient approach to avoid future re-emergence. Copyright © 2015 Elsevier B.V. All rights reserved.
Article
The current Ebola virus epidemic in West Africa has been spreading at least since December 2013. The first confirmed case of Ebola virus in Sierra Leone was identified on May 25. Based on viral genetic sequencing data from 72 individuals in Sierra Leone collected between the end of May and mid June, we utilize a range of phylodynamic methods to estimate the basic reproductive number (R0). We additionally estimate the expected lengths of the incubation and infectious periods of the virus. Finally, we use phylogenetic trees to examine the role played by population structure in the epidemic. The median estimates of R0 based on sequencing data alone range between 1.65-2.18, with the most plausible model yielding a median R0 of 2.18 (95% HPD 1.24-3.55). Importantly, our results indicate that, at least until mid June, relief efforts in Sierra Leone were ineffective at lowering the effective reproductive number of the virus. We estimate the expected length of the infectious period to be 2.58 days (median; 95% HPD 1.24-6.98). The dataset appears to be too small in order to estimate the incubation period with high certainty (median expected incubation period 4.92 days; 95% HPD 2.11-23.20). While our estimates of the duration of infection tend to be smaller than previously reported, phylodynamic analyses support a previous estimate that 70% of cases were observed and included in the present dataset. The dataset is too small to show a particular population structure with high significance, however our preliminary analyses suggest that half the population is spreading the virus with an R0 well above 2, while the other half of the population is spreading with an R0 below 1. Overall we show that sequencing data can robustly infer key epidemiological parameters. Such estimates inform public health officials and help to coordinate effective public health efforts. Thus having more sequencing data available for the ongoing Ebola virus epidemic and at the start of new outbreaks will foster a quick understanding of the dynamics of the pathogen.