ArticlePDF Available

Recombinant Chicken Interferon-α Inhibits H9N2 Avian Influenza Virus Replication In Vivo by Oral Administration

Authors:

Abstract and Figures

Chicken interferon-alpha (ChIFN-α) has been demonstrated to be an important cytokine in antiviral immunity. However, the preventive or therapeutic effect of ChIFN-α as an oral antiviral agent on avian influenza virus (AIV) infection has not been fully clarified in chickens systemically. In the present study, we investigated the anti-H9N2 AIV effect of ChIFN-α on a cohort of 7- and 33-day-old specific pathogen-free (SPF) chickens by oral administration. Results showed that both the ChIFN-α preventive and therapeutic groups exhibited significantly reduced viral load in trachea when compared with the virus-challenged control group. The therapeutic effect was better than the preventive effect on 7-day-old SPF chickens, which is opposite to 33-day-old SPF chickens. We speculated that T-dependent lymphocyte system of 33-day-old SPF chickens might be easier to be stimulated by ChIFN-α than that of 7-day-old SPF chickens. In addition, there was no side effect on the body weight of chickens treated with ChIFN-α. We also found that IFN-stimulated genes (ISGs) (2',5'-oligoadenylate synthetase and Mx1) were upregulated in groups treated by ChIFN-α and/or virus, indicating that these 2 ISGs not only participated in anti-AIV response in vivo but also could be induced by oral administration of ChIFN-α. The present study suggested that ChIFN-α could be used as a potential preventive and therapeutic antiviral agent against H9N2 AIV infection by oral administration.
Content may be subject to copyright.
Recombinant Chicken Interferon-aInhibits H9N2 Avian
Influenza Virus Replication In Vivo by Oral Administration
Shanshan Meng,
1,2
Limin Yang,
1
Chongfeng Xu,
1,2
Zhuoming Qin,
3
Huaiying Xu,
3
Youling Wang,
3
Lei Sun,
1
and Wenjun Liu
1,4
Chicken interferon-alpha (ChIFN-a) has been demonstrated to be an important cytokine in antiviral immunity.
However, the preventive or therapeutic effect of ChIFN-aas an oral antiviral agent on avian influenza virus (AIV)
infection has not been fully clarified in chickens systemically. In the present study, we investigated the anti-H9N2
AIV effect of ChIFN-aon a cohort of 7- and 33-day-old specific pathogen-free (SPF) chickens by oral administration.
Results showed that both the ChIFN-apreventive and therapeutic groups exhibited significantly reduced viral load
in trachea when compared with the virus-challenged control group. The therapeutic effect was better than the
preventive effect on 7-day-old SPF chickens, which is opposite to 33-day-old SPF chickens. We speculated that T-
dependent lymphocyte system of 33-day-old SPF chickens might be easier to be stimulated by ChIFN-athan that of
7-day-old SPF chickens. In addition, there was no side effect on the body weight of chickens treated with ChIFN-a.
We also found that IFN-stimulated genes (ISGs) (20,50-oligoadenylate synthetase and Mx1) were upregulated in
groups treated by ChIFN-aand/or virus, indicating that these 2 ISGs not only participated in anti-AIV response
in vivo but also could be induced by oral administration of ChIFN-a. The present study suggested that ChIFN-acould
be used as a potential preventive and therapeutic antiviral agent against H9N2 AIV infection by oral administration.
Introduction
Chicken interferon-a(ChIFN-a) belongs to type I IFNs
and plays an essential role in the host antiviral response
through stimulating T-dependent lymphocyte system and
induction of numerous IFN-stimulated genes (ISGs) (Rose
1979; Sekellick and others 1994; Li and others 2005; Cheva-
liez and Pawlotsky 2009). There is evidence that ChIFN-a
administrated by oral ingestion or intravenous injection in-
hibits many epidemic avian viruses, such as infectious bron-
chitis virus, infectious bursal disease virus, and Newcastle
disease virus (Marcus and others 1999; Mo and others 2001;
Pei and others 2001). It has also been reported that intranasal
administration of human IFN-acan reduce the morbidity of
seasonal influenza A virus in ferrets (Kugel and others 2009).
More importantly, the H5N1 influenza viral replication at
early stage in mice can be controlled by the type I IFN
(Szretter and others 2009).
It has been elucidated that ChIFN-acould induce the
expression of numerous ISGs. Two of these genes, Mx1 and
20,50-oligoadenylate synthetase (20,50-OAS), were transcrip-
tionally increased at 3 days postinfection (dpi) in protecting
H9N2-infected 14-day-old specific pathogen-free (SPF)
chickens (Reemers and others 2009). However, whether the
expression levels of chicken Mx1 and 20,50-OAS in vivo can
be used as indicators of the antiviral effect of exogenous
ChIFN-aremains unknown.
The H9N2 subtype avian influenza virus (AIV), which was
first isolated from chickens (Chen and others 1994), has
gained considerable attention in China since it was rapidly
spread across China and even transmitted from animals to
humans (Chen and others 1994; Peiris and others 1999, 2001;
Guo and others 2000; Saito and others 2001; Liu and others
2003; Choi and others 2004; Xu and others 2004, 2007; Li and
others 2005). H9N2-infected chickens can serve as reservoir
host and transmit this kind of virus to mammals such as
pigs and humans (Webster and others 1992; Alexander 2000),
thus increasing the potential risk in controlling the viral
mutants. Moreover, H9N2-infected chickens are vulnerable
to secondary infection by pathogenic microbes, which may
consequently cause severe commercial loss. ChIFN-amay be
clinically used as an exogenous antiviral reagent to boost host
innate immunity responses for controlling low-pathogenicity
AIV infection (Marcus and others 2007). Although it has been
1
CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing,
People’s Republic of China.
2
Graduate University of Chinese Academy of Sciences, Beijing, People’s Republic of China.
3
Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan, People’s Republic of China.
4
China-Japan Joint Laboratory of Molecular Immunology and Molecular Microbiology, Institute of Microbiology, Chinese Academy of
Sciences, Beijing, People’s Republic of China.
JOURNAL OF INTERFERON & CYTOKINE RESEARCH
Volume 31, Number 7, 2011
ªMary Ann Liebert, Inc.
DOI: 10.1089/jir.2010.0123
533
identified that ChIFN-acould reduce the mortality of H9N2
(A/Beijing/1/96/H9N2) AIV in 9-day-old SPF chicken em-
bryos and 1-day-old SPF chickens (Xia and others 2004), the
clinical effect of ChIFN-aas a preventive and therapeutic or-
ally administrated antiviral agent against H9N2 AIV remains
to be demonstrated.
In the present study, several experiments were performed
to determine whether orally administrated ChIFN-ahad the
capability of protecting chickens from H9N2 AIV challenge
in SPF chickens. The expression level of chicken Mx1 and
20,50-OAS in tissues and body weights were also measured to
evaluate the antiviral effect of ChIFN-ain vivo.
Materials and Methods
Virus stocks
Avian influenza A virus, subtype H9N2, isolate A/
Chicken/GuangDong/05, was provided by the Institute of
Poultry Science, Shandong Academy of Agricultural Sci-
ences. The virus was reproduced in 10-day-old SPF chicken
embryonated eggs. The allantoic fluids were harvested,
stored at 708C, and used for testing the egg infectious dose
(EID
50
).
Preparation of ChIFN-a
The recombinant ChIFN-aprotein was prepared from
Escherichia coli. In brief, the ChIFN-agene was amplified by
polymerase chain reaction (PCR) from chicken liver (pBV/
chiIFNafor: 50-GAATTCATGTGCAACCACCTTCGCCCC
CA-30; pBV/chiIFNarev: 50-AGATCTTTAAGTGCGCGTGT
TGCC-30), cloned into the vector pBV220, and then induced
expression in E. coli strain BL21 (DE3) at 428C for 5 h. To
purify the ChIFN-aprotein, the cells were harvested and
treated using an ultrasonic cell disruptor. After that, the in-
clusion body was separated by centrifugation and washed
with PBST (pH 7.4; 137 mM NaCl, 2.7 mM KCl, 10 mM
Na
2
HPO
4
, 1.76 mM KH
2
PO
4
, and 1% triton), 2 M urea, and
1 M NaCl in turn. Then the inclusion body was dissolved in
8 M urea. The denatured protein was refolded in the re-
folding buffer [50 mM Tris (pH 8.0), 0.5 M Arg, 2 mM EDTA,
0.5 mM glutathione (oxidized), and 20% glycerol] at 48C for
48 h. After refolding, the ChIFN-awas added to cation ex-
change chromatography at the rate of 0.3 mL min
1
and then
eluted using PBS (pH 7.4) and 1 M NaCl at the rate of 1 mL
min
1
. The purified ChIFN-awere then detected by SDS-
PAGE analysis with Coomassie brilliant blue staining. The
antiviral titer of ChIFN-awas performed using vesicular
stomatitis virus/Madin Darby bovine kidney cells according
to previously described protocols (Chen and others 2009).
Chickens
Seven-day-old and 33-day-old white leghorn SPF chickens
were housed under SPF conditions according to the People’s
Republic of China National Standard ‘‘SPF chicken microbi-
ological quality control’’ (GB/T17998-1 999, China).
Testing the preventive and therapeutic
effects of ChIFN-aon chickens
Chickens were divided into 5 groups, namely the pre-
ventive group, therapeutic group, virus-challenged control
group, ChIFN-acontrol group, and blank control group.
Each chicken was orally fed with ChIFN-aby a 1-mL med-
ical syringe without needle. The preventive group (n¼20)
was successively orally fed with ChIFN-aonce a day for 4
days and subsequently infected with H9N2 AIV. The thera-
peutic group (n¼20) was first infected with H9N2 AIV and
then orally fed with ChIFN-aonce a day for 5 days. For
7-day-old SPF chickens, the preventive and therapeutic
groups were orally fed 0.510
4
U of ChIFN-afor each chick
per day. The dose of ChIFN-aused for 33-day-old SPF
chickens was 2-fold that of 7-day-old SPF chickens. About
110
6
EID
50
bird
1
H9N2 AIV was administered to 7-day-
old SPF chickens by intramuscular injection in the breast
muscle and 110
7
EID
50
bird
1
H9N2 AIV was administered
to 33-day-old SPF chickens by intravenous injection in the
wing vein. For comparison, day-matched virus-challenged
control group only infected with H9N2 AIV, ChIFN-acon-
trol group only treated with ChIFN-a, and blank control
group without any treatment were used.
Body weight was measured every 2 days. The body weight
gain was calculated as the mean body weight of each group
from 2 to 10 dpi minus the corresponding mean body
weight of groups at 4 dpi.
RNA isolation and real-time PCR
The transcriptional level of 2 ISGs (Mx1 and 20,50-OAS) was
analyzed and H9N2 AIV matrix protein gene (M1) mRNA
level was measured to determine the viral load in the tissues.
Table 1. Primers for Amplification of Interferon-Stimulated Genes and M1 of H9N2 Avian
Influenza Virus Used in Real-Time Polymerase Chain Reaction
Genes Primer Sequences of primers (50–30)
Size of
amplicon (bp)
Annealing
temperature (8C)
GenBank
accession no.
b-Actin
a
b-actin-RT-F GAGAAATTGTGCGTGACATCA 152 60 L08165
b-Actin-RT-R CCTGAACCTCTCATTGCCA
20,50-OAS
a
20,50-OAS-RT-F CACGGCCTCTTCTACGACA 103 62 AB037592
20,50-OAS-RT-R TGGGCCATACGGTGTAGACT
Mx1 Mx1-RT-F AGACCTTGCTTTTGGATGTGCT 105 62 AB088533.1
Mx1-RT-R TTGCTCAGGCGTTTATTTGCT
M1 M1-RT-F GGCTAAAGACAAGACCAATCCTG 87 60 AF536727.1
M1-RT-R GTCCTCGCTCACTGGGCAC
a
These primers were referred from Li and others (2007). Other primers were designed by Primer Express software v.1.5 (Applied
Biosystems).
20,50-OAS, 20,50-oligoadenylate synthetase; F, forward primer; R, reverse primer.
534 MENG ET AL.
For the RNA preparation, the upper part tissues of trachea of
four 7- and 33-day-old SPF chickens were collected at 4 and
3 dpi, respectively. Briefly, total RNA was extracted from the
trachea using RNAprep pure animal tissue kit (Tiangen).
Approximately 1 mg RNA was used to synthesize cDNA
using TIANScript RT Kit (Tiangen) according to the manu-
facturer’s directions. An aliquot (1/25) of the cDNA was used
as the template for a real-time PCR using SYBR
Premix Ex
Taqkit (Takara). The sequences of primers are listed in
Table 1 and b-actin was used as the internal control. All real-
time PCRs were performed in the Corbett Rotor-Gene 6000
(Corbett Research). The C
t
values of mRNA were determined
by Rotor-gene 6000 series software (Corbett Research) and the
relative gene expression was calculated using the 2
DDCt
method (Livak and Schmittgen 2001). The tracheal tissues of
7- and 33-day-old SPF chickens without any treatment were
collected at 4 dpi and used as calibrator of corresponding
animal experiment, respectively.
Statistical analysis
All data were analyzed using SPSS version 13.0 for Windows
software (SPSS, Inc., Chicago, IL). Comparison between dif-
ferent groups was performed using the Mann–Whitney Utest.
For all tests, a two-sided Pvalue of <0.05 was considered
significant.
Results
Preparation of recombinant ChIFN-a
In this study, the ChIFN-agene was cloned from chicken
liver and the deduced amino acid sequence without signal
sequence (32–193 aa) exhibited 100% identity with IFN-aof
Gallus (GenBank accession No. ABB05335). The recombinant
ChIFN-aprotein was expressed in E. coli strain BL21 (DE3)
and purified as previously described. The anti-vesicular
stomatitis virus activity of ChIFN-awas 110
7
Umg
1
, and
the ChIFN-awas used in the subsequent experiments.
The body weight of chickens was not influenced
by ChIFN-a
The body weight gain was measured every 2 days. Figure
1A shows that the body weight gain of all infected groups of
7-day-old SPF chickens gradually increased during the ex-
periment and there was no significant difference among them.
It showed that the body weight of 7-day-old SPF chickens was
not influenced by ChIFN-aand/or H9N2 AIV inoculation.
The body weight gain of the 33-day-old SPF chickens is shown
in Fig. 1B. We found that the body weight gain of the 3 in-
fected groups was significantly lower than that of the blank
control from 2 to 10 dpi and the preventive and therapeutic
groups recovered the body weight gain quicker when com-
pared with the virus-challenged control group. Moreover, the
body weight gain of the ChIFN control group was close to that
of the blank control group, which indicated that the body
weight of 33-day-old SPF chickens was influenced by H9N2
AIV and not by ChIFN-a.
ChIFN-asignificantly reduced the viral
load in trachea
The relative trachea viral load from each group was de-
termined using real-time PCR in both 7- and 33-day-old SPF
chickens. For 7-day-old SPF chickens, the M1 mRNA level of
the preventive and therapeutic groups was significantly in-
hibited by ChIFN-aat 3 dpi. The M1 mRNA level in trachea
was reduced by about 12- and 3-fold in the therapeutic and
preventive groups, respectively, compared with that of the
virus-challenged control group (Fig. 2A). We further ana-
lyzed the M1 mRNA level in trachea from 33-day-old SPF
chickens and found that the M1 mRNA level was about 10-
and 85-fold down in the therapeutic and preventive groups,
respectively, over the virus-challenged control group (Fig.
2B). These data indicated that ChIFN-agiven by oral ad-
ministration could significantly reduce the replication of
H9N2 AIV in trachea of 7- and 33-day-old SPF chickens.
FIG. 1. Body weight gain of 7-day-old (A) and 33-day-old
(B) specific pathogen-free (SPF) chickens. The body weight
gain was calculated as the mean body weight of each group
from 2 to 10 days postinfection (dpi) minus the corre-
sponding mean body weight of groups at 4 dpi. Data were
shown as means SEM. The Mann–Whitney Utest was used
to compare the differences of body weight gain between each
group and corresponding blank control group of the same
postinfection day. A Pvalue of <0.05 was considered sta-
tistically significant. *P<0.05.
CHIFN-aINHIBITS AIV REPLICATION BY ORAL ADMINISTRATION 535
The changes of the ISG expression levels
in chickens
We tried to identify whether Mx1 and 20,50-OAS could be
used as indexes to evaluate the antiviral effect of exogenous
ChIFN-ain chickens. The expression levels of these 2 ISGs
were also determined by real-time PCR. As showed in Table
2, both the virus-challenged control group and the ChIFN
control group could increase the expression levels of 20,50-
OAS and Mx1 when compared with the blank control group.
Moreover, these 2 ISG levels of the preventive and thera-
peutic groups were higher than those of the virus-challenged
control group and the ChIFN control group in both 7- and
33-day-old SPF chickens. As a result, 20,50-OAS and Mx1
could be used as reference indexes to evaluate the antiviral
effect of exogenous ChIFN-a.
Discussion
Low-pathogenicity AIV cause the outbreaks of febrile re-
spiratory infection ranging from mild illness such as weight
loss (<10%), ruffled feathers, and elevated temperatures to
severe illness (Belser and others 2007). Given the commercial
loss of AIV infection, finding a potential antiviral agent and
immunopotentiator is urgently needed. In the present study,
we demonstrated that oral administration of ChIFN-anot
FIG. 2. Viral load of 7-day-old (A) and 33-day-old (B) SPF chickens in trachea at 3 dpi, assayed by real-time polymerase
chain reaction. Data were shown as means SEM. The Mann–Whitney Utest was used to compare the differences of viral
load between each group and corresponding virus-challenged control group. A Pvalue of <0.05 was considered statistically
significant. *P<0.05.
Table 2. Induction of Interferon-Stimulated Genes of 7- and 33-Day-Old Specific
Pathogen-Free Chickens at 3Days Postinfection
Fold induction in 7-day-old SPF chickens
a
20,50-OAS Mx1
Preventive group 9.17 0.27 11.51 0.04
Therapeutic group 11.49 0.36 14.11 0.49
b
Virus-challenged control 7.85 1.00 4.35 1.75
ChIFN control 2.14 0.07 2.52 0.07
Blank control 0.92 0.14 0.91 0.14
Fold induction in 33-day-old SPF chickens
a
20,50-OAS Mx1
Preventive group 24.01 0.33 66.30 1.15
Therapeutic group 30.61 0.53 74.07 1.28
Virus-challenged control 15.99 0.91 64.15 2.22
ChIFN control 4.51 0.06 25.45 0.22
Blank control 0.70 0.01 0.71 0.01
b-Actin was used as the internal control.
The Mann–Whitney Utest was used to compare the differences of the relative expression level of each ISG between each group and
corresponding virus-challenged control group. A Pvalue of <0.05 was considered statistically significant.
a
Fold induction was calculated as the relative expression level of each individual gene compared with the level of normal control at 2 dpi
by 2
DDCt
and represented as mean SEM.
b
P<0.05.
SPF, specific pathogen-free; ChIFN, chicken interferon-alpha; dpi, days postinfection.
536 MENG ET AL.
only led to rapid recovery of the body weight gain in the
virus-challenged 33-day-old SPF chickens, but also showed
antiviral ability in both 7- and 33-day-old chickens.
Our study indicated that ChIFN-acould be used as a pre-
ventive and therapeutic agent against AIV. Further, we found
that the therapeutic effect was better than the preventive
effect on 7-day-old SPF chickens, which is opposite to 33-day-
old SPF chickens. These findings also indicated that the
T-dependent lymphocyte system of 33-day-old SPF chickens
was more mature than that of 7-day-old SPF chickens (Rose
1979), and hence, major histocompatibility complex class I
(MHC I) molecules might be upregulated efficiently (Sam-
uel 2001; Chevaliez and Pawlotsky 2009). Further studies
such as NK cell cytotoxicity assays are needed to detect the
change of T-dependent lymphocyte system in chickens, which
was caused by exogenous ChIFN-a( Jarosinski and others
2001).
We used real-time PCR to quantify the viral load in trachea,
which was very sensitive for assessing the antiviral action of
ChIFN-a. Further, oral administration of ChIFN-awas used in
our studies. Although ChIFN-aadministrated by oral inges-
tion can inhibit infectious bursal disease virus and Newcastle
disease virus (Marcus and others 1999; Pei and others 2001),
there is still no report about the anti-H9N2 activity of ChIFN-a
by oral administration until now. In this study, the viral load
of trachea in both 7- and 33-day-old chickens proved that oral
administration of ChIFN-awas an efficient and convenient
way against H9N2 infection. As ChIFN-aused in our study
was prepared from E. coli, a low-cost treatment of H9N2 in-
fection was also provided at the same time.
All type I IFNs regulate the transcription of ISGs by stim-
ulating classical JAK-STAT (signal transducer and activator of
transcription) signaling pathways (Li and others 2005).
However, very little information is available about the sup-
pressive effect of ISGs in vivo when ChIFN-ais administrated
by oral route. In the present study, 20,50-OAS and Mx1 could
be stimulated by oral ChIFN-aalone, which fit the previous
study that 20,50-OAS and Mx1 were 2 key ISGs involved in
IFN-induced pathways (Samuel 2001). The present findings
also suggested that 20,50-OAS and Mx1 actively participate in
regulating anti-AIV response, which was further reinforced
by the observation of upregulated expression of Mx1 and
20,50-OAS in AIV challenge control. However, chickens trea-
ted with ChIFN-aafter H9N2 AIV infection in the present
study exhibited a stronger effect on increasing the expression
level of both 20,50-OAS and Mx1 when compared with the
virus-challenged control. Taken together, we thought that
20,50-OAS and Mx1, at least in part, were involved in the an-
tiviral process of ChIFN-a, although the Mx1 protein of
chicken has been proved to lack antiviral activity in former
studies (Bernasconi and others 1995; Daviet and others 2009;
Benfield and others 2010). Therefore, the expression level of
Mx1 and 20,50-OAS in this study could not fully explain the
antiviral mechanism of ChIFN-a. This meant that other anti-
viral ISGs such as protein kinase (PKR) and RNA adenosine
deaminase (ADAR1) should also be investigated in the anti-
AIV response in future studies (Herbert and others 1995; Ko
and others 2004; Li and others 2010).
In summary, we extended the preventive and therapeutic
effect of ChIFN-ato chickens of different ages and demon-
strated that ChIFN-aadministrated by oral route not only
led to rapid recovery of the body weight gain, but also
showed a potential to suppress virus replication. Thus, oral
administration of recombinant ChIFN-aprovides a new
option in the prevention and therapy of H9N2 AIV infection.
Acknowledgments
This study was funded by the Knowledge Innovation
Program of the Chinese Academy of Sciences (KSCX2-YW-
R-158, KSCX2-YW-N-054) and the Major Special Program
of the Ministry of Science and Technology of China
(2009ZX10004-109).
Author Disclosure Statement
No competing financial interests exist.
References
Alexander DJ. 2000. A review of avian influenza in different bird
species. Vet Microbiol 74:3–13.
Belser JA, Lu X, Maines TR, Smith C, Li Y, Donis RO, Katz JM,
Tumpey TM. 2007. Pathogenesis of avian influenza (H7) virus
infection in mice and ferrets: enhanced virulence of Eurasian
H7N7 viruses isolated from humans. J Virol 81:11139–11147.
Benfield CT, Lyall JW, Tiley LS. 2010. The cytoplasmic location
of chicken mx is not the determining factor for its lack of
antiviral activity. PLoS One 5:e12151.
Bernasconi D, Schultz U, Staeheli P. 1995. The interferon-
induced Mx protein of chickens lacks antiviral activity. J In-
terferon Cytokine Res 15:47–53.
Chen BL, Zhang ZJ, Chen WB. 1994. Isolation and preliminary
serological characterization of type A influenza viruses from
chickens. Chin J Vet Med 20:3–5.
Chen X, Xue Q, Zhu R, Fu X, Yang L, Sun L, Liu W. 2009.
Comparison of antiviral activities of porcine interferon type I
and type II. Sheng Wu Gong Cheng Xue Bao 25:806–812.
Chevaliez S, Pawlotsky JM. 2009. Interferons and their use in
persistent viral infections. Handb Exp Pharmacol 189:203–241.
Choi YK, Ozaki H, Webby RJ, Webster RG, Peiris JS, Poon L, Butt
C, Leung YH, Guan Y. 2004. Continuing evolution of H9N2
influenza viruses in Southeastern China. J Virol 78:8609–8614.
Daviet S, Van Borm S, Habyarimana A, Ahanda ML, Morin V,
Oudin A, Van Den Berg T, Zoorob R. 2009. Induction of Mx
and PKR failed to protect chickens from H5N1 infection. Viral
Immunol 22:467–472.
Guo YJ, Krauss S, Senne DA, Mo IP, Lo KS, Xiong XP, Norwood
M, Shortridge KF, Webster RG, Guan Y. 2000. Characteriza-
tion of the pathogenicity of members of the newly established
H9N2 influenza virus lineages in Asia. Virology 267:279–288.
Herbert A, Lowenhaupt K, Spitzner J, Rich A. 1995. Chicken
double-stranded RNA adenosine deaminase has apparent
specificity for Z-DNA. Proc Natl Acad Sci U S A 92:7550–7554.
Jarosinski KW, Jia W, Sekellick MJ, Marcus PI, Schat KA. 2001.
Cellular responses in chickens treated with IFN-alpha orally
or inoculated with recombinant Marek’s disease virus ex-
pressing IFN-alpha. J Interferon Cytokine Res 21:287–296.
Ko JH, Asano A, Kon Y, Watanabe T, Agui T. 2004. Character-
ization of the chicken PKR: polymorphism of the gene and
antiviral activity against vesicular stomatitis virus. Jpn J Vet
Res 51:123–133.
Kugel D, Kochs G, Obojes K, Roth J, Kobinger GP, Kobasa D,
Haller O, Staeheli P, von Messling V. 2009. Intranasal ad-
ministration of alpha interferon reduces seasonal influenza A
virus morbidity in ferrets. J Virol 83:3843–3851.
Li C, Yu K, Tian G, Yu D, Liu L, Jing B, Ping J, Chen H. 2005.
Evolution of H9N2 influenza viruses from domestic poultry in
Mainland China. Virology 340:70–83.
CHIFN-aINHIBITS AIV REPLICATION BY ORAL ADMINISTRATION 537
Li YP, Handberg KJ, Juul-Madsen HR, Zhang MF, Jorgensen PH.
2007. Transcriptional profiles of chicken embryo cell cultures
following infection with infectious bursal disease virus. Arch
Virol 152:463–478.
Li Z, Wolff KC, Samuel CE. 2010. RNA adenosine deaminase
ADAR1 deficiency leads to increased activation of protein
kinase PKR and reduced vesicular stomatitis virus growth
following interferon treatment. Virology 396:316–322.
Liu H, Liu X, Cheng J, Peng D, Jia L, Huang Y. 2003. Phyloge-
netic analysis of the hemagglutinin genes of twenty-six avian
influenza viruses of subtype H9N2 isolated from chickens in
China during 1996–2001. Avian Dis 47:116–127.
Livak KJ, Schmittgen TD. 2001. Analysis of relative gene ex-
pression data using real-time quantitative PCR and the 2(-Delta
Delta C(T)) Method. Methods 25:402–408.
Marcus PI, Girshick T, van der Heide L, Sekellick MJ. 2007.
Super-sentinel chickens and detection of low-pathogenicity
influenza virus. Emerg Infect Dis 13:1608–1610.
Marcus PI, van der Heide L, Sekellick MJ. 1999. Interferon action
on avian viruses. I. Oral administration of chicken interferon-
alpha ameliorates Newcastle disease. J Interferon Cytokine
Res 19:881–885.
Mo CW, Cao YC, Lim BL. 2001. The in vivo and in vitro effects of
chicken interferon alpha on infectious bursal disease virus and
Newcastle disease virus infection. Avian Dis 45:389–399.
Pei J, Sekellick MJ, Marcus PI, Choi IS, Collisson EW. 2001.
Chicken interferon type I inhibits infectious bronchitis virus
replication and associated respiratory illness. J Interferon
Cytokine Res 21:1071–1077.
Peiris JS, Guan Y, Markwell D, Ghose P, Webster RG, Shortridge
KF. 2001. Cocirculation of avian H9N2 and contempo-
rary ‘‘human’ H3N2 influenza A viruses in pigs in south-
eastern China: potential for genetic reassortment? J Virol 75:
9679–9686.
Peiris M, Yuen KY, Leung CW, Chan KH, Ip PL, Lai RW, Orr
WK, Shortridge KF. 1999. Human infection with influenza
H9N2. Lancet 354:916–917.
Reemers SS, van Haarlem DA, Groot Koerkamp MJ, Vervelde L.
2009. Differential gene-expression and host-response profiles
against avian influenza virus within the chicken lung due to
anatomy and airflow. J Gen Virol 90:2134–2146.
Rose ME. 1979. The immune system in birds. J R Soc Med
72:701–705.
Saito T, Lim W, Suzuki T, Suzuki Y, Kida H, Nishimura SI,
Tashiro M. 2001. Characterization of a human H9N2 influenza
virus isolated in Hong Kong. Vaccine 20:125–133.
Samuel CE. 2001. Antiviral actions of interferons. Clin Microbiol
Rev 14:778–809.
Sekellick MJ, Ferrandino AF, Hopkins DA, Marcus PI. 1994.
Chicken interferon gene: cloning, expression, and analysis.
J Interferon Res 14:71–79.
Szretter KJ, Gangappa S, Belser JA, Zeng H, Chen H, Matsuoka
Y, Sambhara S, Swayne DE, Tumpey TM, Katz JM. 2009. Early
control of H5N1 influenza virus replication by the type I in-
terferon response in mice. J Virol 83:5825–5834.
Webster RG, Bean WJ, Gorman OT, Chambers TM, Kawaoka Y.
1992. Evolution and ecology of influenza A viruses. Microbiol
Rev 56:152–179.
Xia C, Liu J, Wu ZG, Lin CY, Wang M. 2004. The interferon-
alpha genes from three chicken lines and its effects on H9N2
influenza viruses. Anim Biotechnol 15:77–88.
Xu C, Fan W, Wei R, Zhao H. 2004. Isolation and identification
of swine influenza recombinant A/Swine/Shandong/1/2003
(H9N2) virus. Microbes Infect 6:919–925.
Xu KM, Li KS, Smith GJ, Li JW, Tai H, Zhang JX, Webster RG,
Peiris JS, Chen H, Guan Y. 2007. Evolution and molecular ep-
idemiology of H9N2 influenza A viruses from quail in southern
China, 2000 to 2005. J Virol 81:2635–2645.
Address correspondence to:
Dr. Lei Sun
CAS Key Laboratory of Pathogenic Microbiology
and Immunology
Institute of Microbiology
Chinese Academy of Sciences
No. 1 West Beichen Road
Chaoyang District
Beijing 100101
People’s Republic of China
E-mail: sunlei362@im.ac.cn
Dr. Wenjun Liu
CAS Key Laboratory of Pathogenic Microbiology
and Immunology
Institute of Microbiology
Chinese Academy of Sciences
No. 1 West Beichen Road
Chaoyang District
Beijing 100101
People’s Republic of China
E-mail: liuwj@im.ac.cn
Received 2 September 2010/Accepted 6 January 2011
538 MENG ET AL.
... A few studies demonstrated that chicken interferon-alpha showed an inhibitory effect on avian influenza viruses in vitro [119], in ovo [120], and in vivo [120][121][122]. ...
Article
Full-text available
This review article describes the current knowledge about the use of antiviral chemotherapeutics in avian species, such as farm poultry and companion birds. Specific therapeutics are described in alphabetical order including classic antiviral drugs, such as acyclovir, abacavir, adefovir, amantadine, didanosine, entecavir, ganciclovir, interferon, lamivudine, penciclovir, famciclovir, oseltamivir, ribavirin, and zidovudine, repurposed drugs, such as ivermectin and nitazoxanide, which were originally used as antiparasitic drugs, and some others substances showing antiviral activity, such as ampligen, azo derivates, docosanol, fluoroarabinosylpyrimidine nucleosides, and novel peptides. Most of them have only been used for research purposes and are not widely used in clinical practice because of a lack of essential pharmacokinetic and safety data. Suggested future research directions are also highlighted.
... Type I IFNs may work by promoting the transcription of IFN-stimulating genes (ISGs), leading to the production of antiviral proteins such as myxovirus resistance protein (Mx) and 2'-5 ' oligoadenylate synthetases (2',5'-OAS) (Katze et al., 2002). Chicken type I IFN (ChIFN) has been studied and proven to inhibit the replication of Newcastle disease virus (NDV), infectious bronchitis virus (IBV), Marek's disease virus (MDV), Rous sarcoma virus (RSV) and the avian influenza A/H9N2 virus in poultry (Davison & Nair, 2005;Jarosinski et al., 2001;Meng et al., 2011;Mo et al., 2001). Large-scale production of ChIFN alpha (ChIFNα) may thereby have enormous potential and economic impact on the poultry industry. ...
Article
The poultry industry in Vietnam is vulnerable to many viral diseases. Since antibiotics and vaccination provide inadequate protection, it will be beneficial to have alternative immunostimulants to boost non-specific immunity in poultry. Chicken interferon-alpha (ChIFNα) has been described previously with antiviral activity against many pathogens. Therefore, the purpose of this study is to clone and express recombinant ChIFNα in Escherichia coli. The ChIFNα gene was successfully cloned into the pET32a(+) vector, then expressed in the E. coli Rosetta strain. Different expression conditions were tested for the best yield of the expressed protein. Results showed that recombinant ChIFNα was expressed in Rosetta E. coli as inclusion bodies with a yield of 30 mg/100 L culture after induction with 0.5 mM IPTG in 4 hours at 37 oC. The recombinant protein was purified using affinity column chromatography under denaturing conditions with the purity > 94%. Western blot analysis indicated that recombinant ChIFNα reacted specifically with its antibody. Further studies will be carried out to characterize the biological activities of recombinant ChIFNα and its application in the poultry industry.
... Globally, H9N2 AI viruses have become widespread and represent a genuine threat in poultry over the last 2 decades [20]. Given the commercial loss of H9N2 AI virus, finding a potential antiviral agent is urgently needed [21]. ...
Article
Avian influenza (AI) has become a disease of great importance for human and animal health. Beside adverse side effects, there is resistance mutation for about all the conventional drugs that target viral proteins. This study aimed to evaluate antiviral activity of silver nanoparticles combined with epigallocatechingallate (EGCG-AgNPs) and co-administered with zinc sulphate (Zn+2) as alternative treatment strategy to control AI H9N2. EGCG conjugated silver nanoparticles (EGCG-AgNPs) were synthesized. Virus propagation was performed using embryonated Specific-Pathogen-Free (SPF) hen's eggs. Viral EID50 titers were determined before and after treatments. The antiviral activity was determined as Log virucidal reduction. A commercial tetrazolium MTS assay kit was used to determine cytotoxicity. Results showed that 50 μM EGCG was the most significant concentration reduced the logEID50/mL of AI H9N2. Co-treatment with zinc sulphate (1.3 mg/mL) increased the EGCG antiviral effect. The most effective antiviral activity was obtained when combined EGCG-AgNPs with zinc sulphate with the greatest virucidal log reduction. No cytotoxic effect in Vero cells was observed among all of these forms at concentrations of interest used in this study. In conclusion, the topical application of EGCG-AgNPs/ZnSO4 demands additional antiviral strategies against H9N2 AI. This combination may prevent virus transmission, inhibit virus replication within neighboring cells and inhibit microbial resistance by making microbial adaptability very difficult.
... More recently, Meng et al., in a study on SPF chickens experimentally infected with H9N2 avian influenza virus (AIV), demonstrated that oral administration of ChIFN-α not only ensured the rapid recovery of body weight gain in infected chickens, but also exerted an antiviral effect. This study therefore suggested that ChIFN-α could be used as both a preventive and a therapeutic agent against AIV [105]. This study showed that ChIFN-α treated chickens, following infection with AIV H9N2, increased their expression of both 2′,5′-oligoadenylate synthetase (2′,5′-OAS) and myxovirus resistance proteins 1 (Mx1) compared to the control ones. ...
Article
Full-text available
Cytokines are important molecules that orchestrate the immune response. Given their role, cytokines have been explored as drugs in immunotherapy in the fight against different pathological conditions such as bacterial and viral infections, autoimmune diseases, transplantation and cancer. One of the problems related to their administration consists in the definition of the correct dose to avoid severe side effects. In the 70s and 80s different studies demonstrated the efficacy of cytokines in veterinary medicine, but soon the investigations were abandoned in favor of more profitable drugs such as antibiotics. Recently, the World Health Organization has deeply discouraged the use of antibiotics in order to reduce the spread of multi-drug resistant microorganisms. In this respect, the use of cytokines to prevent or ameliorate infectious diseases has been highlighted, and several studies show the potential of their use in therapy and prophylaxis also in the veterinary field. In this review we aim to review the principles of cytokine treatments, mainly IFNs, and to update the experiences encountered in animals.
Article
African swine fever (ASF) is a highly contagious and deadly disease that affects domestic and wild pigs. No commercial vaccine or antiviral is currently available against ASF. The control of ASF primarily relies on implementing effective biosecurity measures during the breeding process. Here, we evaluated the preventive and therapeutic potential of the interferon (IFN) cocktail (a mixture of recombinant porcine IFN α and γ) on ASF. The IFN cocktail treatment delayed the onset of ASF symptoms and ASF virus (ASFV) replication for approximately one week. However, IFN cocktail treatment could not prevent the death of the pigs. Further analysis showed that IFN cocktail treatment increased the expression of multiple IFN-stimulated genes (ISGs) in porcine peripheral blood mononuclear cells in vivo and in vitro. Additionally, IFN cocktail modulated the expression of pro- and anti-inflammatory cytokines and reduced tissue injury in the ASFV-infected pigs. Collectively, the results suggest that the IFN cocktail restricts the progression of acute ASF by inducing high levels of ISGs, contributing to the pre-establishment of antiviral status, and modulating the balance of pro- and anti-inflammatory mediators to lessen cytokine storm-mediated tissue damage.
Article
Hydropericardium-hepatitis syndrome (HHS) caused by fowl adenovirus (FAdV) serotype 4 strains is a highly contagious disease that causes significant economic loss to the global poultry industry. However, subunit vaccine against FAdV-4 infection is not yet commercially available to date. This study aims to explore the potential for oral immunization of recombinant Saccharomyces cerevisiae expressing Fiber-2 of FAdV-4 as a subunit vaccine. Here, we constructed recombinant S. cerevisiae (ST1814G/Fiber-2) expressing recombinant Fiber-2 (rFiber-2), which was displayed on the cell surface. To evaluate the immune response and protective effect of live recombinant S. cerevisiae, chickens were orally immunized with the constructed live ST1814G/Fiber-2, three times at 5-day intervals, and then challenged with FAdV-4. The results showed that oral administration of live ST1814G/Fiber-2 could stimulate the production of humoral immunity, enhance the body's antiviral activity and immune regulation ability, improve the composition of gut microbiota, provide protection against FAdV-4 challenge, reduce viral load in the liver, and alleviate the pathological damage of heart, liver, and spleen for chicken. In addition, we found the synergistic effect in combining the ST1814G/Fiber-2 yeast and inactivated vaccine to trigger stronger humoral immunity and mucosal immunity. Our results suggest that oral live ST1814G/Fiber-2 is a potentially safer auxiliary preparation strategy in controlling FAdV-4 infection.
Article
Full-text available
Infectious bursal disease virus (IBDV) is one of the most important infectious diseases of poultry around the world. Gut-associated lymphoid tissues (GALT) are the first line of defense of the host against the infection. The purpose of this study was to investigate the role of innate immune antiviral signaling triggered by Toll-like receptor 3 (TLR3), as well as macrophage activation and cytokine response in the intestinal lamina propria (ILP) cells after the oral challenge of IBDV in relation to IBDV virulence and disease pathogenesis. The results showed that the expression levels of TLR3, IRF7, IFN-α/β and the corresponding downstream antiviral factors OAS, PKR and Mx were all upregulated in the SPF chicken ILP cells at 8 h post-infection (hpi) and 12 hpi. Similarly, macrophages were activated, with the initial macrophage M1 activation observed at 8 hpi, but then it rapidly shifted to a non-protective M2-type. Both Th1 (IFN-γ, TNF-α, IL-12) and Th2 (IL-4 and IL-10) types of cytokines were differentially upregulated during the early stage of infection; however, the Th1 cytokines exhibited stronger activation before 8 hpi compared to those of the Th2 cytokines. Interestingly, differential regulations of gene expression induced by different IBDV strains with different virulence were detected. The HLJ0504-like very virulent (vv) IBDV strain NN1172 induced stronger activation of TLR3-IFN-α/β pathway, macrophages and the Th1/2 cytokines' expression, compared to those induced by the attenuated strain B87 at 8 hpi and 12 hpi in the ILP cells. In conclusion, the innate antiviral response mediated by the TLR3-IRF7 pathway, macrophage activation and cytokine expression in the GALT cells at the early stage of IBDV infection was differentially modulated, and the HLJ0504-like vvIBDV strain triggered stronger activation than the attenuated vaccine strain, and that may play an important role in the progression of disease.
Article
Full-text available
Kanatlı hayvanlarda hastalıkların önlenmesi ve sağaltımı aşıların ve antibiyotiklerin kullanılması ile sağlanır. Antibiyotiklerin uzun yıllar boyunca kullanılması antibiyotiğe dirençli bakterilerin ortaya çıkması ile ilgili sorunları beraberinde getirmiştir. Hastalıkların önlenmesinde kullanılan aşılardaki adjuvantlar, sağlık üzerinde yan etkilere sahip olabilir ve immun yanıtı yetersiz bir şekilde uyarabilir. Bu nedenle kanatlı endüstrisinde yeni aşı stratejilerinin geliştirilmesi oldukça önemli bir konu haline gelmiştir. Sitokinler, yangı reaksiyonlarında hayati rol oynayan hücreler tarafından salgılanan immun sistem hücrelerinin aktivasyonu ve düzenlenmesini sağlayan peptitlerdir. Kanatlı immunolojisi ve genetik alanındaki gelişmeler, özellikle tavukta çeşitli sitokinlerin keşfedilmesine ve bu sitokinlerin işlevsel özelliklerinin ve mekanizmalarının anlaşılmasını sağlamıştır. Kanatlı hayvanlarda enfeksiyöz ajanlara karşı kullanılan aşılarda sitokinlerin potansiyel bir aşı adjuvantı olarak kullanılması yönünde birçok çalışma gerçekleştirilmiştir. Bu derlemede kanatlı sitokinlerinin çeşitleri, fonksiyonel özellikleri ve sitokinlerin aşı adjuvantı olarak kullanımı hakkında bilgi vermek amaçlandı.
Article
Full-text available
Chicken interferon-α (chIFN-α) is an important antiviral cytokine and represents one of the first lines of the chicken’s innate immune system. The current study is the first-ever report of chicken IFN (chIFN) production in Pakistan. In this study, we have used live and UV-irradiated Newcastle disease virus (NDV) to induce the expression of chIFN-α in chicken embryo fibroblast (CEF) cells. ChIFN-α was partially purified in a two-step protocol; ultracentrifugation followed by treatment with anti-chIFN-β antibodies. The purified chIFN-α was ana-lysed via sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and the in vitro antiviral potential of chIFN-α was determined against the H9N2 avian influenza virus (AIV) via a cytopathic inhibition assay. The relative mRNA level of the IFN-stimulated genes (ISGs) in the IFN-stimulated CEF cells was measured at various time intervals by a quantitative polymerase chain reaction (qPCR). The stability of natural chIFN-α to the temperature, pH, and ultraviolet (UV) light was also determined. The in vivo therapeutic potential of chIFN-α was determined in 7-day-old broiler chickens challenged with AIV. We found that a higher chIFN-α expression level was induced by the UV-irradiated NDV in the CEF cells as compared to the live NDV. The UV-irradiated NDV induced the maximum IFN production in the CEF cells at 24 h post-infection. Two bands of 21 kDa on SDS-PAGE confirmed the presence of the chIFN-α protein. The cytopathic inhibition assay indicated the strong antiviral activity of chIFN-α against AIV. Our results of the stability analysis showed that chIFN-α was stable at a wide range of temperatures and pH levels. However, a little exposure to UV-light resulted in a significant loss of antiviral activity. We also observed that the antiviral activity of chIFN-α is related to the expression levels of the antiviral ISGs. The results of the in vivo study showed that the chIFN-α therapy via the oral route resulted in a significant improvement in the tracheal pathology of chickens challenged with AIV. In conclusion, we suggest that chIFN-α could be an important therapeutic tool to control avian influenza infection in poultry.
Article
Full-text available
In this review we examine the hypothesis that aquatic birds are the primordial source of all influenza viruses in other species and study the ecological features that permit the perpetuation of influenza viruses in aquatic avian species. Phylogenetic analysis of the nucleotide sequence of influenza A virus RNA segments coding for the spike proteins (HA, NA, and M2) and the internal proteins (PB2, PB1, PA, NP, M, and NS) from a wide range of hosts, geographical regions, and influenza A virus subtypes support the following conclusions. (i) Two partly overlapping reservoirs of influenza A viruses exist in migrating waterfowl and shorebirds throughout the world. These species harbor influenza viruses of all the known HA and NA subtypes. (ii) Influenza viruses have evolved into a number of host-specific lineages that are exemplified by the NP gene and include equine Prague/56, recent equine strains, classical swine and human strains, H13 gull strains, and all other avian strains. Other genes show similar patterns, but with extensive evidence of genetic reassortment. Geographical as well as host-specific lineages are evident. (iii) All of the influenza A viruses of mammalian sources originated from the avian gene pool, and it is possible that influenza B viruses also arose from the same source. (iv) The different virus lineages are predominantly host specific, but there are periodic exchanges of influenza virus genes or whole viruses between species, giving rise to pandemics of disease in humans, lower animals, and birds. (v) The influenza viruses currently circulating in humans and pigs in North America originated by transmission of all genes from the avian reservoir prior to the 1918 Spanish influenza pandemic; some of the genes have subsequently been replaced by others from the influenza gene pool in birds. (vi) The influenza virus gene pool in aquatic birds of the world is probably perpetuated by low-level transmission within that species throughout the year. (vii) There is evidence that most new human pandemic strains and variants have originated in southern China. (viii) There is speculation that pigs may serve as the intermediate host in genetic exchange between influenza viruses in avian and humans, but experimental evidence is lacking. (ix) Once the ecological properties of influenza viruses are understood, it may be possible to interdict the introduction of new influenza viruses into humans.
Article
Full-text available
Chicken Mx belongs to the Mx family of interferon-induced dynamin-like GTPases, which in some species possess potent antiviral properties. Conflicting data exist for the antiviral capability of chicken Mx. Reports of anti-influenza activity of alleles encoding an Asn631 polymorphism have not been supported by subsequent studies. The normal cytoplasmic localisation of chicken Mx may influence its antiviral capacity. Here we report further studies to determine the antiviral potential of chicken Mx against Newcastle disease virus (NDV), an economically important cytoplasmic RNA virus of chickens, and Thogoto virus, an orthomyxovirus known to be exquisitely sensitive to the cytoplasmic MxA protein from humans. We also report the consequences of re-locating chicken Mx to the nucleus. Chicken Mx was tested in virus infection assays using NDV. Neither the Asn631 nor Ser631 Mx alleles (when transfected into 293T cells) showed inhibition of virus-directed gene expression when the cells were subsequently infected with NDV. Human MxA however did show significant inhibition of NDV-directed gene expression. Chicken Mx failed to inhibit a Thogoto virus (THOV) minireplicon system in which the cytoplasmic human MxA protein showed potent and specific inhibition. Relocalisation of chicken Mx to the nucleus was achieved by inserting the Simian Virus 40 large T antigen nuclear localisation sequence (SV40 NLS) at the N-terminus of chicken Mx. Nuclear re-localised chicken Mx did not inhibit influenza (A/PR/8/34) gene expression during virus infection in cell culture or influenza polymerase activity in A/PR/8/34 or A/Turkey/50-92/91 minireplicon systems. The chicken Mx protein (Asn631) lacks inhibitory effects against THOV and NDV, and is unable to suppress influenza replication when artificially re-localised to the cell nucleus. Thus, the natural cytoplasmic localisation of the chicken Mx protein does not account for its lack of antiviral activity.
Article
Full-text available
Sampling the complete organ instead of defined parts might affect analysis at both the cellular and transcriptional levels. We defined host responses to H9N2 avian influenza virus (AIV) in trachea and different parts of the lung. Chickens were spray-inoculated with either saline or H9N2 AIV. Trachea and lung were sampled at 1 and 3 days post-inoculation (p.i.) for immunocytochemistry, real-time quantitative RT-PCR and gene-expression profiling. The trachea was divided into upper and lower parts and the lung into four segments, according to anatomy and airflow. Two segments contained the primary and secondary bronchi, cranial versus caudal (parts L1 and L3), and two segments contained the tertiary bronchi, cranial versus caudal (parts L2 and L4). Between the upper and lower trachea in both control and infected birds, minor differences in gene expression and host responses were found. In the lung of control birds, differences in anatomy were reflected in gene expression, and in the lung of infected birds, virus deposition enhanced the differences in gene expression. Differential gene expression in trachea and lung suggested common responses to a wide range of agents and site-specific responses. In trachea, site-specific responses were related to heat shock and lysozyme activity. In lung L1, which contained most virus, site-specific responses were related to genes involved in innate responses, interleukin activity and endocytosis. Our study indicates that the anatomy of the chicken lung must be taken into account when investigating in vivo responses to respiratory virus infections.
Article
Full-text available
Widespread distribution of highly pathogenic avian H5N1 influenza viruses in domesticated and wild birds continues to pose a threat to public health, as interspecies transmission of virus has resulted in increasing numbers of human disease cases. Although the pathogenic mechanism(s) of H5N1 influenza viruses has not been fully elucidated, it has been suggested that the ability to evade host innate responses, such as the type I interferon response, may contribute to the virulence of these viruses in mammals. We investigated the role that type I interferons (alpha/beta interferon [IFN-alpha/beta]) might play in H5N1 pathogenicity in vivo, by comparing the kinetics and outcomes of H5N1 virus infection in IFN-alpha/beta receptor (IFN-alpha/betaR)-deficient and SvEv129 wild-type mice using two avian influenza A viruses isolated from humans, A/Hong Kong/483/97 (HK/483) and A/Hong Kong/486/97 (HK/486), which exhibit high and low lethality in mice, respectively. IFN-alpha/betaR-deficient mice experienced significantly more weight loss and more rapid time to death than did wild-type mice. HK/486 virus caused a systemic infection similar to that with HK/483 virus in IFN-alpha/betaR-deficient mice, suggesting a role for IFN-alpha/beta in controlling the systemic spread of this H5N1 virus. HK/483 virus replicated more efficiently than HK/486 virus both in vivo and in vitro. However, replication of both viruses was significantly reduced following pretreatment with IFN-alpha/beta. These results suggest a role for the IFN-alpha/beta response in the control of H5N1 virus replication both in vivo and in vitro, and as such it may provide some degree of protection to the host in the early stages of infection.
Article
The interferon-α genes from three chicken lines were cloned by a direct PCR technique, and the effects of recombinant protein expressed in a prokaryotic system on highly pathogenic H9N2 influenza viruses were investigated. The cloned ChIFN-α gene encoded a protein of 193 amino acids with a signal sequence of 31 amino acids and mature peptides of 162 amino acids. Comparison of ChIFN-α sequences, detected six amino acids substitutions at positions 50, 58, 65, 81, 181, and 183. Homology analysis indicated that ChIFN-α genes could be subdivided into two lineages, SH-ChIFN-α and WJ-ChIFN-α. In addition, both SH-ChIFN-α and WJ-ChIFN-α were expressed with the N-terminal 6 consecutive histidine residues in a high-level prokaryotic expression system. Recombinant chicken interferon-α (rChIFN-α) protein has anti-VSV activity of more than 1 × 10 U/mg. Moreover, High concentration (10,000 U) of rSH-ChIFN-α resulted in over 40% inhibition of the H9N2 virus infection in chicken embryos (Ovo), and 100% inhibition from one day-old to five day-old chickens (Vivo). The results suggested that rChIFN-α is a potential agent against many Chicken viral strains.
Article
The two most commonly used methods to analyze data from real-time, quantitative PCR experiments are absolute quantification and relative quantification. Absolute quantification determines the input copy number, usually by relating the PCR signal to a standard curve. Relative quantification relates the PCR signal of the target transcript in a treatment group to that of another sample such as an untreated control. The 2(-DeltaDeltaCr) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments. The purpose of this report is to present the derivation, assumptions, and applications of the 2(-DeltaDeltaCr) method. In addition, we present the derivation and applications of two variations of the 2(-DeltaDeltaCr) method that may be useful in the analysis of real-time, quantitative PCR data. (C) 2001 Elsevier science.
Article
We are currently facing a global threat caused by a highly pathogenic avian H5N1 influenza virus (hpH5N1). Death occurs in 48 h in infected chickens, suggesting that they fail to eliminate the virus. Little is known about the immune response in chickens after hpH5N1 infection, or how the virus is evolving to modify and evade host protective responses. Therefore, to better understand the chicken immune response following hpH5N1 infection, we set up an experimental infection of chickens with an hpH5N1 strain, and quantified the mRNA expression of several cytokines and antiviral proteins at different time points post-infection. We show here that a weak host immune response is observed in vivo, in spite of the induction of IL-6, myxovirus resistance protein (Mx), and protein kinase R (PKR). This weak immune response, probably due in part to the absence of type I interferon, was not sufficient to counteract the hpH5N1 virus and protect the chicken from death.
Article
Two size forms of ADAR1 adenosine deaminase are known, one constitutively expressed (p110) and the other interferon (IFN)-induced (p150). To test the role of ADAR1 in viral infection, HeLa cells with ADAR1 stably knocked down and 293 cells overexpressing ADAR1 were utilized. Overexpression of p150 ADAR1 had no significant effect on the yield of vesicular stomatitis virus. Likewise, reduction of p110 and p150 ADAR1 proteins to less than approximately 10 to 15% of parental levels (ADAR1-deficient) had no significant effect on VSV growth in the absence of IFN treatment. However, inhibition of virus growth following IFN treatment was approximately 1 log(10) further reduced compared to ADAR1-sufficient cells. The level of phosphorylated protein kinase PKR was increased in ADAR1-deficient cells compared to ADAR1-sufficient cells following IFN treatment, regardless of viral infection. These results suggest that ADAR1 suppresses activation of PKR and inhibition of VSV growth in response to IFN treatment.
Article
Interferons (IFNs) are natural proteins produced by wide variety of cells in response to viral infection or other biological inducers, and they execute diversified functions as antiviral defense, immune activation and cell growth regulation. Four genes encoding porcine interferons (PoIFN), PoIFN-alpha, PoIFN-gamma, PoIFN-alphagamma or PolFN-omega, were cloned and sequenced. The four types of porcine interferon genes were subcloned into the pET-His vector, and expressed in Escherichia coli Rosetta (DE3). The recombinant products were purified and renaturalized from inclusion bodies to obtain a native state of well biological activity. Antiviral activity assays for porcine interferons were performed and evaluated by standard procedures in following cell/virus test systems: Marc-145/PRRSV, Marc-145/VSV, PK-15/VSV, Vero/VSV or MDBK/VSV. The data showed that both PoIFN-alpha and PoIFN-alpagamma demonstrated significant antiviral activities, and the titer of them against PRRSV was up to 10(8) U/mg. PoIFN-gamma had approximately half or one-thirds antiviral activity of PoIFN-alpha. PoIFN-omega showed inconspicuous antiviral activity.