Sei sulla pagina 1di 8

Journal of Virological Methods 182 (2012) 1–8

Contents lists available at SciVerse ScienceDirect

Journal of Virological Methods


journal homepage: www.elsevier.com/locate/jviromet

Development of an in vitro system to measure the sensitivity to the antiviral Mx


protein of fish viruses
Katherine Lester, Malcolm Hall, Katy Urquhart, Suresh Gahlawat 1 , Bertrand Collet ∗
Marine Scotland, Marine Laboratory, 375 Victoria Road, Aberdeen AB11 9DB, United Kingdom

a b s t r a c t

Article history: Mx is a structural protein, induced by type I interferon (IFN), with direct antiviral properties. In fish
Received 18 November 2011 the inherent contribution of Mx protein to viral protection is unknown. The transgenic Chinook salmon
Received in revised form 12 January 2012 embryonic (CHSE)-TOF cell line was genetically modified to express the rainbow trout Mx (rbtMx1) pro-
Accepted 16 January 2012
tein under the control of the tetracycline derivative, doxycycline (DOX). Two clones CHSE-TOF-MX8 and
Available online 1 March 2012
CHSE-TOF-MX10 were isolated and characterised by qPCR. The level of resistance to Infectious Pancreatic
Necrosis Virus (IPNV), Salmon Alphavirus (SAV), Infectious Haematopoietic Necrosis Virus (IHNV) and
Keywords:
Epizootic Haematopoietic Necrosis Virus (EHNV) of the CHSE-TOF, CHSE-TOF-MX8 and CHSE-TOF-MX10
Mx protein
Interferon
cell lines cultivated with and without DOX was measured. A novel method was established to measure
Salmon accurately the level of sensitivity of any given viral isolate to Mx protein. IPNV and SAV viruses were
Tet-Off highly sensitive to the presence of rbtMx1 in the cells whereas IHNV and EHNV showed partial resistance
Viral resistance suggesting contrasting viral evasion strategies between these categories of viruses.
Crown Copyright © 2012 Published by Elsevier B.V. All rights reserved.

1. Introduction range of RNA viruses. It accumulates in the cytoplasm of IFN-treated


cells, associating partly with the endoplasmic reticulum. It inter-
Mx is an antiviral protein that was discovered initially as an feres with the transport of the Bunyavirus nucleocapsid protein
agent conferring protection to myxoviridae (Haller et al., 1979, (N) to the Golgi apparatus where the virus assembles naturally
1980; Horisberger et al., 1980; Lindenmann, 1964; Staeheli et al., and also prevents the incoming Thogoto virus nucleocapsids from
1986). In mice, disruption of the single Mx1 gene causes complete being transported into the nucleus, the site of viral transcription
loss of innate immunity against mouse-adapted influenza virus, and replication (Kochs and Haller, 1999a,b). However, the contri-
leading to overwhelming infection and rapid death (Haller, 1981; bution of the Mx protein to the general IFN-dependent protection
Haller et al., 1998). against viruses is not clear and very little is known on the inher-
The Mx gene is induced by type I interferon (IFN) the main ent ability of teleost Mx protein to prevent viral replication. The
cytokine regulating innate antiviral immune functions (see for artificial over expression of Mx gene in mammalian and fish cells
review, Arnheiter et al., 1990; Haller et al., 2007a,b). In fish, IFN has been shown to confer resistance to a wide range of viruses. In
activity has been detected for decades and a gene encoding IFN fish, over expression of the Mx gene has provided different levels
was isolated for the first time in 2003 in zebrafish (Altmann et al., of resistance to Infectious Pancreatic Necrosis Virus (IPNV), Infec-
2003). Before the discovery of the IFN genes, Mx was used as a tious Salmon Anaemia Virus (ISAV), and sole aquabirnavirus in the
marker of the presence of IFN by measurement of the induction of Chinook salmon embryonic (CHSE) cell line (Larsen et al., 2004;
its gene. Mx is a member of dynamin superfamily of large GTPase Fernández-Trujillo et al., 2008; Kibenge et al., 2005), reovirus in
characterised by a large molecular weight (75 kDa), and the abil- rare minnow (Su et al., 2009), Hirame rhabdovirus (HIRRV) and
ity to self-assemble (Haller et al., 2007a,b; Lee and Vidal, 2002). In Viral Hemorrhagic Septicaemia Virus (VHSV) in the Hirame Natu-
humans, MxA was found to have antiviral activity against a wide ral Embryo (HINAE) cell line (Caipang et al., 2003), Yellow Grouper
Nervous Necrosis Virus (YGNNV) in grouper brain 3 (GB3) cells
(Lin et al., 2006), and nodavirus in grouper fin cells GF-1 (Chen
et al., 2008). In contrast, it does not provide good protection against
∗ Corresponding author at: Immunology and Infection, Marine Scotland, Marine Infectious Haematopoietic Necrosis Virus (IHNV) in CHSE cells
Laboratory, PO Box 101, 375 Victoria Road, Aberdeen AB11 9DB, United Kingdom. (Trobridge et al., 1997) or against iridovirus in a baramundi cell line,
Tel.: +44 01224 425512; fax: +44 01224 295511.
cBB (Wu and Chi, 2007). Silencing experiments have also demon-
E-mail address: Bertrand.Collet@scotland.gsi.gov.uk (B. Collet).
1
Permanent address: Department of Biotechnology, Chaudhary Devi Lal Univer- strated the involvement of Mx protein in protection against viral
sity, Sirsa-125055, India. nervous necrosis (VNN), nodavirus and IPNV in cBB cells (Wu et al.,

0166-0934/$ – see front matter. Crown Copyright © 2012 Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.jviromet.2012.01.014
2 K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8

Table 1
Names and sequences of all primers used in this study.

Name Function Sequence 5 –3 % efficiency

MX1F1 GCAGTGGGCATCAGATAGCAGAAA
MX1F2 CTTGCTTTTTATTTAGGTTGTATC
Amplification of the full length Mx ORF
MX1R1 CAGCAGGAATATAAGCCACAAGTT
MX1R2 TTATCAGGCAGGTTCCCACTCCAC
MX1MLUIF GGGGGGACGCGTATGAATAATACGCTCAACCAA
Subcloning into pTRE2-hyg
MX1SALIR GGGGGGTCGACTAGAACTCAACTAGGTAGCT
TRE2F AAAGTGAAAGTCGAGCTCGGTACC
Sequencing
TRE2R CACCCTGAAAACTTTGCCCC
ELF-F CCCCTCCAGGACGTTTACAAA 98.3
ELF-R qPCR housekeeping control CACACGGCCCACAGGTACA
ELF-p (MGB) 6FAM-ATCGGTGGTATTGGAAC
tMX-F GATGCTGCACCTCAAGTCCTACTA 87.7
qPCR verification of level of MX expression in
tMX-R CGGATCACCATGGGAATCTGA
CHSE-TOF-MX clones
tMX-p (MGB) 6FAM-CTGGATATCCAGTCAGCGTC

2010; Wu and Chi, 2007). These independent studies are based on 2.2. Construction of the pTRE2hyg-rbtMx1 plasmid
different fish species, cellular systems and methods. It is therefore
difficult to compare the levels of resistance against different viruses The full coding region from the rbtMx1 was obtained by 3
conferred by Mx. rounds of PCR amplification using the high fidelity antibody-based
In rainbow trout, three Mx isoforms have been isolated among Hot Start KOD DNA PCR kit (Takara Bio Europe/Clontech, Saint-
which is rbtMx1. When over-expressed in CHSE-214 cells, rbtMx1 Germain-en-Laye, France). The reaction mix was as follows in a
is located in the cytoplasm (Trobridge and Leong, 1995; Trobridge 25 ␮l reaction: 2.5 ␮l 10× buffer for KOD Hot Start DNA poly-
et al., 1997). It shares 99% sequence identity with the rbtMx3 iso- merase, 1.5 ␮l 25 mM MgSO4 , 2.5 ␮l 2 mM each dNTP, 0.75 ␮l
form and relates to the Atlantic salmon Mx1 and Mx2, respectively 10 ␮M each primer, 1 ␮l template, 0.5 ␮l 1 U/␮l KOD Hot Start
(Robertsen et al., 1997) and its gene has been used as a marker DNA polymerase and 15.5 ␮l PCR grade water. The sequences of
for IFN activity in fish for many years (Collet et al., 2003; McBeath primers used (designed from Genbank OMU30253) and cycling
et al., 2007). Based on their sub cellular localisation, rbtMx1 and 3 condition are given in detail in Tables 1 and 2, respectively.
and Atlantic salmon Mx1 and Mx2, are more related to the human The template for the first round PCR was a pool of cDNA made
MxA, than to the MxB (Altmann et al., 2004). from RNA purified from kidney from two rainbow trout infected
In order to study the inherent role of Mx protein in the cellular with VHSV. The first round of PCR was carried out with MX1F1
protection against a wide range of viruses, a CHSE inducible cell and MX1R1 primers generating a 2.2 kb fragment that was sepa-
line expressing the rainbow trout Mx1 gene encoding for the rain- rated on a 1% Agarose–Ethidium Bromide gel, excised and purified
bow trout Mx1 isoform (rbtMx1) was established. The comparison using MiniElute purification kit according to the manufacturer’s
of viral sensitivity of these new cell lines gives some novel infor- instructions (Qiagen, Crawley, UK). The second round of PCR was
mation on the contribution of Mx protein to the viral resistance in performed with MX1F2 and MX1R2 primers on un-diluted purified
salmonids. first round PCR product as template, generating a 2.1 kb fragment
that was separated and purified as described above. The third
round was carried out on un-diluted second round PCR product
2. Materials and methods as template and with the primers MX1MLUIF and MX1SALIR con-
taining a MluI and SalI restriction site, respectively. It generated a
2.1. Cell culture 1.8 kb fragment that was separated and purified as described above.
Two microlitres of purified PCR product was digested overnight at
The recombinant cell line CHSE-TOF (Collet and Lester, 2011a) 37 ◦ C with MluI and SalI (Promega, Southampton, UK) alongside the
was grown as monolayer in Eagle’s Minimum Essential Medium pTRE2hyg plasmid (Takara Bio Europe/Clontech, Saint-Germain-
(Life Technologies, Paisley, UK) supplemented with 10% foetal en-Laye, France). Both digested fragments were separated, purified
bovine serum (FBS, PAA Laboratories, Pasching, Austria), 20 mM from gel as described above and ligated using the T4 DNA ligase
l-glutamine (Life Technologies, Paisley, UK) and 500 ␮g/ml G418 and 2× ligation mix (Promega) at 16 ◦ C for 16 h. TOP10 compe-
(Life Technologies, Paisley, UK). Stable cell lines CHSE-TOF-MX8 tent cells (Life Technologies, Paisley, UK) were transformed using
and CHSE-TOF-MX10 were selected and maintained in the above 5 ␮l of the ligation mix and plated on LB-Agar 100 ␮g/ml Ampi-
medium supplemented with 16 ␮g/ml Hygromycin (Sigma, Irvine, cillin plates. In parallel, TOP10 cells were transformed with 1 ␮l of
UK) and 1 ␮g/ml DOX (Sigma, Irvine, UK). To induce Mx protein in pTet-Off and pTRE2hyg-Luc vectors (Takara Bio Europe/Clontech,
CHSE-TOF-MX8 or CHSE-TOF-MX10 DOX was omitted.

Table 2
Primer combinations and cycling protocols used in this study for plasmid construction and sequence verification.

Use Primers Cycling protocol

Forward Reverse 1 cycle 35 cycles 1 cycle 1 cycle

Round I PCR MX1F1 MX1R1 95 ◦ C 95 ◦ C 53.7 ◦ C


Round II PCR MX1F2 MX1R2 2 min 30 s 10 s 70 ◦ C 70 ◦ C 4 ◦C
Round III PCR MX1MLUIF MX1SALIR 60 ◦ C 10 s 45 s 10 min hold
Screening PCR TRE2F MX1SALIR 55.7 ◦ C 10 s

Sequencing TRE2F TRE2R According to manufacturer’s instructions (Beckman Coulter)


K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8 3

Saint-Germain-en-Laye, France). Colonies were screened by PCR Reverse Transcriptase to correct for direct amplification from plas-
(Biotaq PCR kit, Bioline, London, UK) with the primers and condi- mid DNA potentially carried over during the RNA preparations.
tions given in Tables 1 and 2, respectively. Plasmids were purified Real-time PCR assays were performed on a LC480 96-well plate
from 3 ml of LB medium containing 100 ␮g/ml Ampicillin with the real time PCR machine (Roche, Burgess Hill, UK). The sequences
Miniprep kit (Qiagen, Crawley, UK) according to the manufacturer’s of the primers and TaqMan® probes to amplify the ELF or rbtMx1
instructions. The inserts were partially sequenced with primers genes are presented in Table 1. One microlitre of cDNA was added
given in Tables 1 and 2 and using the GenomeLabTM DTCS Quick to the following mix contained in individual wells of a 96-well opti-
Start Kit (Beckman Coulter, High Wycombe, UK). Sequencing reac- cal plate (Life Technologies, Paisley, UK): 10 ␮l of TaqMan® 2× PCR
tions were run on a QE8000 capillary sequencer (Beckman Coulter, mix with UNG (Life Technologies, Paisley, UK), 8 ␮l of dH2 O and 1 ␮l
High Wycombe, UK). of a 20× mix containing forward primer (18 ␮M), reverse primer
After sequence verification, a clone was used to inoculate 300 ml (18 ␮M) and probe (5 ␮M). The standard cycling conditions were
LB broth with 100 ␮g/ml Ampicillin. A solution of plasmid free of 50 ◦ C for 2 min, 95 ◦ C for 10 min followed by 50 cycles of 95 ◦ C for
endotoxins (pTRE2hyg-rbtMx1, pTet-Off and pTRE2hyg-Luc) was 15 s and 60 ◦ C for 1 min. The fluorescence output for each cycle
produced using the Endofree Maxiprep kit (Qiagen, Crawley, UK) was measured and recorded upon the completion of the entire run.
according to the manufacturer’s instructions. Absolute quantification of transcripts was carried out.
For each qPCR assay, a standard curve was generated using a
2.3. Isolation of CHSE-TOF-MX8 and CHSE-TOF-MX10 cell lines 10-fold serial dilution of plasmid (calibrator) containing the tar-
get sequence. A linear statistical regression between crossing point
CHSE-TOF cells were detached by Trypsin–EDTA (Life Technolo- (Cp) and log(concentration calibrator) was used to estimate the
gies, Paisley, UK) action and pelleted by centrifugation at 4200 × g expression level of the corresponding gene from its measured Cp
for 5 min, washed once with culture medium and twice with phos- value. The percentage efficiency was also calculated (Table 1). The
phate buffered saline (PBS, Life Technologies, Paisley, UK). The expression level of the gene relative to ELF was calculated by divid-
pellet was drained and re-suspended in solution R (Neon kit, ing the gene expression level by ELF expression level. To correct for
Life Technologies, Paisley, UK) at a density of 1.5 × 107 cells/ml. potential genomic DNA carry-over of a given sample, the expres-
pTRE2hyg-rbtMx1 plasmid was added to half of the cell suspension sion level of the “no RT” control was subtracted from the cDNA
at a concentration of 0.2 ␮g/␮l and the other half was left without expression level.
any DNA as a mock-transfected control. Two transfections were car-
ried out in a Neon® Transfection System (Life Technologies, Paisley, 2.5. Viral titration in CHSE-TOF-MX8, CHSE-TOF-MX10 and
UK) using a Neon® Transfection System 10 ␮L Kit (Life Technolo- CHSE-TOF
gies, Paisley, UK) set to 2 pulses for 20 ms at 1300 V, according to
prior optimisation trials for CHSE (data not shown). The two sets CHSE-TOF, CHSE-TOF-MX8 or CHSE-TOF-MX10 cells were
of transfected cells were added to 5 ml of medium supplemented seeded on 96-well plates (Nunc, Roskilde, Denmark) at a density of
with 16 ␮g/ml hygromycin (Sigma, Irvine, UK) and 1 ␮g/ml DOX 20,000 cells/cm2 (approx. 50% confluency) in culture medium with
(Sigma, Irvine, UK) in a 20 cm diameter Petri dish (Nunc, Roskilde, or without DOX supplemented with 25 mM HEPES buffer (Life Tech-
Denmark). nologies, Paisley, UK) and incubated at 15 ◦ C for 24 h and during the
following infection. Six to 8 wells were left empty. Tenfold dilu-
2.4. Quantitative real-time PCR (qPCR) characterisation of tions of virus inoculums (approx. 106 –107 TCID50 /ml) were added
transgenic cell lines in six replicate wells from dilution 10 to 1012 . IPNV isolate A2 or A5,
Salmon Alphavirus (SAV) isolates F93-125 or 4640, IHNV and Epi-
The transcription activity of the transgene was measured by zootic Haematopoietic Necrosis Virus (EHNV, Langdon et al., 1988)
qPCR. Each CHSE-TOF-MX clone was seeded and cultured with or were used (Table 3). Sixteen to 18 wells were left un-infected.
without DOX in a 6-well plate for 72 h. The total RNA was purified The cells were checked regularly for onset of cytopathic effect
using the RNA/DNA/Protein purification kit (Qiagen, Crawley, UK). (CPE). After 14 days, the cells were washed with PBS, fixed for
The Mx expression levels relative to elongation factor 1 ␣ gene (ELF) 10 min with 10% saline formalin, washed with PBS and incubated
were measured by qPCR in CHSE-TOF-MX clones. for 30 min with 0.1% crystal violet (Sigma, Irvine, UK), washed with
RNA was reverse transcribed to cDNA using the TaqMan® distilled water, drained and dried at room temperature. After the
Reverse Transcription Reagent kit (Life Technologies, Paisley, plates were photographed under a light box, the crystal violet dye
UK) with oligo-d(T)16 as follows: 9.625 ␮l of total RNA (approx. was redissolved in 100 ␮l 1% SDS solution (Sigma, Irvine, UK) and
0.5 ␮g) and 1.25 ␮l 50 ␮M oligo-d(T)16 were mixed and heated the optical density (OD) was read on a 96 well plate at 590 nm with
to 70 ◦ C for 10 min and chilled on ice. The final volume was a PowerWave X spectrophotometer (BioTek, Potton, UK).
adjusted to 25 ␮l by adding Master mix comprising the fol-
lowing: 1× RT buffer (25 mM Tris–HCl pH 8.3, 37.5 mM KCl, 2.6. Statistical analysis
5.5 mM MgCl2 ), 0.5 mM each dNTP, 0.4 U RNase inhibitor and
1.25 U Multiscribe Reverse Transcriptase. Reactions were incu- Analysis was intended to characterise the response of selected
bated at 48 ◦ C for 90 min, heat inactivated at 95 ◦ C for 5 min combinations of cell lines and medium (referred to as treatment
and stored at −80 ◦ C until use. For each sample, a mock reac- combinations) to different dilutions (referred to as doses) of a viral
tion “no RT” control was made by omitting the Multiscribe strain. The response of each treatment combination to the log10

Table 3
Taxonomy information on the viruses used in this study.

Full name Family Genus genome

Infectious Pancreatic Necrosis Virus Birnaviridae Aquabirnavirus dsRNA


Salmon Alpha Virus Togaviridae alphavirus ssRNA Pos
Infectious Haematopoeitic Necrosis Virus Rabdoviridae Novirabdovirus ssRNA Neg
Epizootic Haematopoietic Necrosis Virus Iridoviridae Ranavirus dsDNA
4 K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8

dose of the viral strain was measured as an optical density. Further CHSE-TOF with or without DOX. This can be seen visually (Fig. 2)
statistical analysis was conducted using version 2.12.0 of the R and was confirmed by statistical analysis (Table 4).
statistical computing environment (R Development Core Team, CHSE-TOF-MX10 and CHSE-TOF showed the same level of sen-
2010). Mean optical densities and 95% confidence intervals for sitivity against IPNV isolates A2 and A5 and against SAV isolate
defined treatment combinations at each dose were calculated and F93-125 irrespective of presence or absence of DOX in the cul-
plotted. An un-weighted four-parameter logistic dose–response ture medium (Fig. 2). In contrast, CHSE-TOF-MX10 was completely
curve was then fitted to the individual optical densities for the resistant to the SAV isolate 4640 irrespective of presence or absence
selected treatment combinations using the supplementary R of DOX in the culture medium (Fig. 2).
package drc 2.0-1 (Ritz and Streibig, 2005). The statistical fit was There was a clear effect of the DOX removal on the resistance
assessed by comparing the residual sums of squares obtained from of CHSE-TOF-MX8 cells to IPNV isolates A2 and A5 (Fig. 3 – lines
the model to the residual sums of squares from a one way analysis 1 and 2). The cell lines exhibited complete resistance to IPNV iso-
of variance. Two parameters are of particular interest. The first is lates A2 and A5 in the absence of DOX whereas in presence of DOX,
the TCID50 , a measure of the sensitivity of a treatment combination the sensitivity was similar to the parental line CHSE-TOF. This was
to a viral strain. The second is the lower asymptote, a measure of confirmed by statistical analysis (p < 0.001; Table 4; Fig. 4A and B).
the maximum sensitivity of the treatment combination to the viral However, CHSE-TOF-MX8 cells were completely resistant to SAV
strain. The predicted values were plotted using a monotone Her- isolates F93-125 and 4640 even in the presence of DOX (Table 4;
mite spline. Differences in response between cell lines cultivated Fig. 3 – lines 3 and 4).
with or without DOX and virus strain combinations were evaluated Both parental CHSE-TOF and Mx-expressing CHSE-TOF-MX8 cell
by comparing the residual variance of the dose–response curve lines were sensitive to EHNV infection. A significant effect of DOX
for each cell line and virus strain combination assuming that there on the resistance to EHNV infection could be observed in the CHSE-
was either no difference between responses between DOX or dif- TOF-MX8 cell line (p < 0.001; Table 4; Fig. 3 – line 5).
ferences between responses for DOX. The residual variance of the A partial resistance to IHNV was observed when comparing the
dose–response curve assuming no difference in responses to DOX parental CHSE-TOF with and without DOX (Fig. 3 – line 6). This
will be greater than the residual variance of the dose–response was confirmed by quantification and statistical analysis (p < 0.05;
curves assuming a difference in responses to DOX when there is a Table 4; Fig. 4C and D).
difference in response to DOX. The difference in response between
presence and absence of DOX was regarded as statistically signifi-
cant when the probability of the variance ratio occurring by chance 4. Discussion
alone was ≤5%. This approach evaluates differences between
curves rather than comparing the specific parameters. This is the first report on the production of a tetracycline
inducible expression system for salmon through the production of
3. Results a double recombinant fish cell line. The principle of tetracycline
dependent expression systems was investigated in 2005 in carp
3.1. Isolation of CHSE-TOF-MX8 and CHSE-TOF-MX10 cells (Munoz et al., 2005) but has never been applied since. Single
recombinant cell lines producing a reporter gene such as luciferase
After transfection with pTRE2hyg-rbtMx1, five clones were have been established in the past as a tool to study specific gene
characterised by qPCR. Among those, the clone CHSE-TOF-MX8 regulatory sequences, monitor toxicity of samples to fish cells or
showed the highest level of Mx induction with a fold increase upon study signalling mechanisms (see for review Martin et al., 2008).
DOX removal of 27.95 (Fig. 1). No induction of IFN gene could be In this study, the biological function of rbtMx1 was investigated in
detected by qPCR in any of the cell lines established in the present the Tet-Off Chinook salmon cell line CHSE-TOF recently established
report (CHSE-TOF, CHSE-TOF-MX clones, data not shown). (Collet and Lester, 2011b).
In fish, the Mx gene has been shown to be induced by many
3.2. Resistance tests to IPNV, SAV, IHNV and EHNV viruses (McBeath et al., 2007; Fernández-Trujillo et al., 2011) and
related to early viral protection (McLauchlan et al., 2003). However,
There was no significant difference in the titres of IPNV isolates the significance of this induction is difficult to evaluate because
A2 or A5, SAV isolates 4640 or SAV F93-125 in the parental cell line virus infection induces the production and release of type I and/or
II IFN which in turn induces not only the Mx gene expressing the
Mx protein but also many other molecules with direct or indirect
35
0.78 antiviral properties, most of which have never been fully charac-
1.63
33 terised in mammalian or other vertebrates (Boo and Yang, 2010).
Cp DOX
31 Cp NO DOX 5.75 Therefore, the exact contribution of Mx protein to the overall viral
29
protection in fish cells is not known.
It was observed that the Mx protein was less able to confer resis-
Average Cp

8.70
27
tance to IHNV than to other viruses such as SAV or IPNV. This is in
1.07
25 27.95 agreement with previous work where it was reported that Mx was
23 unable to inhibit IHNV nucleoprotein (N) production in CHSE cells
21
(Trobridge et al., 1997) but its effect on viral replication is unknown.
The accumulation of Mx protein conferred complete resistance
19
to IPNV infection which is in agreement with previous data (Larsen
17 et al., 2004), where it was associated with an inhibition of viral
15 protein production. In Larsen’s study a stable cell line of CHSE
TOF TOF-MX4 TOF-MX7 TOF-MX8 TOF-MX9 TOF-MX10
over-expressing Mx was generated and demonstrated that Mx
CHSE-TOF-MX clones
conferred a higher degree of protection to IPNV when compared to
the parental cell line and control cell line. However, the expression
Fig. 1. Level of expression of Mx in CHSE-TOF and clones CHSE-TOF-MX4, 7, 8, 9 and
10. The average fold increase relative to the control cultivated in absence of DOX is of Mx protein declined with increasing passage number making
indicated above the bars. Data represent average Cp values (N = 3) ± SE. it difficult to compare the level of resistance against a panel of
K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8 5

Fig. 2. Resistance level of CHSE-TOF and CHSE-TOF-MX10 cultivated with or without DOX. The cells were plated in a 96-well plate with or without DOX. Twenty four hours
later, they were infected with serial 10-fold dilutions of virus inoculum, incubated 14 days at 15 ◦ C, fixed, stained and photographed. The viruses tested were IPNV (isolates
A2 and A5) and SAV (isolates F93-125 and 4640).

different viruses and isolates. The authors hypothesised that there number currently reaching 35. As a matter of fact, a decrease in
was a detrimental effect of artificially high levels of Mx expression. the rate of cell division (data not shown) was observed upon DOX
The inducible system presented here would allow long term main- removal suggesting that the unregulated over expression of Mx
tenance of such a cell line with stable properties over high passage gene is indeed affecting the cell growth. In the presence of DOX,

Table 4
Relevant parameters of combination cell line–culture condition–virus.

Cell line Virus DOX TCID50 p Lower asymptote

Mean St dev Mean St dev

+ 4.95 0.14 −0.04 0.13


IPNV A2 NS
− 5.20 0.60 0.00 0.11
+ 6.02 0.07 0.01 9
IPNV A5 NS
− 5.56 0.25 0.02 0.09
+ Poor model fit Poor model fit
SAV 4640 No test
− 5.10 0.22 0.00 0.14
CHSE-TOF5
+ 6.52 0.12 0.03 0.08
SAV F93-125 NS
− 6.46 0.10 0.02 0.08
+ 6.49 0.14 −0.04 0.11
IHNV NS
− 6.03 0.2 −0.05 0.11
+ 6.09 0.05 0.06 0.03
EHNV NS
− 6.08 0.05 0.12 0.03

+ 2.90 0.13 0.29 0.20


IPNV A2 <0.001
− −0.18 38.67 3.02 2.48
+ 4.57 0.15 0.01 0.12
IPNV A5 <0.001
− −2.72 1216.2 2.83 6.34
+ 0.39 ND 1.14 ND
SAV 4640 NS
− 2.30 1.26 2.75 0.14
CHSE-TOF5-MX8
+ −0.79 8.99 0.20 26.47
SAV F93-125 NS
− 2.39 1.08 3.13 0.05
+ 5.50 0.13 −0.07 0.09
IHNV 0.015
− 4.87 0.21 −0.06 0.10
+ 4.91 0.08 −0.02 0.04
EHNV <0.001
− 5.02 0.06 −0.02 0.04

+ 3.49 0.42 2.33 0.11


SAV 4640 <0.001
− 3.68 0.21 1.59 0.09
CHSE-TOF5-MX10
+ Poor model fit No Poor model fit
SAV F93-125
− 5.25 0.16 test 0.06 0.13
6 K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8

Fig. 3. Resistance level of CHSE-TOF and CHSE-TOF-MX8 cultivated with or without DOX. The cells were plated in a 96-well plate with or without DOX. Twenty four hours
later, they were infected with serial 10-fold dilutions of virus inoculum, incubated 14 days at 15 ◦ C, fixed, stained and photographed. The viruses tested were IPNV (isolates
A2 and A5), SAV (isolates F93-125 and 4640), EHNV and IHNV.

in spite of a leakage in Mx expression, the cells showed a normal fish macrophage-like cell line (unpublished results). Cytopathic
rate of division very similar to the parental cell line. effect was observed for isolate F93-125 but not for isolate 4640. The
In a cell line constitutively expressing Mx1 but IFN-deficient, relatively high rbtMx1 sensitivity and high Mx gene inducing abil-
it was demonstrated that different isolates of the influenza virus ity of SAV isolate 4640 contrast with the low rbtMx1 sensitivity and
were affected differently by the over-expression of Mx1 (Dittmann the low Mx gene inducing ability of isolate F93-125. In addition,
et al., 2008). To date no fish cell lines deficient in IFN production viral gene nsP1 expression levels were much higher in cells infected
are available. In this study, the production effect of large amounts with isolate F93-125 rather than with isolate 4640 (unpublished
of rbtMx1 proteins induced a range of levels of protection against results). It is likely that the relative resistance of isolate F93-125
several fish virus types and isolates. This cellular system is therefore results in an excess of viral protein that could not be neutralised
a good model to compare the early protection conferred by rbtMx1 by rbtMx1 in CHSE-TOF-MX10. CHSE-TOF-MX8 cells produce a
proteins against different virus types and isolates. larger amount of rbtMx1 protein, even in presence of DOX, capable
A clear difference could be seen on the resistance level of neutralising viral proteins from the two isolates. A given virus
of CHSE-TOF-MX10 cells against SAV isolates 4640 and F93-125. isolate can be characterised in vitro by its ability to induce the Mx
CHSE-TOF-MX10 was found to express a relatively lower amount of gene and to resist to the rbtMx1 protein in CHSE-TOF-MX8/10.
rbtMx1 when compared to CHSE-TOF-MX8 which exhibited a com- Whether this correlates accurately with the virulence measured in
plete resistance to both isolates irrespective of the presence of DOX. vivo remains to be established but it can potentially constitute a
The parental CHSE-TOF cell line showed similar sensitivity levels to method of prediction that does not require the use of animals.
both isolates. These results demonstrated that isolate 4640 is more Unlike SAV, IPNV was able to propagate in cells expressing mod-
sensitive to the antiviral effect of rbtMx1 than isolate F93-125. SAV erate levels of rbtMx1. This was observed in CHSE-TOF-MX8 cells
isolate 4640 produced a fast and strong induction of the Mx gene cultured in the presence of DOX expressing a leakage level of Mx
whereas isolate F93-125 gave a delayed and lower response in a protein and showing sensitivity to IPNV but not to SAV (Fig. 2). This
K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8 7

A 3.0 B

3.0
2.5

2.5
TCID50 = 2.9±0.13
Lower asymptote = 0.29±0.2
optical density

optical density
2.0

2.0
1.5

1.5
1.0

1.0
TCID50 = -0.18±38.67
Lower asymptote = 3.02±2.48
0.5

0.5
0.0

0.0
1 2 3 4 5 1 2 3 4 5
Log10 dilution Log10 dilution

C D
3.0
3.0

2.5
2.5

TCID50 = 4.87±0.21 TCID50 = 5.5±0.13


Lower asymptote = -0.06±0.1 Lower asymptote = -0.07±0.09
2.0
2.0

optical density
optical density

1.5
1.5

1.0
1.0

0.5
0.5

0.0
0.0

1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8
Log10 dilution Log10 dilution

Fig. 4. Comparative resistance levels of CHSE-TOF-MX8 against IPNV isolate A2 with (A) or without (B) DOX, or against IHNV with (C) or without (D) DOX. Data are average
optical density (ROD) ± SE and the 4-parameter logistic model.

suggests that IPNV propagation is less affected by rbtMx1 protein response to this virus in fish. It is interesting to note that of all the
than SAV since the moderate leakage expression of Mx protein is viruses tested, EHNV is the only one showing complete resistance
sufficient to provide protection against SAV but not against IPNV. to rbtMx1 and the only one having a nuclear genome replication.
This can be explained by a lack of affinity of the Mx oligomers for Other fish iridoviruses such as the Orange-spotted Grouper virus
IPNV proteins or the ability of IPNV to counteract the viral protein have been shown to induce Mx protein (Chen et al., 2006).
sequestration by Mx through an active viral evasion mechanism These results give valuable information on the type of strategy
(Versteeg and Garcia-Sastre, 2010). Although this result agrees employed by different categories of viruses to escape the effect of
with past observations that IPNV is capable of inhibiting IFN action IFN. For example, IPNV which is highly sensitive to rbtMx1, has
(Collet et al., 2003), this study demonstrates a direct interaction developed strategies to limit the production of Mx by blocking steps
between IPNV and rbtMx1 protein whilst previous work empha- of the IFN signalling pathway (Collet et al., 2007). It is likely that SAV
sised the effect of IPNV on the regulation of Mx gene expression. has a similar strategy, as documented in other alphaviruses such as
A wider comparison showed that the presence of Mx protein in the Semliki Forest virus (SFV), where nsP2 has been described as a
the cell confers a range of resistance levels to different viruses. In strong inhibitor of IFN production (Breakwell et al., 2007).
this study SAV is the virus found to be most sensitive to the Mx pro- The use of optical densities to characterise the cellular response
tein, followed by IPNV and IHNV. The ability of EHNV to induce IFN to a viral isolate combined to a model-based analysis is a method-
has not been reported and very little is known about the immune ological innovation. It contributes to reduce the subjectivity of
8 K. Lester et al. / Journal of Virological Methods 182 (2012) 1–8

scoring by eye, facilitates the estimation of parameters character- Dittmann, J., Stertz, S., Grimm, D., Steel, J., Garcia-Sastre, A., Haller, O., Kochs, G., 2008.
istic of the response and allows identification of partially resistant Influenza A virus strains differ in sensitivity to the antiviral action of Mx-GTPase.
J. Virol. 82, 3624–3631.
cell responses. It is reasonable to suggest that this approach is Fernández-Trujillo, M.A., Novel, P., Manchado, M., Sepulcre, M.P., Mulero, V., Borrego,
promising but is not necessarily problem free. The reliance on a J.J., Alvarez, M.C., Béjar, J., 2011. Three Mx genes with differential response to
statistical modelling approach to generate results means that the VNNV infection have been identified in Gilthead seabream (Sparus aurata). Mol.
Immunol. 48 (9–10), 1216–1223.
results are only as good as the model. Whilst there is no evidence Fernández-Trujillo, M.A., García-Rosado, M.E., Borrego, J.J., Alvarez, M.C., Béjar, J.,
that the models presented in this report are not satisfactory two 2008. In vitro inhibition of sole Aquabirnavirus by recombinant Senegalese sole
possible improvements can be envisaged. One is a form of weight- Mx. Fish Shellfish Immunol. 24, 187–193.
Haller, O., 1981. Inborn resistance of ice to orthomyxoviruses. Curr. Top. Microbiol.
ing which takes into account the different amounts of variation
Immunol. 92, 25–52.
in optical densities for given doses. The second is the use of a Haller, O., Staeheli, P., Kochs, G., 2007a. Interferon-induced Mx proteins in antiviral
five-parameter logistic dose–response curve which would correct host defense. Biochimie 89, 812–818.
Haller, O., Stertz, S., Kochs, G., 2007b. The Mx GTPase family of interferon-induced
for any asymmetry in the relationship if present. Despite these
antiviral proteins. Microbes Infect. 9, 1636–1643.
shortcomings there is no evidence that these more sophisticated Haller, O., Frese, M., Kochs, G., 1998. Mx proteins: mediators of innate resistance to
methods would affect the conclusions of this report. RNA viruses. Rev. Sci. Tech. 17, 220–230.
The isolation of the novel cell lines CHSE-TOF-MX8/10 has pro- Haller, O., Arnheiter, H., Lindenmann, J., Gresser, I., 1980. Host gene influences sen-
sitivity to interferon action selectively for influenza virus. Nature 283, 660–662.
vided an opportunity to study the intimate relationship between Haller, O., Arnheiter, H., Gresser, I., Lindenmann, J., 1979. Genetically determined,
viral proteins and the rbtMx1 protein in fish. More specifically, it interferon-dependent resistance to influenza virus in mice. J. Exp. Med. 149,
allows understanding how diverse group of viruses have developed 601–612.
Horisberger, M.A., Haller, O., Arnheiter, H., 1980. Interferon-dependent genetic resis-
ways to successfully evade the effect of IFN (see for review Versteeg tance to influenza virus in mice: virus replication in macrophages is inhibited
and Garcia-Sastre, 2010). Further studies are being undertaken to at an early step. J. Gen. Virol. 50, 205–210.
test a wider range of SAV isolates and establish correlates between Kibenge, M.J.T., Munir, K., Kibenge, F.S.B., 2005. Constitutive expression of Atlantic
salmon Mx1 protein in CHSE-214 cells confers resistance to Infectious Salmon
in vivo virulence and rbtMx1 sensitivity using CHSE-TOF-MX8/10 Anaemia virus. Virol. J. 2, 75.
cell lines. In addition, in the absence of DOX, CHSE-TOF-MX8 cells Kochs, G., Haller, O., 1999a. Interferon-induced human MxA GTPase blocks nuclear
are completely resistant to IPNV or SAV making it the ideal material import of Thogoto virus nucleocapsids. Proc. Natl. Acad. Sci. U. S. A. 96,
2082–2086.
to test for risk of emergence of virulence in vitro.
Kochs, G., Haller, O., 1999b. GTP-bound human MxA protein interacts with the nucle-
ocapsids of thogoto virus (Orthomyxoviridae). J. Biol. Chem. 274, 4370–4376.
Langdon, J.S., Humphrey, J.D., Williams, L.M., 1988. Outbreaks of EHNV-like iri-
Acknowledgements dovirus in cultured rainbow trout, Salmo gairdneri Richardson, in Australia. J.
Fish Dis. 11, 93–96.
Larsen, R., Rokenes, T.P., Robertsen, B., 2004. Inhibition of infectious pancre-
S.K. Gahlawat is grateful to the Department of Biotechnology,
atic necrosis virus replication by Atlantic salmon Mx1 protein. J. Virol. 78,
Ministry of Science & Technology, and Government of India for 7938–7944.
providing a fellowship under Biotechnology Overseas Associate- Lee, S.H., Vidal, S.M., 2002. Functional diversity of Mx proteins: variations on a theme
of host resistance to infection. Genome Res. 12, 527–530.
ship and the authorities of CCS Haryana Agricultural University,
Lin, C.H., Christopher John, J.A., Lin, C.-H., Chang, C.Y., 2006. Inhibition of nervous
Hisar (India) for allowing uptake of the fellowship. Part of this work necrosis virus propagation by fish Mx proteins. Biochem. Biophys. Res. Commun.
was funded by EU FP6 IMAQUANIM project no. 7103. The authors 351, 534–539.
are grateful to the virology staff at Marine Scotland for providing Lindenmann, J., 1964. Inheritance of resistance to influenza virus in mice. Proc. Soc.
Exp. Biol. Med. 116, 506–509.
viruses. McBeath, A.J., Snow, M., Secombes, C.J., Ellis, A.E., Collet, B., 2007. Expression kinetics
of interferon and interferon-induced genes in Atlantic salmon (Salmo salar) fol-
lowing infection with infectious pancreatic necrosis virus and infectious salmon
References anaemia virus. Fish Shellfish Immunol. 22, 230–241.
McLauchlan, P.E., Collet, B., Ingerslev, E., Secombes, C.J., Lorenzen, N., Ellis, A.E.,
Altmann, S.M., Mellon, M.T., Distel, D.L., Kim, C.H., 2003. Molecular and functional 2003. DNA vaccination against viral haemorrhagic septicaemia (VHS) in rainbow
analysis of an interferon gene from the zebrafish, Danio rerio. J. Virol. 77, trout: size, dose, route of injection and duration of protection–early protection
1992–2002. correlates with Mx expression. Fish Shellfish Immunol. 15, 39–50.
Altmann, S.M., Mellon, M.T., Johnson, M.C., Paw, B.H., Trede, N.S., Zon, L.I., Kim, Martin, S.A.M., Collet, B., MacKenzie, S., Evensen, O., Secombes, C.J., 2008. Genomic
C.H., 2004. Cloning and characterization of an Mx gene and its corresponding tools for examining immune gene function in salmonid fish. Rev. Fish Sci. 16,
promoter from the zebrafish, Danio rerio. Dev. Comp. Immunol. 28, 295–306. 110–116.
Arnheiter, H., Skuntz, S., Noteborn, M., Chang, S., Meier, E., 1990. Transgenic mice Munoz, I., Carrillo, M., Zanuy, S., Gomez, A., 2005. Regulation of exogenous gene
with intracellular immunity to influenza virus. Cell 62, 51–61. expression in fish cells: an evaluation of different versions of the tetracycline-
Boo, K.H., Yang, J.S., 2010. Intrinsic cellular defenses against virus infection by antivi- regulated system. Gene 363, 173–182.
ral type I interferon. Yonsei Med. J. 51, 9–17. R Development Core Team, 2010. R: A Language and Environment for Statistical
Breakwell, L., Dosenovic, P., Karlsson Hedestam, G.B., D’Amato, M., Liljestrom, P., Computing. R Foundation for Statistical Computing, Vienna, Austria, ISBN 3-
Fazakerley, J., McInerney, G.M., 2007. Semliki Forest virus nonstructural pro- 900051-07-0, http://www.R-project.org.
tein 2 is involved in suppression of the type I interferon response. J. Virol. 81, Ritz, C., Streibig, J.C., 2005. Bioassay analysis using R. J. Stat. Software 12, 5.
8677–8684. Robertsen, B., Trobridge, G., Leong, J.A., 1997. Molecular cloning of double-stranded
Caipang, C.M., Hirono, I., Aoki, T., 2003. In vitro inhibition of fish rhabdoviruses by RNA inducible Mx genes from Atlantic salmon (Salmo salar L.). Dev. Comp.
Japanese flounder, Paralichthys olivaceus Mx. Virology 317, 373–382. Immunol. 21, 397–412.
Chen, Y.-M., Su, Y.-L., Lin, J.H.-Y., Yang, H.-L., Chen, T.-Y., 2006. Cloning of an orange- Staeheli, P., Haller, O., Boll, W., Lindenmann, J., Weissmann, C., 1986. Mx protein:
spotted grouper (Epinephelus coioides) Mx cDNA and characterisation of its constitutive expression in 3T3 cells transformed with cloned Mx cDNA confers
expression in response to nodavirus. Fish Shellfish Immunol. 20, 58–71. selective resistance to influenza virus. Cell 44, 147–158.
Chen, Y.-M., Su, Y.-L., Shie, P.-S., Lin, J.H.-Y., Yang, H.-L., Chen, T.-Y., 2008. Grouper Su, J., Yang, C., Zhu, Z., Wang, Y., Jang, S., Liao, L., 2009. Enhanced grass carp reovirus
Mx confers resistance to nodavirus and interacts with coat protein. Dev. Comp. resistance of Mx-transgenic rare minnow (Gobiocypris rarus). Fish Shellfish
Immunol. 32, 825–836. Immunol. 26, 828–835.
Collet, B., McDonald, C., Secombes, C.J., 2003. The promoter for the Interferon Reg- Trobridge, G.D., Leong, J.A., 1995. Characterization of a rainbow trout Mx gene. J.
ulatory Factor (IRF)-2 in the rainbow trout Oncorhynchus mykiss: cloning and Interferon Cytokine Res. 15, 691–702.
reporter gene activity. Fish Shellfish Immunol. 15, 473–477. Trobridge, G.D., Chiou, P.P., Leong, J.A., 1997. Cloning of the rainbow trout
Collet, B., Munro, E.S., Gahlawat, S., Acosta, F., Garcia, J., Roemelt, C., Zou, J., Sec- (Oncorhynchus mykiss) Mx2 and Mx3 cDNAs and characterization of trout Mx
ombes, C.J., Ellis, A.E., 2007. Infectious Pancreatic Necrosis Virus suppresses type protein expression in salmon cells. J. Virol. 71, 5304–5311.
I interferon signalling in rainbow trout gonad cell line but not in Atlantic salmon Versteeg, G.A., Garcia-Sastre, A., 2010. Viral tricks to grid-lock the type I interferon
macrophages. Fish Shellfish Immunol. 22, 44–56. system. Curr. Opin. Microbiol. 13, 508–516.
Collet, B., Lester, K., 2011a. Isolation of an inducible expression cell line in Atlantic Wu, Y.C., Lu, Y.-F., Chi, S.C., 2010. Anti-viral mechanism of barramundi Mx against
salmon Salmo salar. J. Biotech. 154, 209–211. betanodavirus involves the inhibition of viral RNA synthesis through the inter-
Collet, B., Lester, K., 2011b. Establishment of a Chinook salmon cell line with an ference of RdRp. Fish Shellfish Immunol. 28 (3), 467–475.
inducible gene expression system? In vitro Cell. Dev. Biol. – Anim. 47 (10), Wu, Y.C., Chi, S.C., 2007. Cloning and analysis of antiviral activity of a barramundi
695–697. (Lates calcarifer) Mx gene. Fish Shellfish Immunol. 23, 97–108.

Potrebbero piacerti anche