Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Developmental and Comparative Immunology 74 (2017) 178e189

Contents lists available at ScienceDirect

Developmental and Comparative Immunology


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

Oral DNA vaccines based on CS-TPP nanoparticles and alginate


microparticles confer high protection against infectious pancreatic
necrosis virus (IPNV) infection in trout
Sohrab Ahmadivand a, Mehdi Soltani a, b, *, Mahdi Behdani c, Øystein Evensen d,
Ehsan Alirahimi c, Reza Hassanzadeh e, Ellahe Soltani f
a
Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, P.O. Box 14155-6453, Tehran, Iran
b
Centre of Excellence of Aquatic Animal Health, University of Tehran, Tehran, Iran
c
Biotechnology Research Center, Venom & Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
d
Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
e
Central Veterinary Laboratory, Iran Veterinary Organization, Tehran, Iran
f
Department of Microbiology, Faculty of Sciences, University of Tehran, Tehran, Iran

a r t i c l e i n f o a b s t r a c t

Article history: Infectious pancreatic necrosis virus (IPNV) is the etiological agent of a contagious viral disease causing
Received 19 March 2017 remarkable mortalities in different fish species. Despite the availability of commercial vaccines against
Received in revised form IPN, the disease still constitutes one of the main threats to the aquaculture industry worldwide. In this
3 May 2017
study, we developed a DNA vaccine encoding the VP2 gene of IPNV and evaluated its ability to induce
Accepted 3 May 2017
Available online 4 May 2017
protective immunity in rainbow trout fry (3 g) at doses of 10 and 25 mg/fish and boosting with the same
doses two weeks later through the oral route using chitosan/tripolyphosphate (CS-TPP) nanoparticles
and alginate microparticles incorporated into fish feed. The distribution of the administered vaccines in
Keywords:
IPNV
different organs and transcription of VP2 gene were confirmed by RT-PCR assay at day 30 post boost-
Trout vaccination. Transcript levels of IFN-1, Mx-1, IgM, IgT and CD4 genes was dependent on vaccine dose
Oral DNA vaccine and was significantly up-regulated in head kidney of all orally vaccinated fish groups compared to
CS-TPP nanoparticles controls (pcDNA3.1). Cumulative mortalities post-challenge with virulent isolate of the virus were lower
Alginate microparticles in the vaccinated fish and a relative percentage survival (RPS) of 59% and 82% were obtained for the 10
and 25 mg/fish pcDNA3.1-VP2 groups, respectively. Vaccination with the same amount of pcDNA3.1-VP2
encapsulated with CS-TPP nanoparticles resulted in RPS of 47 %and 70%, respectively. Detectable anti-
IPNV antibodies were shown until 90 days postvaccination. The orally administrated vaccines signifi-
cantly decreased VP4 transcripts thus contributing to reducing viral load in surviving fish on day 45 post-
challenge. In conclusion, these results show good to high protection post-vaccination alongside with
significant up-regulation of key immune genes and detectable levels of circulating antibodies after oral
administration of the DNA vaccine formulated in CS-TPP nanoparticles and alginate microparticles in fish
feed.
© 2017 Elsevier Ltd. All rights reserved.

1. Introduction viral infections (Brudeseth et al., 2013; Gudding et al., 2014).


However, a number of vaccines such as live, formalin or heat
Vaccination is the most effective method for prevention of in- inactivated whole virus, fusion protein, subunit, virus-like particles,
fectious diseases, however, most commercially available vaccines and DNA vaccines have been tested experimentally against some
are targeting bacterial diseases and there are relatively few against viral diseases of fish (Gudding et al., 2014; Munang'andu and
Evensen, 2015). DNA vaccine technology can provide safe and un-
der given conditions more effective vaccine, and have been shown
to induce strong innate immune responses, as well as humoral and
* Corresponding author. Faculty of Veterinary Medicine, University of Tehran,
Tehran, Iran.
cellular adaptive immunities (Heppell and Davis, 2000; Kurath,
E-mail address: msoltani@ut.ac.ir (M. Soltani). 2005. Overview of recent DNA vaccine development for fish. In

http://dx.doi.org/10.1016/j.dci.2017.05.004
0145-305X/© 2017 Elsevier Ltd. All rights reserved.
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 179

Progress in Fish Vaccinology (ed. Midlyng and Karger). Basel; pp. Altun et al., 2010; Leal et al., 2010). Research into alginate de-
201e213.) livery of DNA vaccines against viral pathogens of fishes such as
DNA vaccines consist of plasmid DNA that are delivered through LCDV, IHNV and IPNV suggests an alternative effective delivery
the intramuscular route and encodes specific proteins expressed in strategy (Tian et al., 2008; de las heras et al., 2010; Ballesteros et al.,
cells of the inoculated host inducing strong and long-lasting im- 2015).
munity. Oral delivery is preferred from a user viewpoint and is IPNV is the etiological agent of a highly contagious viral disease,
almost stress-free to the fish (Embregts and Forlenza, 2016). Some infectious pancreatic necrosis (IPN), causing high mortalities in
antigen-encapsulation methods have been tested for oral delivery different fish species worldwide (Reno, 1999; Evensen and Santi,
of vaccines against lymphocystis disease virus (LCDV; Tian et al., 2008). The IPNV belongs to the family Birnaviridae, genus Aqua-
2008), infectious pancreatic necrosis virus (IPNV; de las heras birnavirus with the bisegmented genome (A and B) of double-
et al., 2010), infectious hematopoietic necrosis virus (IHNV; stranded RNA where segment A is the largest segment (approxi-
Adomako et al., 2012; Ballesteros et al., 2015) and betanodavirus mately 3.1 kbp). Segment A encodes VP2 that contains most of the
infection (Vimal et al., 2014). neutralizing epitopes of the virus (Tarrab et al., 1993; Frost et al.,
Many studies have been performed evaluating the microparti- 1995; Fridholm et al., 2007) and structural protein VP3 and the
cles and nanoparticles (NPs) for oral delivery of DNA vaccines in protease, VP4. It also encodes a non-structural protein of unknown
fish, however, nanoparticles are thought to be superior to micro- function, VP5 (Evensen and Santi, 2008). IPNV infects both fry and
particles because of the size consistency compared to microparti- juveniles of farmed and wild fish e.g. Atlantic salmon, brook trout
cles (Gregory et al., 2013; Vimal et al., 2014; Ballesteros et al., 2015). and rainbow trout resulting in high morbidity and mortality
Immune responses due to intestinal antigen uptake and the (Evensen and Santi, 2008). Moreover, the IPNV establishes lifelong
related mechanisms are well documented as reviewed elsewhere asymptomatic carriers shedding and spreading of the virus
(Lokka and Koppang, 2016). Encapsulation of DNA vaccine is a (Rodriguez Saint-Jean et al., 1991; Johansen and Sommer, 1995).
suitable strategy to deliver antigens at mucosal surface for inducing Oral DNA vaccines are considered as a new strategy and ideal way
immunity by targeting microfold (M)-like cells in higher verte- to immunize large numbers of small fish against IPNV (reviewed in
brates and translocation to antigen presenting cells in the sub- Evensen and Leong, 2013; Gudding et al., 2014; Hølvold et al., 2014;
epithelial compartment (dendritic cells and macrophages) for Embregts and Forlenza, 2016). Ballesteros et al. (2014) showed high
initiation of CD8 cytotoxic T cell responses (MHC-I pathway), CD4 protection (RPS ¼ 78%) of rainbow trout fry (1.5 g) against IPNV
helper T cells engagement (MHC-II pathway), as well as natural using administration of 3 consecutive days of 10 mg/day/fish of VP2-
killer (NK) cell activation (Tonheim et al., 2008; Fuglem et al., 2010; encoding DNA vaccine encapsulated in alginate microsphere in
Mutoloki et al., 2015). The orally delivered antigen can also activate feed. Nonetheless, considering the renewal period of intestinal
the antigen-specific B cells and antibody production via CD4 cells epithelium in the second gut segment depends on temperature and
resulting in adaptive humoral immune response (Tonheim et al., fish species (10e15 days at 20  C) and antigen breakdown in the
2008; Embregts and Forlenza, 2016). Furthermore, plasmid back- harsh gastric and the high tolerogenic gut environment, optimizing
bone contains unmethylated CpG motifs be adjuvants that bind to the time and dose of DNA plasmid delivery as well as the appro-
Toll-like receptor 9 (TLR9) that initiates signal transduction priate vaccine design is important for successful oral DNA vacci-
resulting in cellular immune response (Pietretti and Wiegertjes, nation (Embregts and Forlenza, 2016).
2014). Moreover, up-regulation of many genes related to innate In the present study, we constructed a DNA vaccine encoding
and adaptive-immune responses have been reported after oral DNA the VP2 protein of a prevalent isolate of IPNV (Sp strain) in trout
vaccination of fish (Ballesteros et al., 2012, 2014 and 2015). None- farms and examined its efficacy by testing different doses and
theless, antigen type, dose, encapsulation as well as delivery boosting 2 weeks later using oral delivery with CS-TPP nano-
methods influence all of the mentioned immune responses and particles and alginate microparticles incorporated into fish feed.
vaccine efficacy (reviewed in Embregts and Forlenza, 2016).
Chitosan and alginate are cationic and anionic biopolymers,
respectively, have received much attention and been extensively
2. Materials and methods
studied for nano/micro encapsulation in different types of fish
vaccines that their properties and formulation methods have been
2.1. Virus and cell culture
extensively reviewed elsewhere (Plant and Lapatra, 2011; Ji et al.,
2015; Mutoloki et al., 2015; Embregts and Forlenza, 2016).
A prevalent isolate of the IPNV (Sp strain) in Iranian trout farms
Chitosan is non-toxic, biocompatible and biodegradable
was propagated in CHSE-214 cells (Fryer et al., 1965) for use in
biopolymer with the advantage of mucoadhesion which results in
immunoassays and for challenge of fish. The cells were cultured in
the particles adhering to the mucus membrane facilitating delivery
Minimal Essential Medium (MEM) supplemented with 10% FBS, L-
(Takeuchi et al., 1996; Rao and Sharma, 1997). An alternative to
glutamine, 100 IU penicillin G and 100 mg/ml of streptomycin. The
chitosan-based delivery systems for oral application is the chitosan/
virus was titrated according to Reed and Muench (1938).
tripolyphosphate (CS-TPP) nanoparticles, which is simple to pre-
pare and safe for use and has been used to encapsulate peptides and
proteins (Xu and Du, 2003), siRNA (Katas and Alpar, 2006), shRNA
(Wang et al., 2009), deliver drugs (Lam et al., 2006) as well as oral 2.2. Fish
plasmid DNA (Csaba et al., 2009; Vimal et al., 2012, 2014). The safety
and efficacy of CS-TPP nanoparticles have also been shown for oral Healthy rainbow trout (O. mykiss) weight of 3 ± 0.32 g were
delivery of DNA vaccines against bacterial and viral pathogens of obtained from a local hatchery in Tehran province (Firuzkuh city).
fish (Vimal et al., 2012, 2014). As Vimal et al. (2014) reported, Fish were maintained at the faculty of veterinary medicine (Uni-
administration of 100 mg/fish of an oral DNA vaccine encapsulated versity of Tehran, Tehran) laboratory with a flow-through system of
with CS-TPP nanoparticles inducing the 60% protection in 10e15 g fresh water (7 L/min) at 15  C, and fed daily with a pelleted diet
Asian sea bass (Lates calcarifer) against nodavirus. (Skretting, UK). Fish were acclimatized for 2 weeks in the labora-
However, alginate particles to deliver some bacterins to fishes tory before vaccination. All applicable guidelines for the care and
have not resulted in an enough protection (Romalde et al., 2004; use of animals were followed.
180 S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189

2.3. Construction of the DNA vaccine (pcDNA3.1-VP2) RNA was extracted from 20 mg of the homogenized tissues using
the RiboEx SL Total RNA extraction kit (GeneAll, Korea). The cDNA
The VP2 gene of IPNV was cloned into a eukaryotic expression synthesis was carried out in a total volume of 25 ml from 5 ml of
vector pcDNA3.1 (Invitrogen, USA) under the control of the cyto- extracted RNA using HyperScript™ First Strand Synthesis Kit
megalovirus (CMV) promoter, yielding pcDNA3.1-VP2. The (GeneAll, Korea) according to the manufacturer's recommenda-
pcDNA3.1-VP2 was verified using HindIII and XhoI endonuclease tions. Primer pairs, SVP2-F (50 GTTCGACAAGCCATACGTCC 30 ) and
analysis and the recombinant plasmid was then amplified in SVP2-R (50 GCTTGGTGATGTTCTCGGTC 30 ) were used to RT-PCR
Escherichia coli (TOP10), and the cells were grown in LB broth with amplification of exogenous VP2 gene with an expected size of
ampicillin. The constructed plasmid DNA was then isolated with 405 bp. The RT-PCR amplifications were performed in a final vol-
the Endofree Plasmid Mega Purification Kit (Qiagen, USA) according ume of 25 ml containing 12.5 ml Taq DNA Polymerase Mix Red
to the manufacturer's instructions. The DNA concentration was (GeneAll, Korea), 2 ml of cDNA, 1 ml of each primer pair (10 pmol)
measured using a spectrophotometer (NanoDrop 2000, Thermo and 8.5 ml nuclease-free water. Amplification was carried out with
scientific, Spain), and stored at 20  C until use. 40 cycles of 94  C for 30 s, 58  C for 30 s and 72  C for 45 s followed
by a final extension at 72  C for 10 min. The amplification products
2.4. Encapsulation of DNA vaccine were resolved by electrophoresis using a 1% agarose under UV light.

The pcDNA3.1-VP2 and pcDNA3.1 plasmids were encapsulated 2.7. Immune gene transcription (RT-qPCR)
in sodium alginate microparticles and CS-TPP nanoparticles as
described previously (Bozkir and Saka, 2004; de las heras et al., At days 3, 7, 15 and 22 post-vaccination 3 trout from each group
2010), lyophilized and stored at 4  C. Briefly, an equal volume of were sacrificed with overexposure to clove oil then the head-
heated CS-TPP (prepared by ionic gelation process; Vimal et al., kidney tissue was removed aseptically, and relative expression of
2012) and pcDNA3.1-VP2 solutions was quickly mixed together IFN-1 and Mx-1 mRNA and genes related to adaptive immune re-
and vortexed at 2500 rpm for 30 s. The pcDNA3.1-VP2 loaded CS- sponses consisting of IgM, IgT, CD4 and CD8 was assessed 15 and 30
TPP particles were separated by centrifugation at 20,000 rpm for days post vaccination (dpv) were quantified using Real-Time PCR
30 min at 10  C. (Applied Biosystems) and SYBR Green qPCR Master Mix. RNA
For preparation of microparticles, 1.5 mL of pcDNA-VP2 (1 mg/ extraction and cDNA synthesis were performed as described above
ml) were mixed with 2.5 mL of sodium alginate (3% w/v), then the (section 2.6).
mixture after stirrer (10 min at 500 rpm) was emulsified (30 min at Each RT-qPCR reaction (containing 12.5 ml 2SYBR Green PCR
900 rpm) with Span 80 (0.5 ml) and paraffin oil (100 mL). A volume Master Mix, 200 nM of each primer 100 ng cDNA template and
of 2.5 ml of 0.15 M CaCl2 was then added to the emulsion and nuclease-free water up to final volume of 25 ml) were carried out in
stirred at 900 rpm for 2 h. Alginate microparticles were collected by duplicate and incubated for 15 min at 95  C (1 cycle), followed by 40
centrifugation (10 min at 1000g) and washed with ethanol (70%). cycles at 95  C for 15 s, 59  C for 1 min. The melting curve of each
The unbound pcDNA-VP2 content in the supernatants was amplicon was examined. The expression of the target genes was
quantified by UV spectrophotometer at 260 nm. The encapsulation corrected based on the endogenous control expression (EF-1 a) and
efficiency (EE) was calculated using the following equation: calculated relative to empty plasmid according to the 2-DDCt method
EE ¼ [(Total amount of pcDNA3.1-VP2  Free amount of as fold change (Livak and Schmittgen, 2001). However, relative
pcDNA3.1-VP2 in supernatant)/Total amount of pcDNA3.1- expression of VP4 gene in challenged fish was showed as 2-DCt,
VP2]  100. where DCt was determined by subtracting the average EF1-a Ct
value from the average target Ct. All amplifications were performed
2.5. Vaccination in duplicate. The primers used are listed in Table 1.

Rainbow trout (3 ± 0.32) g were orally immunized with feed 2.8. IPNV challenge and viral replication
pellets at 5% of body weight containing alginate microparticles and
CS-TPP nanoparticles loaded with pcDNA3.1-VP2 (named At 30 days post booster vaccination 30 fish form each group in
AlgepcDNAVP2 and CS-TPPepcDNAVP2, respectively) in separate two subgroups (n ¼ 15) were challenged by IP injection of 0.2 ml/
trial groups (each group 90 fish) at temperature 15  C for 90 days. fish with IPNV at 107 TCID50 mL1 and unvaccinated fish were
The feed pellets containing vaccine were prepared according to injected with the same volume of PBS. The mortality was recorded
Ballesteros et al. (2014). daily for 30 days and the identity of IPNV in the dead fish was
Two groups of trout were immunized with vaccine-coated feed confirmed through amplification of the VP4 viral gene. At day 45
pellets containing CS-TPP nanoparticles loaded with 10 or 25 mg of post-challenge when fish did not exhibit clinical symptoms, three
pcDNA3.1-VP2 and boosted 15 days later with the same amount of trout from each group were sacrificed and spleen and head kidney
plasmid. In control group, fish orally immunized with CS-TPP were removed for RT-qPCR analysis of IPNV-VP4 gene as an indi-
nanoparticles containing 10 mg of empty-plasmid (pcDNA3.1) and cator of viral replication according to Ballesteros et al. (2014).
boosted 15 days later with the same amount of empty-plasmid.
Furthermore, the same experiment was performed using alginate 2.9. Indirect enzyme-linked immunosorbent assay (ELISA)
microparticles loaded with the same concentration of pcDNA3.1-
VP2. Unvaccinated fish feed with commercial pellets used as On days 15, 30, 45, 60 and 90 post-vaccination, three fish of each
negative controls (Fig. 1). group were anesthetized by clove oil and the blood samples were
collected from the caudal vein, clotted and serua samples were
2.6. Transcription and tissue distribution of DNA vaccine collected by centrifuging at 500g for 10 min. Serua samples were
(pcDNAvp2) by RT-PCR stored at 20  C until used. Briefly, 96-well ELISA plates were
coated by with 100 ml (10 7) well1 IPNV (previously propagated in
At day 30 post-vaccination three fish from each group were CHSE-214 cells) dissolved in coating buffer (carbonate-bicarbonate
sacrificed by overexposure to clove oil, and then tissues of spleen, solution, pH ¼ 9.6) cells in and incubated overnight at 4  C.
gill, head kidney, liver and intestine were removed aseptically, and Following a wash in phosphate buffered saline containing 0.05%
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 181

Fig. 1. Protocol of immunization, virus challenge and sampling times. Fish were orally immunized with alginate microparticles and CS-TPP nanoparticles containing 10 and 25 mg of
pcDNA3.1-VP2 and boosted 15 days later with the same amount of vaccine. Control fish groups were fed with alginate microparticles and CS-TPP nanoparticles containing 10 mg of
empty-plasmid and boosted 15 days. Unvaccinated fish were used as negative controls. On 30 days post-booster vaccination fish were challenged by IP injection of 0.2 ml/fish with
IPNV at 107 TCID50 mL1.

Table 1
The primers used for the RT-qPCR.

Genes Acc. No FW-Primer (50 -30 ) Rev-Primer (50 -30 )

VP4 M18049 AGGAGATGACATGTGCTACACCG CCAGCGAATATTTTCTCCACCA


IgM X65263 ACCTTAACCAGCCGAAAGGG TGTCCCATTGCTCCAGTCC
IgT AY870265 AGCACCAGGGTGAAACCA GCGGTGGGTTCAGAGTCA
CD4 AY973030 CCTGCTCATCCACAGCCTAT CTTCTCCTGGCTGTCTGACC
CD8 NM_001124263 AGTCGTGCAAAGTGGGAAAG GGTTGCAATGGCATACAGTC
IFN-1 NM_001124531 AAAACTGTTTGATGGGAATATGAAA CGTTTCAGTCTCCTCTCAGGTT
MX-1 NM_001171901 AGCTCAAACGCCTGATGAAG ACCCCACTGAAACACACCTG
EF-1 a AF498320 GATCCAGAAGGAGGTCACCA TTACGTTCGACCTTCCATCC

Tween 20 (PBS-T), the wells were blocked with 3% dried skimmed 2.10. Statistics analysis
milk in phosphate buffered saline (300 ml/well) and incubated for
2 h at 22  C. Plates were washed with PBS-T then serum samples Data was statistically analyzed by one-way analysis of variance
serially diluted with PBST-5% BSA, were added to the wells (100 ml (ANOVA) using SPSS package (SPSS 1998). Differences were
well1) and incubate for 3 h at 22  C. PBS was used as negative considered statistically significant at p < 0.05.
control. The plates were again washed with PBS-T then 100 mL of
the anti-rainbow trout IgM monoclonal antibody (Aquatic Di- 3. Results
agnostics Ltd, Stirling, Scotland) were added to the wells and
incubate for 60 min at 22  C. After washing with PBS-T, the bound The encapsulation efficiency of alginate sodium and CS-TPP to
antibodies were detected by adding 100 ml well1 anti-mouse IgG- encapsulate pcDNA3.1-VP2 were determined and the results
HRP (Bio-Rad) diluted 1/1000 in conjugate buffer (1% BSA solution) revealed high encapsulation efficiencies with 87.8% and 84.2% of
and incubation for 60 min at 22  C. After washing with PBS-T DNA binding with alginate sodium microparticles and CS-TPP
antibody binding was then visualized by adding 100 mL of tetra- nanoparticles, respectively.
methylbenzidine dihydrochloride (TMB; BioLegend, USA) to each
well and incubation for 10 min at 22  C. The reactions were stopped
by adding 50 mL of stop the solution (2 M H2SO4 in distilled water) 3.1. Transcription analysis of AlgepcDNAVP2 and CS-
to the wells. The plates were read at 450 nm in Epoch Microplate TPPepcDNAVP2
Spectrophotometer (BioTek).
Transcription analyses of VP2 gene in tissues of spleen, head
kidney, gill, liver and intestine in orally vaccinated fish with
182 S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189

AlgepcDNAVP2 and CS-TPPepcDNAVP2, and the control groups either the AlgepcDNAVP2 or CS-TPPepcDNAVP2 in comparison
(n ¼ 3) at 30 dpv were performed by RT-PCR reaction. The RT-PCR with the control group (pcDNA) or fish vaccinated with 10 mg of the
products with an expected size of 405 bp specific to the VP2 gene of oral vaccines on 15 and 30 dpv. However, no significant change was
IPNV were obtained in the all tested tissue samples of vaccinated observed in CD8 gene expression in the head kidney of vaccinated
fish that confirmed the distribution of the administered oral vac- fish.
cines in different tissues of organism and transcription of VP2 as
well. No amplification was detected in tissue samples from the 3.3. IPNV challenge/cumulative mortality and relative percent
unvaccinated fish (control groups). The results are shown in Fig. 2. survival (RPS)

3.2. The RT-qPCR analysis of immune-related genes in the head The orally vaccinated and control groups were challenged with
kidney of orally vaccinated trout IPNV at 30 days post-vaccination. The cumulative mortality was
recorded daily for 30 days. The results of cumulative mortality and
3.2.1. Transcription of innate immune-related genes (IFN-1 and RPS are shown in Fig. 5AeB. Cumulative mortalities in unvaccinated
MX-1 gene) control were 57%, and 57% and 54% in groups vaccinated with
The effect of oral DNA vaccination with the AlgepcDNAVP2 or empty plasmid encapsulated with alginate microparicles and CS-
CS-TPPepcDNAVP2 through the feed on the expression of IFN-1 and TPP nanoparticles, respectively. Fish vaccinated with alginate
MX-1 genes related to the innate immune response, and IgM, IgT, microparicles of 10 or 25 mg pcDNA3.1-VP2 gave cumulative mor-
CD4 and CD8 genes belong to adaptive-immune response in the talities of 24% and 10%, resulting in RPS of 59% and 82%, respec-
head kidney of vaccinated trout was evaluated by RT-qPCR analysis. tively. Cumulative mortalities in CS-TPPepcDNAVP2 groups were of
The expression of induced genes is shown in fold values 30% and 17%, with corresponding RPS of 47 and 70% for the 10 and
(pcDNAVP2/pcDNA). The RT-qPCR analysis revealed a significant 25 mg groups, respectively.
increase in the expression of IFN-1 and MX-1 genes of vaccinated
fish with 10 or 25 mg dose of either the AlgepcDNAVP2 or CS- 3.4. IPNV challenge/viral load
TPPepcDNAVP2 relative to control group (empty plasmid; pcDNA)
on 3, 7, 15, 22 dpv (Fig. 3). However, the expression levels of IFN-1 The relative expression of VP4 gene in kidney and spleen
and MX-1 genes in fish received 25 mg of vaccines (AlgepcDNAVP2 vaccinated and unvaccinated trout was determined by RT-qPCR in
or CS-TPPepcDNAVP2) were significantly higher than 10 mg of one surviving fish post-challenge (Fig. 6). The results showed signifi-
(about 2 fold). The RT-qPCR for IFN-1 and Mx of the same cDNA cantly lower levels of VP4 mRNA in surviving vaccinated fish at 45
from vaccinated trout showed similar kinetics, with peaking at 3 days post-challenge (P<0.05). The highest level of VP4 expression
dpv, which then decreased by day 15 and increased again on 22 dpv was observed in the head kidney of the non-vaccinated IPNV group,
after boosting at 15 dpv. approximately 15 and 9 folds higher than (AlgepcDNAVP2 and CS-
TPPepcDNAVP2) vaccinated and infected fish. The expression level
3.2.2. Transcription of adaptive immune-related genes (IgM, IgT, of VP4 in head kidney was slightly higher than spleen tissues as
CD4 and CD8 genes) well as in IPNV control (unvaccinated) to empty-plasmid vacci-
The results of RT-qPCR assays for adaptive immune-related nated. The VP4 expression in spleen and kidney of vaccinated trout
genes including CD4, CD8, IgM and IgT genes in the head kidney was nearly undetectable. Nonetheless, VP4 expression in vacci-
of vaccinated fish are shown in Fig. 4. The transcription levels of nated fish with 25 mg of the vaccines (AlgepcDNAVP2 or CS-
IgM and IgT genes were significantly up-regulated in all vaccinated TPPepcDNAVP2) was significantly lower than 10 mg of one (about 3
groups related to control group (pcDNA). In addition, the effect of fold).
the oral vaccines on IgM and IgT genes expression was dose-
dependent and on 30 dpv their levels were significantly higher 3.5. Anti-IPNV serum antibodies of orally vaccinated fish
than 15 dpv. The transcription levels of CD4 gene was significantly
higher in the kidney of orally vaccinated fish with 25 mg dose of The serua samples obtained from orally vaccinated fish with

Fig. 2. RT-PCR analysis of VP2 gene of IPNV in different organs of oral vaccinated fish with different doses (10 mg and 25 mg) of AlgepcDNAVP2 (A) and CS-TPPepcDNAVP2 (B) at day
30 post-vaccination. Lanes are M: 100-bp DNA ladder; 1e2: spleen, 3e4: gill, 5e6: liver, 7e8: kidney, 9e10: intestine; P: positive control (pcDNA3.1-VP2), N: negative control
(empty-pcDNA3.1). RT-PCR products were analyzed on a 1% agarose gel.
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 183

Fig. 3. IFN-1 and MX-1 gene expression in the kidney of orally vaccinated trout with AlgepcDNAVP2 (left) and CS-TPPepcDNAVP2 (right) in feed, at 3, 7, 15 22 days post vaccination.
Rainbow trout fry (weight of 3 g; n ¼ 90) were orally immunized with alginate microparticles (A) and CS-TPP nanoparticles (B) containing 10 and 25 mg of pcDNA3.1-VP2 and
boosted 15 days later with the same amount of vaccine. Control fish groups were fed with alginate microparticles and CS-TPP nanoparticles (B) containing 10 mg of empty-plasmid
and boosted 15 days, and unvaccinated fish were used as negative controls. Data were normalized based on endogenous EF-1a gene and presented as mean fold increase relative to
empty plasmid (2-DDCT method; n ¼ 3e5). Asterisks and black point indicate significant differences (p < 0.05 and >2 folds) between vaccinated and unvaccinated fish or between
vaccinated fish with10 and 25 mg of pcDNA3.1-VP2, respectively.

alginate microparticles or CS-TPP nanoparticles loaded with for mass vaccination of small fish (Embregts and Forlenza, 2016).
pcDNA3.1-VP2 were tested for anti-IPNV antibodies using ELISA. Moreover, subunit and inactivated vaccines predominantly induce
Anti-IPNV antibodies in the serua of all vaccinated fish groups were humoral immune response which may prevent against mortality
detected 90 dpv (Fig. 7AeB). The antibody levels in sera of vacci- but is insufficient to prevent a carrier state or virus shedding
nated trout were progressively increased by 45e60 days post- (Bootland et al., 1995; Melby and Falk, 1995; Urquhart et al., 2008).
vaccination and reached the peak of antibody and then gradually Despite the advances in fish vaccination and the availability of some
decreased. The level antibody in AlgepcDNAVP2 and CS- commercial vaccines, IPN outbreaks occur in vaccinated fish
TPPepcDNAVP2 were significantly higher than control groups because of the above reasons. Therefore, providing an effective
(p<0.05). Also, the levels of Anti-IPNV antibodies expression in vaccine for immunization of a large number of small fish is still a
vaccinated fish with 25 mg pcDNA3.1-VP2 encapsulated with algi- necessary way and oral DNA vaccine delivery is one potential
nate microsphere and CS-TPP nanoparticles were significantly alternative goal.
higher than 10 mg groups. In this study, we examined the efficacy and immune responses
induced by a DNA vaccine encoding VP2 gene of an isolate of IPNV
(Sp strain) delivered by the oral route using CS-TPP nanoparticles
4. Discussion
and alginate microparticles incorporated into fish feed for rainbow
trout fry.
Current commercial vaccines against IPN are mostly inactivated
Several studies on the tissue distribution and persistence of
vaccines, delivered intraperitoneally and a few subunit oral vac-
plasmid DNA have been reported for DNA vaccines after oral
cines are available in Norway (MSD) but in principle no longer in
vaccination using CS-TPP nanoparticles and alginate microparticles
use, in Chile (Centrovet, Chile) and in Canada (Merck Animal
in fish. These formulations were found promising as delivery sys-
Health, USA). IP injection vaccination imposes stress and may result
tems that protect against degradation in the stomach and anterior
in some local side effects. Also, this way of vaccination is unusable
184 S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189

Fig. 4. CD4, CD8, IgM and IgT gene expression in trout after the oral administration of the alginat microparticles (left) and CS-TPP nanoparticles (right) loaded with pcDNA3.1-VP2 in
feed, at 15 and 30 dpv. Rainbow trout fry (weight of 3 g; n ¼ 90) were orally immunized as described in the legend of Fig. 3. Data were normalized based on endogenous EF-1a gene
and presented as mean fold increase relative to empty plasmid (2-DDCT method; n ¼ 5). Asterisks and black point indicate significant differences (p < 0.05 and >2 fold) between
vaccinated and unvaccinated fish or between vaccinated fish with10 and 25 mg of pcDNA3.1-VP2, respectively.

gut, allowing uptake of plasmid or antigen in the posterior of trout fed with feed containing alginate microparticles encapsu-
(Tonheim et al., 2008; de las heras et al., 2010; Vimal et al., 2012, lated DNA vaccine (Ballesteros et al., 2014, 2015). Also, expression of
2014; Ballesteros et al., 2014, 2015). For instance, transcript of the the capsid protein gene of nodavirus has been reported in tissues of
IPNV-VP2 and IHNV-G genes have been shown in different tissues Asian sea bass after oral DNA vaccination using CS-TPP
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 185

Fig. 5. Cumulative mortalities of orally immunized rainbow trout with alginate microparticles (A) and CS-TPP nanoparticles (B) loaded with pcDNA3.1-VP2, and challenged by IP
injection of IPNV at 2  106 TCID50/fish. Unvaccinated fish were injected with PBS. Mortality was recorded daily for 30 days.

nanoparticles (Vimal et al., 2014). Our results revealed transcription that limit viral replication and spread (Robertsen, 2006).
and distribution of the pcDNA3.1-VP2 by RT-PCR in tissues of Here, the transcript levels of IFN-1 and MX-1 genes were
spleen, head kidney, gill, liver and intestine of rainbow trout fry fed significantly up-regulated in all vaccinated fish groups compared to
with feed containing AlgepcDNAVP2 and CS-TPPepcDNAVP2 sug- the control group (pcDNA3.1) dependent on vaccine doses (10 or
gesting a good protection of the vaccines from degradation stomach 25 mg) with similar kinetics, peaking at 3 dpv and decreased by day
condition, and uptake across the intestinal epithelium and distri- 15 with slight increase on 22 dpv after boosting at 15 dpv. An up-
bution to other organs. regulation of IFN-1 and MX-1 genes have been shown in brown
Type I Interferons (IFNs) are the first line of defense against viral trout (Salmo trutta; de las heras et al., 2010; Ballesteros et al., 2012)
infections that act through the JAK-STAT pathway by inducing the and rainbow trout (O. mykiss; de las heras et al., 2010; Ballesteros
transcription of Mx and other interferon stimulated genes (ISGs) et al., 2014) only up to 15 days after oral DNA vaccination against
186 S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189

Fig. 6. Relative expression of the IPNV-VP4 gene in kidney and spleen tissues of vaccinated and non-vaccinated trout infected with IPNV. Transcription of the VP4 gene was
recorded in the kidneys and spleen of vaccinated or non-vaccinated control fish at 45 days post-challenge. Data are represented relative to EF1-a expression (2-DCT method; n ¼ 3).
Different letters indicate significant differences between groups at p < 0.05. IPNV ¼ non-vaccinated, challenged controls; CS-TPP-pcDNA3.1 and Alg-pcDNA3.1 are fish given empty
plasmid, then challenged; others are different vaccine groups, then challenged as indicated.

IPNV. Our results suggest that the both vaccines were processed primary lymphoid tissue and the main source of Ig-secreting B cells
and epitopes presented by MHC receptors, inducing IFN response (Zwollo et al., 2005). IgM and IgT are two distinct lineages of B cells
and may produce specific T-lymphocytes for cellular adaptive im- in fish that are important for mucosal immune responses as their
mune responses. Also, these results reflect the involvement of the populations can be increased after oral vaccination (Hansen et al.,
Type-I IFN pathway in the antiviral response against IPNV and a 2005; Ballesteros et al., 2013). In this study, we also examined the
positive effect made by a booster vaccination on this pathway. IgT and IgM gene expression in the head kidney of orally vaccinated
Regarding the adaptive cellular immune response, we also trout, at 15 and 30 dpv. As expected, the constructed DNA vaccine
found a significant increase in transcript levels of CD4 (a marker of strongly enhanced the expression of these two genes which is
helper T cell) in fish fed with feed containing 25 mg pcDNA-VP2 similar to previous studies that revealed both IgM and IgT are up-
vaccines encapsulated in both CS-TPP nanoparticles and alginate regulated in response to alginate encapsulated oral DNA vaccine
microparticles but no significant enhancement was seen in CD8 (a against IPNV and IHNV in rainbow trout (Ballesteros et al., 2014,
marker of cytotoxic T cell) transcript levels. An induction in CD4 and 2015), as well as in the orally vaccinated grouper larvae (Epi-
CD8 genes has been reported in rainbow trout when an oral IHNV- nephelus coioides) against betanodavirus infection (NNV; Kai et al.,
DNA vaccine (100 mg/fish) encapsulated in alginate microparticles 2014). Overall, our results revealed IgT and IgM gene response to
was administered (Ballesteros et al., 2015). In contrast, despite a oral stimulation and activity of our constructed vaccine to stimulate
sufficient protection, transcription of these T-cell surface markers systemic responses and mucosal immunity and substantiate Ig-
were not significantly increased in trout vaccinated 3 consecutive secreting cells are induced following vaccination with the con-
days with 10 mg/fish of an IPNV-DNA vaccine encapsulated in structed vaccine.
alginate microsphere in feed (Ballesteros et al., 2014). These Moreover, the constructed oral DNA vaccines induced circu-
controversial results might be explained by differences in the lating anti-IPNV antibodies, peaking at 6e8 weeks post vaccination
quality, quantity and vaccine strain. Another possibility is the for each group, gradually decreasing at later time. The up-
wrong choice of the tissue or time for evaluating the CD8 and CD4 regulation of IgM, IgT and CD4 in vaccinated fish suggest activa-
responses. Nonetheless, further verifications using functional tion of B and T cells and consequently antibody production.
studies and the transcript analysis of other CD8/CD4 T cells genes Moreover, the kinetics of release of the encapsulated DNA vaccines
markers such as Eomesodermin (Eomes), Granzyme A, T-bet and and degradation of the pcDNA3.1 vector may explain a gradual
FoxP3 and GATA3 (Munang'andu et al., 2013) are required to find increase of antibody level in sera of vaccinated trout, i.e. an issue of
out the exact cellular immune responses to oral DNA vaccines. timing.
In teleost fish, B cells differentiate occurs in the head kidney, the Exception the trout vaccinated with 10 mg CS-TPPepcDNAVP2
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 187

Fig. 7. Anti-IPNV antibodies in serum of orally vaccinated fish with alginate microsphere (A) and CS-TPP nanoparticles (B) loaded with pcDNA3.1-VP2 in feed. Serua samples were
collected on days 15, 30, 45, 60 and 90 dpv, analyzed by ELISA. Data is mean optical density (OD; at 450 nm) of anti-IPNV antibodies in serum of vaccinated fish at a dilution of 1:100.
Different letters indicate significant differences between groups at p < 0.05. Control ¼ non, vaccinated; CS/TPP-pcDNA3.1 are vaccinated with empty plasmid; others are vaccine
groups as indicated.
188 S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189

(RPS ¼ 47%), a high protection was seen during the challenging the Int. J. Pharm. 382, 05e14.
Cui, L.C., Guan, X.T., Liu, Z.M., Tian, C.Y., Xu, Y.G., 2015. Recombinant lactobacillus
vaccinated fish with the virulent strain of IPNV. Moreover, the RPS
expressing G protein of spring viremia of carp virus (SVCV) combined with
in the trout vaccinated with AlgepcDNAVP2 was also higher than ORF81 protein of koi herpesvirus (KHV): a promising way to induce protective
CS-TPPepcDNAVP2 which may be due to polymer type and/or immunity against SVCV and KHV infection in cyprinid fish via oral vaccination.
encapsulation efficiency (Wendorf et al., 2008). Vaccine 33, 3092e3099.
de las heras, A.I., Rodríguez Saint-Jean, S., Perez-prieto, S.I., 2010. Immunogenic and
RT-qPCR analysis IPNV-VP4 gene in the surviving fish also protective effects of an oral DNA vaccine against infectious pancreatic necrosis
revealed a higher expression in unvaccinated fish compared to virus in fish. Fish. Shellfish Immunol. 28, 562e570.
vaccinated trout suggesting inhibiting and decreasing of viral load, Embregts, C.W., Forlenza, M., 2016. Oral vaccination of fish: lessons from humans
and veterinary species. Dev. Comp. Immunol. 64, 118e137.
the advantage of DNA vaccine over commercially available vaccines Evensen, Ø., Leong, J.A., 2013. DNA vaccines against viral diseases of farmed fish.
for prevention of the carrier state (Bootland et al., 1995; Ballesteros Fish. Shellfish Immunol. 35, 1751e1758.
et al., 2014, 2015; Cui et al., 2015). Evensen, Ø., Santi, N., 2008. Infectious pancreatic necrosis virus. In: Mahy, B.W.J.,
Van Regenmortel, M.H.V. (Eds.), Encyclopedia of Virology, third ed. Academic
In conclusion, this study describes the development of protec- Press, Oxford, pp. 83e89.
tive oral DNA vaccines encoding the VP2 protein of IPNV using CS- Fridholm, H., Eliasson, L., Everitt, 2007. EImmunogenicity properties of authentic
TPP nanoparticles and alginate microsphere incorporated into fish and heterologously synthesized structural protein VP2 of infectious pancreatic
necrosis virus. Viral Immunol. 20, 635e648.
feed. The results show that the constructed DNA vaccine protected Frost, P., Havarstein, L.S., Lygren, B., Stahl, S., Endresen, C., Christie, K.E., 1995.
by CS-TPP nanoparticles as well as alginate microsphere gave a Mapping of neutralization epitopes on infectious pancreatic necrosis viruses.
great protection to IPNV and a well prevention carrier state in the J. General Virol. 76, 1165e1172.
Fryer, J.L., Yusha, A., Pilcher, K.S., 1965. The in vitro cultivation of tissue and cells of
survivors. To our knowledge, this is the first report describing CS-
Pacific salmon and steelhead trout. Ann. N.Y. Acad. Sci. 126, 566e586.
TPP nanoparticles for oral delivery of DNA vaccines against a viral Fuglem, B., Jirillo, E., Bjerkås, I., Kiyono, H., Nochi, T., Yuki, Y., Raida, M., Fischer, U.,
pathogen in rainbow trout. It applicability for other fish pathogens/ Koppang, E.O., 2010. Antigen-sampling cells in the salmonid intestinal epithe-
diseases should be explored. lium. Dev. Comp. Immunol. 34, 768e774.
Gregory, A.E., Titball, R., Williamson, D., 2013. Vaccine delivery using nanoparticles
frontiers in cellular and infection microbiology, 3, 13. http://dx.doi.org/10.3389/
Conflict of interest fcimb.2013.00013.
Gudding, R., Lillehaug, A., Evensen, Ø. (Eds.), 2014. Fish Vaccination. John Wiley &
Sons, Ltd, Chichester, UK. http://dx.doi.org/10.1002/9781118806913.ch22.
Authors declare that they do not have any conflict of interest. Hansen, J.D., Landis, E.D., Phillips, R.B., 2005. Discovery of a unique Ig heavy-chain
isotype (IgT) in rainbow trout: implications for a distinctive B cell develop-
mental pathway in teleost fish. Proc. Natl. Acad. Sci. U. S. A. 102, 6919e6924.
Acknowledgments Heppell, J., Davis, H.L., 2000. Application of DNA vaccine technology to aquaculture.
Adv. Drug Deliv. Rev. 43, 29e43.
Hølvold, L.B., Myhr, A.I., Dalmo, R.A., 2014. Strategies and hurdles using DNA vac-
We are grateful for the generous collaboration of Pasteur Insti-
cines to fish. Veterinary Res. 45 (1), 21.
tute of Iran and Central Veterinary Laboratory of Iran Veterinary  Roher, N., 2015. Nanodelivery systems as new tools for
Ji, J., Torrealba, D., Ruyra, A.,
Organization (National laboratory). This study was funded by the immunostimulant or vaccine administration: targeting the fish immune sys-
Research Council of the University of Tehran and the Centre of tem. Biol. (Basel) 4, 664e696.
Johansen, L.H., Sommer, A.I., 1995. Multiplication of infectious pancreatic necrosis
Excellence of Aquatic Animal Health, University of Tehran, Iran virus (IPNV) in head kidney and blood leukocytes isolated from Atlantic salmon,
(750). Salmo Salar L. J. Fish. Dis. 18, 147e156.
Kai, Y.H., Wu, Y.C., Chi, S.C., 2014. Immune gene expressions in grouper larvae
(Epinephelus coioides) induced by bath and oral vaccinations with inactivated
References betanodavirus. Fish. Shellfish Immunol. 40, 563e569.
Katas, H., Alpar, H.O., 2006. Development and characterisation of chitosan nano-
Adomako, M., St-Hilaire, S., Zheng, Y., Eley, J., Marcum, R.D., Sealey, W., particles for siRNA delivery. J. Control. Release 115, 216e225.
Donahower, B.C., LaPatra, S., Sheridan, P.P., 2012. Oral DNA vaccination of Kurath, G., 2005. Overview of recent DNA vaccine development for fish. Dev. Biol.
rainbow trout, Oncorhynchus mykiss (Walbaum), against infectious haemato- 121, 201e213.
poietic necrosis virus using PLGA [Poly (D,L-Lactic-Co-Glycolic Acid)] nano- Lam, T.D., Hoang, V.D., Lien, L.N., Thinh, N.N., Dien, P.G., 2006. Synthesis and char-
particles. J. Fish. Dis. 35, 203e214. acterization of chitosan nanoparticles used as drug. J. Chem. 44, 105e109.
Altun, S., Kubilay, A., Ekici, S., Didinen, B.I., Diler, O., 2010. Oral vaccination against Leal, C.A.G., Carvalho-Castro, G.A., Sacchetin, P.S.C., Lopes, C.O., Moraes, A.M.,
Lactococcosis in rainbow trout (Oncorhynchus mykiss) using sodium alginate Figueiredo, H.C.P., 2010. Oral and parenteral vaccines against Flavobacterium
and poly (lactide-co-glycolide) carrier. Kafkas Univ. Vet. Fak. Derg. 16, 211e217. columnare: evaluation of humoral immune response by ELISA and in vivo ef-
Ballesteros, N., Rodriguez Saint-Jean, S., Encinas, P.A., Perez-Prieto, S., Coll, J., 2012. ficiency in Nile tilapia (Oreochromis niloticus). Aquac. Int. 18, 657e666.
Oral immunization of rainbow trout to infectious pancreatic necrosis virus Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using
(IPNV) induces different immune gene expression profiles in head kidney and realtime quantitative PCR and the 2DDCt method. Methods 25, 402e408.
pyloric ceca. Fish. Shellfish Immunol. 33, 174e185. Lokka, G., Koppang, E.O., 2016. Antigen sampling in the fish intestine. Dev. Comp.
Ballesteros, N.A., Rodriguez Saint-Jean, S., Perez-Prieto, S.I., 2014. Food pellets as an Immunol. 64, 138e149.
effective delivery method for a DNA vaccine against infectious pancreatic ne- Melby, H.P., Falk, K., 1995. Study of the interaction between a persistent infectious
crosis virus in rainbow trout (Oncorhynchus mykiss,Walbaum). Fish. Shellfish pancreatic necrosis virus (IPNV) infection and experimental infectious salmon
Immunol. 37, 220e228. anaemia (ISA) in Atlantic salmon. Salmo salar L. J. Fish. Dis. 18, 579e586.
Ballesteros, N.A., Alonso, M., Saint-Jean, S.R., Perez-Prieto, S.I., 2015. An oral DNA Munang’andu, H.M., Evensen, O., 2015. A review of intra- and extracellular antigen
vaccine against infectious haematopoietic necrosis virus (IHNV) encapsulated delivery systems for virus vaccines of finfish. J. Immunol. Res. 960859. http://dx.
in alginate microspheres induces dose-dependent immune responses and sig- doi.org/10.1155/2015/960859.
nificant protection in rainbow trout (Oncorrhynchus mykiss). Fish. Shellfish Munang'andu, H.M., Fredriksen, B.N., Mutoloki, S., Dalmo, R.A., Evensen, O., 2013.
Immunol. 45, 877e888. The kinetics of CD4þ and CD8þ T-cell gene expression correlate with protection
Ballesteros, N.A., Castro, R., Abos, B., Rodríguez Saint-Jean, S.S., Perez-Prieto, S.I., in Atlantic salmon (Salmo salar L) vaccinated against infectious pancreatic ne-
Tafalla, C., 2013. The pyloric caeca area is a major site for IgMþ and IgTþ B cell crosis. Vaccine 31, 1956e1963.
recruitment in response to oral vaccination in rainbow trout. PLoS One 8, 66118. Mutoloki, S., Munang'andu, H.M., Evensen, Ø., 2015. Oral vaccination of fish e an-
Bootland, L.M., Dobos, P., Stevenson, R.M.W., 1995. Immunization of adult brook tigen preparations, uptake, and immune induction. Front. Immunol. 6, 519.
trout, Salvelinus fontinalis (Mitchill), fails to prevent the infectious pancreatic http://dx.doi.org/10.3389/fimmu.2015.00519.
necrosis virus (IPNV) carrier state. J. Fish. Dis. 18, 449e458. Pietretti, D., Wiegertjes, G.F., 2014. Ligand specificities of Toll-like receptors in fish:
Bozkir, A., Saka, O.M., 2004. Chitosan nanoparticles for plasmid DNA delivery: effect indications from infection studies. Dev. Comp. Immunol. 43, 205e222.
of chitosan molecular structure on formulation and release characteristics. Drug Plant, K.P., Lapatra, S.E., 2011. Advances in fish vaccine delivery. Dev. Comp.
Deliv. 11, 107e112. Immunol. 35, 1256e1262.
Brudeseth, B.E., Wiulsrød, R., Fredriksen, B.N., Lindmo, K., Løkling, K.E., Bordevik, M., Rao, S.B., Sharma, C.P., 1997. Use of chitosan as a biomaterial: studies on its safety
Steine, N., Klevan, A., Gravningen, K., 2013. Status and future perspectives of and hemostatic potential. J. Biomed. Mater. Res. 34, 21e28.
vaccines for industrialised fin-fish farming. Fish. Shellfish Immunol. 35, Reed, L.J., Muench, H.A., 1938. Simple method of estimating fifty percent endpoints.
1759e1768. Am. J. Hyg. 27, 493e497.
Csaba, N., Koping Hoggard, M., Alonso, M.J., 2009. Ionically crosslinked chitosan/ Reno, P.W., 1999. Infectious pancreatic necrosis and associated aquatic birnaviruses.
tripolyphosphate nanoparticles for oligonucleotide and plasmid DNA delivery. In: Woo, P.T.K., Bruno, D.W. (Eds.), Fish Diseases and Disorders. CAB
S. Ahmadivand et al. / Developmental and Comparative Immunology 74 (2017) 178e189 189

International, New York, pp. 1e56. Shanks, A.M., Raynard, R.S., 2008. Estimation of infectious dose and viral
Robertsen, B., 2006. The interferon system of teleost fish. Fish. Shellfish Immunol. shedding rates for infectious pancreatic necrosis virus in Atlantic salmon, Salmo
20, 172e191. salar L, post-smolts. J. Fish. Dis. 31, 879e887.
Rodriguez Saint-Jean, S., Vilas Minondo, M.P., Palacios, A., Perez-Prieto, S., 1991. Vimal, S., Abdul, Majeed, S., Nambi, K.S.N., Madan, N., Farook, M.A., Venkatesan, C.,
Detection of infectious pancreatic necrosis virus in a carrier population of Taju, G., Venu, S., Subburaj, R., Thirunavukkarasu, A.R., Sahul Hameed, A.S., 2014.
rainbow trout (Oncorhynchus mykiss, Richardson), by flow cytometry. J. Fish. Dis. Delivery of DNA vaccine using Chitosan-Tripolyphosphate (CS-TPP) nano-
14. particles in Asian sea bass, Lates calcarifer (Bloch, 1790) for protection against
Romalde, J.L., Luzardo-Alvarez, A., Ravelo, C., Toranzo, A.E., Blanco-Wendez, J., 2004. nodavirus infection. Aquaculture 420e421, 240e246.
Oral immunization using alginate microparticles as a useful strategy for booster Vimal, S., Taju, G., Nambi, K.S.N., Abdul Majeed, S., Sarath Babu, V., Ravi, M., Sahul
vaccination against fish lactoccocosis. Aquaculture 236, 119e129. Hameed, A.S., 2012. Synthesis and characterization of CS-TPP nanoparticles for
Takeuchi, H., Yamamoto, H., Niwa, T., Hino, T., Kawashima, Y., 1996. Enteral ab- oral delivery of gene in fish. Aquaculture 358e359, 14e22.
sorption of insulin in rats from mucoadhesive chitosan-coated liposomes. Wang, S.L., Yao, H.H., Guo, L.L., Dong, L., Li, S.G., Gu, Y.P., Qin, Z.H., 2009. Selection of
Pharm. Res. 13, 896e901. optimal sites for TGFB1 gene silencing by chitosaneTPP nanoparticle-mediated
Tarrab, E., Berthiaume, L., Heppell, J., Arella, M., Lecomte, J., 1993. Antigenic char- delivery of shRNA. Cancer Genet. Cytogenet 190, 8e14.
acterization of serogroup ‘A’ of infectious pancreatic necrosis virus with three Wendorf, J., Chesko, J., Kazzaz, J., Ugozzoli, M., Vajdy, M., O'Hagan, D., Singh, M.,
panels of monoclonal antibodies. J. General Virol. 74, 2025e2030. 2008. A comparison of anionic nanoparticles and microparticles as vaccine
Tian, J.Y., Sun, X.Q., Chen, X.G., 2008. Formation and oral administration of alginate delivery systems. Hum. Vaccines 4, 44e49.
microspheres loaded with pDNA coding for lymphocystis disease virus (LCDV) Xu, Y., Du, 2003. Effect of molecular structure of chitosan on protein delivery
to Japanese flounder. Fish. Shellfish Immunol. 24, 592e599. properties of chitosan nanoparticles. Int. J. Pharm. 250, 215e226.
Tonheim, T.Ch, Bøgwald, J., Dalmo, R.A., 2008. What happens to the DNA vaccine in Zwollo, P., Cole, S., Bromage, E., Kaattari, S., 2005. B cell heterogeneity in the teleost
fish? A review of current knowledge. Fish Shellfish Immunol 25, 1e18. kidney: evidence for a maturation gradient from anterior to posterior kidney.
Urquhart, K., Murray, A.G., Gregory, A., O'Dea, M., Munro, L.A., Smail, D.A., J. Immunol. 174, 6608e6616.

You might also like