Next Article in Journal
Replication Fork Protection Factors Controlling R-Loop Bypass and Suppression
Previous Article in Journal
Acknowledgement to Reviewers of Genes in 2016
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Characterization of the Transcriptome and Gene Expression of Brain Tissue in Sevenband Grouper (Hyporthodus septemfasciatus) in Response to NNV Infection

Department of Aqualife Medicine, College of Fisheries and Ocean Science, Chonnam National University, Yeosu 550-749, Korea
*
Author to whom correspondence should be addressed.
Genes 2017, 8(1), 31; https://doi.org/10.3390/genes8010031
Submission received: 10 November 2016 / Revised: 7 January 2017 / Accepted: 9 January 2017 / Published: 13 January 2017
(This article belongs to the Section Microbial Genetics and Genomics)

Abstract

:
Grouper is one of the favorite sea food resources in Southeast Asia. However, the outbreaks of the viral nervous necrosis (VNN) disease due to nervous necrosis virus (NNV) infection have caused mass mortality of grouper larvae. Many aqua-farms have suffered substantial financial loss due to the occurrence of VNN. To better understand the infection mechanism of NNV, we performed the transcriptome analysis of sevenband grouper brain tissue, the main target of NNV infection. After artificial NNV challenge, transcriptome of brain tissues of sevenband grouper was subjected to next generation sequencing (NGS) using an Illumina Hi-seq 2500 system. Both mRNAs from pooled samples of mock and NNV-infected sevenband grouper brains were sequenced. Clean reads of mock and NNV-infected samples were de novo assembled and obtained 104,348 unigenes. In addition, 628 differentially expressed genes (DEGs) in response to NNV infection were identified. This result could provide critical information not only for the identification of genes involved in NNV infection, but for the understanding of the response of sevenband groupers to NNV infection.

1. Introduction

Grouper is one of the highest valued marine fish and has become an important species in the aquaculture industry of various Asian countries. In Korea, sevenband grouper (Hyporthodus septemfasciatus) is one the favorite grouper fish consumed. Its production rate is increasing. However, viral nervous necrosis (VNN) causes high mortality, especially at the larval and juvenile stage of sevenband groupers during the summer season, which has caused vast economic losses [1].
Viral Nervous Necrosis is a serious disease in the world aquaculture industry [2,3,4]. Firstly, it was reported in bigeye trevally (Caranx sexfasciatus) in the 1980s and since then it has been reported in over twenty species [2,3,4]. The infected fish are usually swimming abnormally and having vacuolization and necrosis of the central nervous system in the brain [3]. In Korea, mass mortalities caused by VNN have been reported from various cultured marine fish such as sevenband grouper (Hyporthodus septemfasciatus), rock bream (Oplegnathus fasciatus), red drum (Sciaenops ocellatus) and olive flounder (Paralichthys olivaceus) since 1990 [5,6,7].
Nervous necrosis virus (NNV), the causative agent of VNN, has non-enveloped icosahedral structure and belongs to the family Nodaviridae (genus Betanodavirus). Its genome contains two single-stranded positive senses RNA: RNA1 (approximately 3.1 kb in length) encodes an RNA-dependent RNA polymerase for viral replication, whereas RNA2 (1.4 kb) encrypts a protein α. During RNA replication, a sub-genomic RNA3 was produced from the 3′ terminus of RNA1 that encodes non-structural proteins B2 [8]. Betanodaviruses have five genogroups based on the T4 region sequence of RNA2 as barfin flounder nervous necrosis virus (BFNNV), red-spotted grouper nervous necrosis virus (RGNNV), striped jack nervous necrosis virus (SJNNV), tiger puffer nervous necrosis virus (TPNNV) [9], and turbot nervous necrosis virus [10].
Although virological and genetic characterizations of NNV have been reported, its infection mechanisms and disease outbreak mechanisms remain unclear. Therefore, systematic approaches are needed to determine its infection mechanisms.
Recently, rapid progress has been made in next generation sequencing (NGS) technology and bioinformatics. They are powerful tools for transcriptome analysis. RNA-Seq by NGS is one of the most useful methods to survey the landscape of a transcriptome since it produces millions of data on gene expression. Numerous novel genes and unraveling expression profiles related phenotypic changes, such as development stages, were identified using RNA-Seq [11,12]. Several studies using RNA-Seq have reported the immune relevant genes of fish after pathogen challenges. For example, zebrafish (Danio rerio) [13], orange-spotted grouper (Epinephelus coioides) [14], large yellow croaker (Pseudosciaena crocea) [15], turbot (Scophthalmus maximus) [16], Japanese sea bass (Lateolabrax japonicus) [17] and common carp (Cyprinus carpio) [18] in response to pathogen challenges have been analyzed.
In this study, we analyzed the brain transcriptome of sevenband grouper in response to NNV infection. We annotated the transcripts using Gene Ontology (GO) and non-redundant (nr) databases of GenBank. In addition, we obtained differential expression genes (DEGs) between sevenband grouper brain infected by NNV and non-infected sevenband grouper brain. To the best of our knowledge, this is the first study that reports the transcriptome of brain tissue of NNV-infected sevenband grouper by RNA-Seq. Our transcriptome analysis data will provide valuable genomic information to determine the functional roles of these genes related to NNV infection and VNN outbreak in the future.

2. Materials and Methods

2.1. Ethics Statment

The experiments using sevenband grouper were carried out in strict accordance with the recommendations of the Institutional Animal Care and Use Committee of Chonnam National University (Permit Number: CNU IACUC-YS-2015-4).

2.2. Experimental Fish and Virus

Sevenband groupers were purchased from an aqua-farm without history of VNN occurrence. Prior to experiment, brain tissues of 10 fish were randomly sampled from the aqua-farm and subjected to reverse transcription polymerase chain reaction (RT-PCR) analysis to determine betanodavirus infection according to a previous report [9]. The NNV (SGYeosu08) [19] isolated from sevenband grouper aqua-farm in Korea was propagated in the striped snakehead (SSN-1) cell line. SSN-1 cells were grown at 25 °C in Leibovitz L-15 medium (Sigma Aldrich, St. Louis, MO, USA) containing 10% fetal bovine serum (FBS, Gibco, Waltham, MA, USA), 100 µg/mL streptomycin and 150 U/mL penicillin G. NNV was inoculated onto SSN-1 cell monolayer and incubated at 25 °C for the virus propagation. After the cells were completely lysed, virus titer was calculated by the Reed and Muench method [20]. Viral samples were aliquoted into small volumes and stored at −80 °C until use.

2.3. Virus Challenge

Twenty fish (mean body weight, 20 g) were reared in two 40 L tanks (n = 10/tank) at 25 °C. Ten fish were intramuscularly injected with NNV at doses of 103.8 TCID50/fish. The remaining 10 fish were injected with L15 medium as a control. The challenged fish were observed daily. The NNV infected fish died from day 3 after infection and showed 100% of cumulative mortality after 1 week. The moribund fish at days 3 and 4 were selected for sampling. Brain tissues of three of ten challenged fish from mock and the virus-challenge group were collected and pooled for NGS analysis, respectively.

2.4. Next Generation Sequencing of Transcriptome

To obtain high-throughput transcriptome data of sevenband grouper, complementary DNA (cDNA) libraries were prepared for 100 bp paired-end sequencing using a TruSeq RNA Sample Preparation Kit (Illumina, San Diego, CA, USA) according to the manufacturer’s protocols. They were then paired-end (2 × 100 bp) sequenced using an Illumina HiSeq2500 system (Illumina, San Diego, CA, USA).

2.5. Transcriptome Assembly and Functional Annotation

Prior to de novo assembly, paired-end sequences were filtered and cleaned using an NGS QC toolkit [21] to remove low quality reads (Q < 20) and adapter sequences. In addition, bases of both ends less than Q20 of filtered reads were removed additionally. This process is to enhance the quality of reads due to mRNA degradation in both ends of it as time goes on [22]. Only high quality reads were used for de novo assembly performed by Trinity (version 20130225) using default values [23]. To remove the redundant sequences, CD-HIT-EST [24] was used. NCBI Blast (version 2.2.28) was applied for the homology search to predict the function of unigenes. The function of unigenes was predicted by Blastx to search all possible proteins against the NCBI Non-redundant (NR) database (accessed on 17 July 2013). The criterion regarding significance of the similarity was set at Expect-value less than 1 × 10−5.

2.6. Differentially Expressed Genes Analysis

After obtaining the assembled transcriptome data using Trinity, gene expression level was measured with RNA-Seq by Expectation Maximization (RSEM), a tool for measuring the expression level of transcripts without any information on its reference [25]. The TCC package was used for DEG analysis through the iterative DEGES/DEseq method [26]. Normalization was progressed three times to search meaningful DEGs between comparable samples [27]. The DEGs were identified based on the p-value of less than 0.05.

2.7. GO Enrichment of Differentially Expressed Genes

The GO database classifies genes according to the three categories of Biological Process (BP), Cellular Component (CC) and Molecular Function (MF) and provides information on the function of genes. To characterize the identified genes from DEG analysis, a GO based trend test was carried out through the Fisher’s exact test. Selected genes with p-values of less than 1 × 10−5 were regarded as statistically significant.

2.8. Data Deposition

All the raw read files were submitted to the sequence reads archive (SRA), NCBI database (accession number—SRR5091816).

3. Results

3.1. Sequence Analysis of the Transcriptome

Sequencing of the two libraries (mock and NNV-infected brain tissue) using the Illumina Hiseq 2500 platform generated a total of 45,101,102 (5,682,738,852 bases) and 34,715,846 (4,374,196,596 bases) raw reads, respectively (Table 1). After the cleansing step with an NGS QC Toolkit and removal of low quality (Q < 20) reads, 39,932,160 (5,006,434,933 bases) and 31,353,144 (3,932,946,324 bases) remained as clean reads, respectively (Table 1). The percentages of clean reads were 88.1% and 89.9%, respectively (Table 1). All the clean reads were submitted to the Trinity for de novo assembly. Unigenes were identified after removing redundant sequences from assembled transcripts. The number of unigenes was 104,348, the total length and the average length of the unigenes were 88,123,224 bp and 845 bp, respectively (Table 1). The length distribution of unigenes is presented in Figure 1. Among these unigenes, 66,204 unigenes (63.4%) were no more than 500 bp. A total of 15,382 unigenes (14.7%) were 501–1000 bp, 6991 unigenes (6.7%) were 1001–1500 bp, 4727 unigenes (4.5%) were 1501–2000 bp, 3332 unigenes (3.2%) were 2001–2500 bp, and 7712 unigenes (7.4%) were longer than 2500 bp.

3.2. The Most Abundantly Expressed Gene in the Transcriptome Profile

To estimate gene expression levels, we calculated the abundance of reads in the transcriptome. The top 20 most highly expressed transcripts are shown in Table 2. Commonly, the most abundant genes in both the mock and NNV-infected groups were ribosomal proteins, such as ribosomal protein (RPS) 15a, RPL39, RPS28, RPS14, RPLS2, RPS27a, RPL21 and RPL32 essential for biological metabolism. Ubiquitin-like protein 4a (UBL4a), C-C motif chemokine 2 (CCL2), lysozyme g (LYG_EPICO) and two novel genes (ID: SGU016297, SGU008676) were highly expressed in the NNV-infected group compared to that in the mock group. Of them, Ubiquitin-like protein 4a was the most abundant gene in the NNV-infected group.

3.3. Functional Annotations

Putative annotations of these transcripts were performed using BlastX as mentioned in the method section. After gene annotation by using BlastX against the non-redundant (nr) database, the putative functions of 43,280 sequences (41.5%) of 104,348 unigenes sequences were identified.

3.4. Immune Relevant DEGs Involved in NNV Infection

A total of 3418 unigenes were differentially expressed based on DEG analysis using the TCC package. A total of 372 genes from the total of 3418 DEGs were annotated (Table S1). Immune relevant DEGs were further manipulated manually (Table 3). In immune relevant genes, a variety of cytokines were intensely up-regulated after NNV infection.
Several cytokine genes induced by NNV infection belonged to the chemokine family, including C-C motif Chemokine Ligand 2 (CCL2), CCL34, CCL19, CCL4, C-X-C motif chemokine ligand 13 (CXCL13), CXCL6, CXCL8, CXCL9, Interleukin-12 subunit alpha (IL12A) and beta (IL12B), and IL18B. CCL2 was the most critically expressed gene in the infected group showing 10.66 Log Fold Change (FC). CCL2 is involved in neuroinflammatory processes taking place in the central nervous system in various diseases [28].
Cathepsins are lysosomal cysteine enzymes with important roles in cellular homeostasis and innate immune response [29]. Among a dozen members of the Cathepsin family, subtypes L, H, K, O, S and Z were up-regulated in the brain of sevenband grouper after NNV infection. Specifically, Cathepsin L was highly expressed in the NNV-infected group showing 8.3 Log FC.
Several lectins were expressed in higher levels in the NNV-infected group compared to the mock group, including C-type lectins (CLEC4M, CLEC10A), galectins (LGALS9, LGALS3), fucolectin (FUCL4), and mannose-binding lectin (MBL). In the case of C-type lectins, its receptor (CD209) was also highly expressed in the infected group (Table 3) indicating that C-type lectin might play specific roles in the response of sevenband grouper to NNV infection.
As expected, a number of antiviral proteins also showed high levels of expression in the NNV-infected group. For example, radical S-adenosyl methionin domain-containing protein 2 (RSAD2), also known as viperin, was highly expressed in the NNV-infected group with 10.40 Log FC. Mx gene (MX), one of the important downstream effectors of interferon (IFN), was also expressed more in the infected group with 8.64 Log FC. Besides Mx, a lot of IFN-induced proteins were upregulated by NNV infection, including IFN-induced protein 44 (IFI44), IFN-induced protein with tetratricopeptide repeats 5 (IFIT5), IFN-induced very large GTPase 1 (GVINP1), IFN-induced double-stranded RNA-activated protein kinase (EIF2Ak2), and IFN-induced helicase C domain-containing protein 1 (IFIH1). Interestingly, of the various heat shock proteins (HSPs), only HSP30 was significantly upregulated in the NNV-infected group with Log 8.42 FC. NK-Lysin, a known antibacterial protein, was also highly expressed in the NNV-infected group with 8.85 Log FC.

3.5. GO Enrichment of Differentially Expressed Genes

GO is a widely used method to classify gene functions and their products in organisms. Therefore, the identified DEGs were subsequently used for GO enrichment analysis. According to GO terms, 2094 (61.3%) of the total of 3418 DEGs were classified into the three categories of molecular function, biological process, and cellular component. “Binding” (1258 genes, 46.3%) was the major subcategory in the molecular function. The largest subcategory found in the biological process category was “cellular process” (1488 genes, 12.3%) while “Cell” (1687 genes, 19.6%) and “cell part” (1687 genes, 19.6%) were the most abundant GO terms in the cellular component category (Figure 2). Because one gene could be categorized into several subcategories, the sum of genes in the subcategories could exceed 100%. GO analysis of the transcriptome revealed nine molecular function subcategories, 62 biological process subcategories, and 12 cellular component subcategories with p value of less than 1 × 10−5) (Table S2).

4. Discussion

NNV infection has caused high mortalities of sevenband groupers in aqua-farms during the summer season, especially at larval and juvenile stages. It has also caused tremendous economic losses [1]. Due to the greater damage to the sevenband grouper industry, investigation on the molecular response of NNV infection is required to understand the outbreak mechanism of disease and develop prevention methods such as vaccines. In this study, we performed a transcriptome analysis of the brain tissue of sevenband grouper infected with NNV compared to mock brain tissue using a RNA-Seq.
The total number of unigenes and the average length of the unigenes were 104,348 and 845 bp, respectively. The number and average length found in this study indicated a fairly good performance compared to other previous NGS transcriptome studies on crimson spotted rainbowfish (107,749 transcripts, 961 bp) [30] common carp (130,292 transcripts, 1401 bp) [18], blunt snout bream (253,439 transcripts, 998 bp) [31], orange-spotted grouper (116,678 transcripts, 685 bp) [32] and Asian seabass (89,026 transcripts, 1175 bp) [33].
Gene annotation by BlastX provides valuable information about the transcripts. In this study, 43,289 unigenes (41.5%) of 104,348 unigenes were annotated. This is similar to the result of orange-spooted grouper (45.8%) [32]. Liu et al. have addressed the possible reasons of such poor annotation: (1) novel genes; (2) sequencing errors; and (3) artefacts by assembly algorithm [33]. Therefore, more genetic studies are needed to understand the biological functions.
The importance of innate defense mechanisms against viral infection has been extensively reviewed [34,35,36]. In this study, we identified innate immune response relevant genes of sevenband grouper involved in NNV infection. Chemokines are critical components of the immune system. The roles of chemokines and their receptors in viral interactions have been reported in various studies [37]. The chemokines family comprises four subfamilies based on N-terminal cystein-motifs: C, C-C, C-X-C, and C-X3-C subfamilies [38]. In this study, we also detected significant up-regulation of CCL2, CCL34, CCL19, CCL4, CXCL13, CXCL6, CXCL8, and CXCL9 in sevenband grouper brain tissue after NNV infection. Especially, CCL2 was highly over expressed at about 10.66 Log FC. CCL2 is a pro-inflammatory chemokine that is induced during several human acute and chronic viral infections, including human immunodeficiency virus (HIV) [39], hepatitis C virus [40], Epstein–Barr virus [41], respiratory synctitial virus [42], Severe Acute Respiratory Syndrome (SARS) [43], herpes simplex virus-1 [44], and Japanese encephalitis virus [45].
Cathepsins are lysosomal cysteines that play important roles in normal metabolism for the maintenance of cellular homeostasis. Cathepsins are one of the superfamilies involved in the regulation of antigen presentation and degradation as well as immune responses, including apoptosis, inflammation, and regulation of hormone processing [46,47,48]. In addition, Chandran et al. have shown that Cathepsin B and Cathepsin L are involved in Ebola virus infection [49]. They are involved in the entry of reovirus [50]. Recently, Cathepsin L has also been shown to be involved in the entry mediated by the SARS coronavirus spike glycoprotein [51] as well as in the process of fusion glycoprotein of Hendra virus [52]. In this study, Cathepsin L and Cathepsin S were found to be notably expressed after NNV infection. Their functional roles in the interaction between grouper and NNV merit further studies.
Lectins are carbohydrate-binding proteins that are highly specific for sugar moieties. They mediate the attachment and binding of viruses to their targets [53]. Lectins are also known to play important roles in the immune system. Within the innate immune system, lectins can help mediate the first-line of defense against invading microorganisms. In this study, several kinds of lectins were found to be highly induced in sevenband grouper brain tissue by NNV infection, such as C-type lectins (CTLs), galectins, fucolectin, and mannose-binding lectin. CTLs are the most well studied lectins. They can promote antibacterial and antiviral immune defense [54]. Many CTLs have been identified in teleost but the exact function of CTLs remains unclear.
Hundreds of interferon stimulated genes (ISGs) were transcribed in sevenband grouper brain tissue during NNV infection. Interferon induced protein 44 (IFI44) was expressed the most. IFI44 is an interferon-alpha inducible protein associated with infection of several viruses. Power et al. have demonstrated that IFI44 can inhibit HIV-1 replication in vitro by suppressing HIV-1 LTR promoter activity [55]. Carlton-Smith and Elliott have screened ISGs related to Bunyamwera orthbunyavirus replication using nonstructural (NSs) protein knock out virus. One of these ISGs that have inhibitory activity is found to be IFI44 [56]. Whether protein B2 of NNV has roles in virus replication and its relationship with ISGs such as IFI44 merit further study.
HSPs are one of the most phylogenetically conserved classes of proteins with critical roles in maintaining cellular homeostasis and protecting cells from various stresses [57]. Ironically, HSP70 has roles to suppress some virus infections, and support their replication in other viruses [58]. In this study, HSP30 was the highly induced gene instead of HSP70. HSP30 has also been reported to be the most induced gene in the NNV infected Asian seabass epithelial cell [33]. However, the function of HSP30 in NNV infection remains unclear.
Krasnov et al. previously reported the effects of NNV on gene expression in Atlantic cod brain using a microarray [59]. Compared to our study, a number of genes show a similar up-regulation result in the study, such as Caspase, Cathepsins, IRF, Radical S-adenosyl methionine domain-containing protein, tripartite motif-containing protein (TRIM) and so on. However, a lot of novel genes (e.g., NK-Lysin, Ubiquitin-like protein 4, Granzyme A, etc.) were identified from our RNA-Seq result because a microarray can only evaluate the genes on a chip.
Our findings are preliminary based on the small scale of the study and the results have not yet been confirmed by an independent technique such as quantitative polymerase chain reaction (qPCR). In future studies, it will be necessary to confirm the expression level of genes and to characterize the function of genes that are highly involved in NNV infection.

5. Conclusions

In conclusion, to the best of our knowledge, this is the first study reporting the transcriptome of brain tissue of NNV-infected sevenband grouper. In this study, we obtained the transcriptome of sevenband grouper. A total of 104,348 transcripts were obtained, including 628 DEGs between NNV infected and non-infected sevenband grouper. A large number of differential expressed genes relevant to immune response were identified as well as several candidate genes (CCL2, Cathepsins, Lectins, Hsp30, and Interferon-induced protein 44) that were intensely induced by NNV. Their functions in sevenband grouper against NNV infection merit further study. The acquired data from such transcriptome analysis provide valuable information for future functional genes related to NNV infection and VNN outbreak.

Supplementary Materials

The following are available online at www.mdpi.com/2073-4425/8/1/31/s1, Table S1: The annotated DEGs, Table S2: Gene ontology of sevenband grouper transcriptome after NNV infection.

Acknowledgments

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (NRF-2015R1C1A1A01054829).

Author Contributions

Jong-Oh Kim and Myung-Joo Oh conceived and designed the experiments; Jong-Oh Kim and Jae-Ok Kim performed the experiments; Jong-Oh Kim and Wi-Sik Kim analyzed the data; Jong-Oh Kim and Myung-Joo Oh wrote the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kim, C.S.; Kim, W.S.; Nishizawa, T.; Oh, M.J. Prevalence of viral nervous necrosis (VNN) in sevenband grouper, Epinephelus septemfasciatus farms. J. Fish Pathol. 2012, 25, 111–116. [Google Scholar] [CrossRef]
  2. Munday, B.L.; Kwang, J.; Moody, N. Betanodavirus infections of teleost fish: A review. J. Fish Dis. 2002, 25, 127–142. [Google Scholar] [CrossRef]
  3. Munday, B.L.; Nakai, T. Special topic review: Nodaviruses as pathogens in larval and juvenile marine finfish. World J. Microbiol. Biotechnol. 1997, 13, 375–381. [Google Scholar] [CrossRef]
  4. Muroga, K. Viral and bacterial diseases of marine fish and shellfish in Japanese hatcheries. Aquaculture 2001, 202, 23–44. [Google Scholar] [CrossRef]
  5. Cha, S.J.; Do, J.W.; Lee, N.S.; An, E.J.; Kim, Y.C.; Kim, J.W.; Park, J.W. Phylogenetic analysis of betanodaviruses isolated from cultured fish in Korea. Dis. Aquat. Org. 2007, 77, 181–189. [Google Scholar] [CrossRef] [PubMed]
  6. Oh, M.J.; Jung, S.J.; Kitamura, S.I. Comparison of the coat protein gene of nervous necrosis virus (NNV) detected from marine fishes in Korea. J. World Auqac. Soc. 2005, 36, 223–227. [Google Scholar] [CrossRef]
  7. Sohn, S.G.; Park, M.A.; Oh, M.J.; Chun, S.K. A fish nodavirus isolated from cultured sevenband grouper, Epinephelus septemfasciatus. J. Fish Pathol. 1998, 11, 97–104. [Google Scholar]
  8. Huang, Y.; Huang, X.; Yan, Y.; Cai, J.; Ouyang, Z.; Cui, H.; Wang, P.; Qin, Q. Transcriptome analysis of orange-spotted grouper (Epinephelus coioides) spleen in response to Singapore grouper iridovirus. BMC Genom. 2011, 12, 556. [Google Scholar] [CrossRef] [PubMed]
  9. Nishizawa, T.; Mori, K.; Furuhashi, M.; Nakai, T.; Furusawa, I.; Muroga, K. Comparison of the coat protein genes of five fish nodaviruses, the causative agents of viral nervous necrosis in marine fish. J. Gen. Virol. 1995, 76, 1563–1569. [Google Scholar] [CrossRef] [PubMed]
  10. Johansen, R.; Sommerset, I.; Torud, B.; Korsnes, K.; Hjortaas, M.J.; Nilsen, F.; Nerland, A.H.; Dannevig, B.H. Characterization of nodavirus and viral encephalophathy and retinopathy in farmed turbot, Scophthalumus maximus (L.). J. Fish Dis. 2004, 27, 591–601. [Google Scholar] [CrossRef] [PubMed]
  11. Jones, S.I.; Vodkin, L.O. Using RNA-Seq to profile soybean seed development from fertilization to maturity. PLoS ONE 2013, 8, e59270. [Google Scholar] [CrossRef] [PubMed]
  12. Tan, M.H.; Au, K.F.; Yablonovitch, A.L.; Wills, A.E.; Chuang, J.; Baker, J.C.; Wong, W.H.; Li, J.B. RNA sequencing reveals a diverse and dynamic repertoire of the Xenopus tropicalis transcriptome over development. Genome Res. 2013, 23, 201–216. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, H.; Fei, C.; Wu, H.; Yang, M.; Liu, Q.; Wang, Q.; Zhang, Y. Transcriptome profiling reveals Th17-like immune responses induced in zebrafish bath-vaccinated with a live attenuated Vibrio anguillarum. PLoS ONE 2013, 8, e73871. [Google Scholar] [CrossRef] [PubMed]
  14. Iwamoto, T.; Mise, K.; Takeda, A.; Okinaka, Y.; Mori, K.I.; Arimoto, M.; Okuno, T.; Nakai, T. Characterization of Striped jack nervous necrosis virus subgenomic RNA3 and biological activities of its encoded protein B2. J. Gen. Virol. 2005, 86, 2807–2816. [Google Scholar] [CrossRef] [PubMed]
  15. Mu, Y.; Li, M.; Ding, F.; Ding, Y.; Ao, J.; Hu, S.; Chen, X. De novo characterization of the spleen transcriptome of the large yellow croaker (Pseudosciaena crocea) and analysis of the immune relevant genes and pathways involved in the antiviral response. PLoS ONE 2014, 9, e97471. [Google Scholar] [CrossRef] [PubMed]
  16. Pereiro, P.; Balseiro, P.; Romero, A.; Dios, S.; Forn-Cuni, G.; Fuste, B.; Planas, J.V.; Beltran, S.; Nova, B.; Figueras, A. High- throughput sequence analysis of turbot (Scophthalmus maximus) transcriptome using 454-pyrosequencing for the discovery of antiviral immune genes. PLoS ONE 2012, 7, e35369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Xiang, L.X.; He, D.; Dong, W.R.; Zhang, Y.W.; Shao, J.Z. Deep sequencing- based transcriptome profiling analysis of bacteria-challenged Lateolabrax japonicus reveals insight into the immune-relevant genes in marine fish. BMC Genom. 2010, 11, 472. [Google Scholar] [CrossRef] [PubMed]
  18. Li, G.; Zhao, Y.; Liu, Z.; Gao, C.; Yan, F.; Liu, B.; Feng, J. De novo assembly and characterization of the spleen transcriptome of common carp (Cyprinus carpio) using Illumina paired-end sequencing. Fish Shellfish Immunol. 2015, 44, 420–429. [Google Scholar] [CrossRef] [PubMed]
  19. Kim, J.O.; Kim, J.O.; Kim, W.S.; Oh, M.J. Development and application of quantitative detection method for nervous necrosis virus (NNV) isolated from sevenband grouper Hyporthodus septemfasciatus. Asian Pac. J. Trop. Med. 2016, 9, 742–748. [Google Scholar] [CrossRef] [PubMed]
  20. Reed, L.J.; Muench, H. A simple method of estimating fifty percent end points. Am. J. Hyg. 1938, 27, 493–497. [Google Scholar]
  21. Andrews, S. FastQC a Quality Control Tool for High Throughput Sequence Data [Online]. 2010. Available online: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 3 March 2016).
  22. Martin, J.A.; Wang, Z. Next-generation transcriptome assembly. Nat. Rev. Genet. 2011, 12, 671–682. [Google Scholar] [CrossRef] [PubMed]
  23. Hass, B.J.; Papanicolaou, A.; Yassour, M.; Grabherr, M.; Blood, P.D.; Bowden, J.; Couger, M.B.; Eccles, D.; Li, B.; Lieber, M.; et al. De novo transcript sequence reconstruction from RNA-seq using the Trinity platform for reference generation and analysis. Nat. Protoc. 2013, 8, 1494–1512. [Google Scholar] [CrossRef] [PubMed]
  24. Fu, L.; Niu, B.; Zhu, Z.; Wu, S.; Li, W. CD-HIT: Accelerated for clustering the next-generation sequencing data. Bioinformatics 2012, 28, 3150–3152. [Google Scholar] [CrossRef] [PubMed]
  25. Li, B.; Dewey, C.N. RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011, 4, 323. [Google Scholar] [CrossRef] [PubMed]
  26. Sun, J.; Nishiyama, T.; Shimizu, K.; Kadota, K. TCC: An R package for comparing tag count data with robust normalization strategies. BMC Bioinform. 2013, 14, 219. [Google Scholar] [CrossRef] [PubMed]
  27. Kadota, K.; Nishiyama, T.; Shimizu, K. A normalization strategy for comparing tag count data. Algorithms Mol. Biol. 2012, 7, 5. [Google Scholar] [CrossRef] [PubMed]
  28. Gerard, C.; Rollins, B.J. Chemokines and disease. Nat. Immunol. 2001, 2, 108–115. [Google Scholar] [CrossRef] [PubMed]
  29. Choi, K.M.; Shim, S.H.; An, C.M.; Nam, B.H.; Kim, Y.O.; Kim, J.W.; Park, C.I. Cloning, characterisation, and expression analysis of the cathepsin D gene from rock bream (Oplegnathus fasciatus). Fish Shellfish Immunol. 2014, 40, 253–258. [Google Scholar] [CrossRef] [PubMed]
  30. Smith, S.; Bernatchez, L.; Beheregaray, L.B. RNA-seq analysis reveals extensive transcriptional plasticity to temperature stress in a freshwater fish species. BMC Genom. 2013, 14, 375. [Google Scholar] [CrossRef] [PubMed]
  31. Tran, N.T.; Gao, Z.X.; Zhao, H.H.; Yi, S.K.; Chen, B.X.; Zhao, Y.H.; Lin, L.; Liu, X.Q.; Wang, W.M. Transcriptome analysis and microsatellite discovery in the blunt snout bream (Megalobrama amblycephala) after challenge with Aeromonas hydrophila. Fish Shellfish Immunol. 2015, 45, 72–82. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, Y.D.; Huang, S.J.; Chou, H.N.; Liao, W.L.; Gong, H.Y.; Chen, J.Y. Transcriptome analysis of the effect of Vibrio alginolyticus infection on the innate immunity-related complement pathway in Epinephelus coioides. BMC Genom. 2014, 15, 1102. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, P.; Wang, L.; Kwang, J.; Yue, G.H.; Wong, S.M. Transcriptome analysis of genes responding to NNV infection in Asian seabass epithelial cells. Fish Shellfish Immunol. 2016, 54, 342–352. [Google Scholar] [CrossRef] [PubMed]
  34. Koyama, S.; Ishii, K.J.; Coban, C.; Akira, S. Innate immune response to viral infection. Cytokine 2008, 43, 336–341. [Google Scholar] [CrossRef] [PubMed]
  35. Takeuchi, O.; Akira, S. Innate immunity to virus infection. Immunol. Rev. 2009, 227, 75–86. [Google Scholar] [CrossRef] [PubMed]
  36. Thompson, M.R.; Kaminski, J.J.; Kurt-Jones, E.A.; Fitzgerald, K.A. Pattern Recognition Receptors and the Innate Immune Response to Viral Infection. Viruses 2011, 3, 920–940. [Google Scholar] [CrossRef] [PubMed]
  37. Abdul, W.A.; Heiken, H.; Meyer-Olson, D.; Schmidt, R.E. CCL2: A potential prognostic marker and target of anti-inflammatory strategy in HIV/AIDS pathogenesis. Eur. J. Immunol. 2011, 41, 3412–3418. [Google Scholar]
  38. Rollins, B.J. Chemokines. Blood 1997, 90, 909–928. [Google Scholar] [PubMed]
  39. Weiss, J.M.; Nath, A.; Major, E.O.; Berman, J.W. HIV-1 Tat induces monocyte chemoattractant protein-1-mediated monocyte transmigration across a model of the human blood-brain barrier and up-regulates CCR5 expression on human monocytes. J. Immunol. 1999, 163, 2953–2959. [Google Scholar] [PubMed]
  40. Fisher, N.C.; Neil, D.A.; Williams, A.; Adams, D.H. Serum concentrations and peripheral secretion of the beta chemokines monocyte chemoattractant protein 1 and macrophage inflammatory protein 1α in alcoholic liver disease. Gut 1999, 45, 416–420. [Google Scholar] [CrossRef] [PubMed]
  41. Gaudreault, E.; Fiola, S.; Olivier, M.; Gosselin, J. Epstein-Barr virus induces MCP-1 secretion by human monocytes via TLR2. J. Virol. 2007, 81, 8016–8024. [Google Scholar] [CrossRef] [PubMed]
  42. Culley, F.J.; Pennycook, A.M.; Tregoning, J.S.; Hussell, T.; Openshaw, P.J. Differential chemokine expression following respiratory virus infection reflects Th1- or Th2-biased immunopathology. J. Virol. 2006, 80, 4521–4527. [Google Scholar] [CrossRef] [PubMed]
  43. Yen, Y.T.; Liao, F.; Hsiao, C.H.; Kao, C.L.; Chen, Y.C.; Wu-Hsieh, B.A. Modeling the early events of severe acute respiratory syndrome coronavirus infection in vitro. J. Virol. 2006, 80, 2684–2693. [Google Scholar] [CrossRef] [PubMed]
  44. Kurt-Jones, E.A.; Chan, M.; Zhou, S.; Wang, J.; Reed, G.; Bronson, R.; Arnold, M.; Knipe, D.M.; Finberg, R.W. Herpes simplex virus 1 interaction with Toll-like receptor 2 contributes to lethal encephalitis. Proc. Natl. Acad. Sci. USA 2004, 101, 1315–1320. [Google Scholar] [CrossRef] [PubMed]
  45. Mishra, M.K.; Kumawat, K.L.; Basu, A. Japanese encephalitis virus differentially modulates the induction of multiple pro-inflammatory mediators in human astrocytoma and astroglioma cell-lines. Cell Biol. Int. 2008, 32, 1506–1513. [Google Scholar] [CrossRef] [PubMed]
  46. Dixit, A.K.; Dixit, P.; Sharma, R.L. Sharma Immunodiagnostic/protective role of Cathepsin L cysteine proteinases secreted by Fasciolaspecies. Vet. Parasitol. 2008, 154, 177–184. [Google Scholar] [CrossRef] [PubMed]
  47. Hsing, L.C.; Rudensky, A.Y. The lysosomal cysteine proteases in MHC class II antigen presentation. Immunol. Rev. 2005, 207, 229–241. [Google Scholar] [CrossRef] [PubMed]
  48. Yasothornsrikul, S.; Greenbaum, D.; Medzihradszky, K.F.; Toneff, T.; Bundey, R.; Miller, R.; Schilling, B.; Petermann, I.; Dehnert, J.; Logvinova, A.; et al. Cathepsin L in secretory vesicles functions as a prohormone-processing enzyme for production of the enkephalin peptide neurotransmitter. Proc. Natl. Acad. Sci. USA 2003, 100, 9590–9595. [Google Scholar] [CrossRef] [PubMed]
  49. Chandran, K.; Sullivan, N.J.; Felbor, U.; Whelan, S.P.; Cunningham, J.M. Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection. Science 2005, 308, 1643–1645. [Google Scholar] [CrossRef] [PubMed]
  50. Ebert, D.H.; Deussing, J.; Peters, C.; Dermody, T.S. Cathepsin L and cathepsin B mediate reovirus disassembly in murine fibroblast cells. J. Biol. Chem. 2002, 277, 24609–24617. [Google Scholar] [CrossRef] [PubMed]
  51. Simmons, G.; Gosalia, D.N.; Rennekamp, A.J.; Reeves, J.D.; Diamond, S.L.; Bates, P. Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. USA 2005, 102, 11876–11881. [Google Scholar] [CrossRef] [PubMed]
  52. Pager, C.T.; Dutch, R.E. Cathepsin L is involved in proteolytic processing of the Hendra virus fusion protein. J. Virol. 2005, 79, 12714–12720. [Google Scholar] [CrossRef] [PubMed]
  53. Bartenschlager, R.; Sparacio, S. Hepatitis C virus molecular clones and their replication capacity in vivo and in cell culture. Virus Res. 2007, 127, 195–207. [Google Scholar] [CrossRef] [PubMed]
  54. Zhou, Z.J.; Sun, L. CsCTL1, a teleost C-type lectin that promotes antibacterial and antiviral immune defense in a manner that depends on the conserved EPN motif. Dev. Comp. Immunol. 2015, 50, 69–77. [Google Scholar] [CrossRef] [PubMed]
  55. Power, D.; Santoso, N.; Dieringer, M.; Yu, J.; Huang, H.; Simpson, S.; Seth, I.; Miao, H.; Zhu, J. IFI44 suppresses HIV-1 LTR promoter activity and facilitates its latency. Virology 2015, 481, 142–150. [Google Scholar] [CrossRef] [PubMed]
  56. Carlton-Smith, C.; Elliott, R.M. Viperin, MTAP44, and protein kinase R contribute to the interferon-induced inhibition of Bunyamwera Orthobunyavirus replication. J. Virol. 2012, 86, 11548–11557. [Google Scholar] [CrossRef] [PubMed]
  57. Hartl, F.U. Molecular chaperones in cellular protein folding. Nature 1996, 381, 571–579. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, M.Y.; Oglesbee, M. Virus-heat shock protein interaction and a novel axis for innate antiviral immunity. Cells 2012, 1, 646–666. [Google Scholar] [CrossRef] [PubMed]
  59. Krasnov, A.; Kileng, O.; Skugor, S.; Jorgensena, S.M.; Afanasyev, S.; Timmerhaus, G.; Sommer, A.I.; Jensen, I. Genomic analysis of the host response to nervous necrosis virus in Atlantic cod (Gadus morhua) brain. Mol. Immunol. 2013, 54, 443–452. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Length distribution of unigenes obtained from transcriptome analysis.
Figure 1. Length distribution of unigenes obtained from transcriptome analysis.
Genes 08 00031 g001
Figure 2. GO annotation of Differentially Expressed Genes (DEGs).
Figure 2. GO annotation of Differentially Expressed Genes (DEGs).
Genes 08 00031 g002
Table 1. Sequencing, assembly and annotation of transcriptome.
Table 1. Sequencing, assembly and annotation of transcriptome.
A. Sequencing and Preprocessing
Sample TypeNot Infected (Mock)NNV Infected
Number of raw reads45,101,10234,715,846
Total number of raw bases5,682,738,8524,374,196,596
Number of clean reads39,932,160 (88.5%)31,353,144 (90.3%)
Total number of clean bases5,006,434,933 (88.1%)3,932,946,324 (89.9%)
B. De novo Assembly and Annotation
Number of unigenes 104,348
Total bases 88,123,224
Average length of unigenes 845 bases
Annotation by BLAST 43,280 (41.5%)
Table 2. The top 20 most abundant unigenes.
Table 2. The top 20 most abundant unigenes.
NoMockInfected
IDNameDescriptionIDNameDescription
1SGU067144-Hypothetical proteinSGU020577UBL4aUbiquitin-like protein 4a
2SGU067145-Hypothetical proteinSGU005369-Hyphothetical protein
3SGU004764EPD1Ependymin-1SGU067144-Hyphothetical protein
4SGU023233MT-CO1Cytochrome c oxidase subunit 1SGU067145-Hyphothetical protein
5SGU005369-Hypothetical proteinSGU051992RPS15a40S ribosomal protein S15a
6SGU023234MT-ND5NADH dehydrogenase subunit 5SGU028675RPL3860S ribosomal protein L38
7SGU051992RPS15a40S ribosomal protein S15aSGU004764EPD1Ependymin-1
8SGU028388CD59CD59 glycoproteinSGU035083RPL3960S ribosomal protein L39
9SGU025576HBAHemoglobin subunit alphaSGU023233MT-CO1Cytochrome c oxidase subunit 1
10SGU035083RPL3960S ribosomal protein L39SGU036307CCL2C-C motif chemokine 2
11SGU021087UBIQP_XENLAPolyubiquitinSGU053409LYG_EPICOLysozyme g
12SGU002473RPS2840S ribosomal protein S28SGU002473RPS2840S ribosomal protein S28
13SGU002418FABP7Fatty acid-binding protein, brainSGU021087UBIQP_XENLAPolyubiquitin
14SGU028675RPL3860S ribosomal protein L38SGU037323RPL2960S ribosomal protein L29
15SGU040127RPS1440S ribosomal protein S14SGU003425RPS27a40S ribosomal protein S27a
16SGU028033RPLP260S acidic ribosomal protein P2SGU016297--
17SGU026642HBB2Hemoglobin subunit beta-2SGU005492UBA52Ubiquitin-60S ribosomal protein L40
18SGU003425RPS27a40S ribosomal protein S27aSGU034075RPL2160S ribosomal protein L21
19SGU034075RPL2160S ribosomal protein L21SGU040127RPS1440S ribosomal protein S14
20SGU003965RPL3260S ribosomal protein L32SGU008676--
Table 3. Immune relevant differentially expressed genes (DEGs) after NNV infection.
Table 3. Immune relevant differentially expressed genes (DEGs) after NNV infection.
NameDescriptionExpression Level (FPKM)Log FCp-ValueFDR
MockInfected
Cytokine
CCL2C-C Motif Chemokine 251.783,844.210.662.3 × 10−6.6.0 × 10−3
IL12BInterleukin-12 Subunit Beta12.514,158.010.155.6 × 10−67.1 × 10−3
CCL34A.4Chemokine (C-C Motif) Ligand 34a, Duplicate 45.87038.810.246.2 × 10−67.4 × 10−3
CXCL13C-X-C Motif Chemokine Ligand 1310.07500.39.551.4 × 10−51.3 × 10−2
CCL19C-C Motif Chemokine 1915.04523.88.248.2 × 10−53.0 × 10−2
CXCL6C-X-C Motif Chemokine Ligand 660.811,405.27.551.8 × 10−45.3 × 10−2
IL18R1Interleukin-18 Receptor 19.21814.27.632.3 × 10−46.0 × 10−2
CXCL8C-X-C Motif Chemokine Ligand 8 (Interleukin-8)6.71307.07.612.7 × 10−46.4 × 10−2
EBI3Interleukin-27 Subunit Beta9.21506.87.363.4 × 10−47.5 × 10−2
CXCL9C-X-C Motif Chemokine Ligand 912.51438.86.856.4 × 10−41.2 × 10−1
CCL4C-C Motif Chemokine 411.71328.86.836.7 × 10−41.2 × 10−1
IL1R2Interleukin-1 Receptor Type 26.7458.66.102.7 × 10−32.9× 10−1
CXCR4C-X-C Chemokine Receptor Type 435.01016.34.868.4 × 10−36.1 × 10−1
CRLF1Cytokine Receptor-like Factor 19.2326.35.159.1 × 10−36.5× 10−1
XCR1Chemokine Xc Receptor 10.897.56.881.0 × 10−27.0 × 10−1
IL13RA1AIl-13 Receptor-alpha-1-a Precursor117.52426.34.371.4 × 10−28.6 × 10−1
CXCR3C-X-C Chemokine Receptor Type 356.71140.04.331.6 × 10−29.3 × 10−1
IL12AInterleukin-12 Subunit Alpha3.5128.35.201.8 × 10−29.9 × 10−1
Cathepsin
CTSLCathepsin L7.52370.08.309.2 × 10−53.2 × 10−2
CTSHCathepsin H418.39396.34.491.1 × 10−27.5 × 10−1
CTSKCathepsin K170.08802.55.692.3 × 10−32.7 × 10−1
CTSOCathepsin O20.0485.04.601.4 × 10−28.6 × 10−1
CTSSCathepsin S388.320,089.45.692.2 × 10−32.7 × 10−1
CTSZCathepsin Z604.210,606.84.131.8 × 10−29.8 × 10−1
Cluster of differentiation
CD274Programmed Cell Death 1 Ligand 15.82255.08.606.7 × 10−52.8 × 10−2
CD4T-cell Surface Glycoprotein CD43.3256.96.273.7 × 10−33.6 × 10−1
CD209ACD209 Antigen-like Protein A3.3251.66.243.9 × 10−33.8 × 10−1
CD48CD48 Antigen30.81969.66.001.7 × 10−32.3 × 10−1
CD209DCD209 Antigen-like Protein D68.33346.35.612.7 × 10−33.0 × 10−1
TSPAN6Tetraspanin-69.7387.85.327.2 × 10−35.6 × 10−1
Complement
C4AComplement C4-a9.62062.47.742.0 × 10−45.6 × 10−2
C1QAComplement C1q Subcomponent Subunit A74.25517.86.221.1 × 10−31.8 × 10−1
C4Complement C41.2220.17.531.3 × 10−31.9 × 10−1
C1QCComplement C1q Subcomponent Subunit C61.74210.06.091.4 × 10−32.0 × 10−1
C1SComplement C1s Subcomponent126.48272.56.031.4 × 10−32.0 × 10−1
C1QBComplement C1q Subcomponent Subunit B116.77135.25.931.7 × 10−32.2 × 10−1
C7Complement Component C730.01288.25.423.9 × 10−33.8 × 10−1
CFBComplement Factor B144.24880.05.085.3 × 10−34.6 × 10−1
Lectin
CLEC4MC-type Lectin Domain Family 4 Member M10.05250.09.042.9 × 10−51.9 × 10−2
LGALS9Galectin-989.28098.86.507.6 × 10−41.3 × 10−1
FUCL4_ANGJAFucolectin-432.51838.85.822.2 × 10−32.7 × 10−1
CLEC10AC-type Lectin Domain Family 10 Member A12.5603.85.594.1 × 10−33.9 × 10−1
MBLMannose-binding Lectin1.7175.06.714.2× 10−34.0 × 10−1
LGALS3Galectin-310.8415.05.267.1× 10−35.5 × 10−1
LGALS3BPAGalectin-3-binding Protein A1083.327,214.64.658.9 × 10−36.5 × 10−1
Ubiquitination
UBL4AUbiquitin-like Protein 4a31.787,332.811.438.0 × 10−76.0 × 10−3
HERC5E3 Ubiquitin-protein Ligase Herc53.14851.110.615.6 × 10−67.1 × 10−3
HERC6E3 Ubiquitin-protein Ligase Herc665.034,045.89.032.2 × 10−51.6 × 10−2
USP18Ubiquitin Carboxyl-terminal Hydrolase 1814.24250.98.238.4 × 10−53.0 × 10−2
USP12Ubiquitin Carboxyl-terminal Hydrolase 129.21986.27.761.9 × 10−45.4 × 10−2
UBR1E3 Ubiquitin-protein Ligase Ubr118.42933.87.322.9 × 10−46.6 × 10−2
TRIM21Tripartite Motif-containing Protein 216.71061.37.314.2 × 10−48.7 × 10−2
TRIM47Tripartite Motif-containing Protein 479.21332.57.184.4 × 10−48.8 × 10−2
RNF213E3 Ubiquitin-protein Ligase Rnf21340.84928.86.924.6 × 10−48.9 × 10−2
TRIM29Tripartite Motif-containing Protein 2958.02261.15.294.3 × 10−34.0 × 10−1
TRIM39Tripartite Motif-containing Protein 397.0354.25.674.7 × 10−34.2 × 10−1
TRIM25Tripartite Motif-containing Protein 252.5182.56.195.8 × 10−34.8 × 10−1
HERC4E3 Ubiquitin-protein Ligase Herc4788.617,971.64.511.1 × 10−27.3 × 10−1
TRIM16Tripartite Motif-containing Protein 1617.5476.74.771.2 × 10−27.7 × 10−1
TRIM14Tripartite Motif-containing Protein 1485.71728.94.331.5 × 10−28.9 × 10−1
Others
RSAD2Radical S-adenosyl Methionine Domain-containing Protein 230.841,672.610.403.4 × 10−67.1 × 10−3
IFI44Interferon-induced Protein 4462.873,980.210.204.3 × 10−67.1 × 10−3
IFIT5Interferon-induced Protein With Tetratricopeptide Repeats 54.23119.89.552.0 × 10−51.6 × 10−2
SOCS1Suppressor Of Cytokine Signaling 125.813,362.69.012.5 × 10−51.7 × 10−2
MXInterferon-induced GTP-binding Protein Mx49.219,664.48.643.9 × 10−52.3 × 10−2
NKLAntimicrobial Peptide Nk-lysin8.33838.88.854.0 × 10−52.3 × 10−2
FCGR1AHigh Affinity Immunoglobulin Gamma Fc Receptor I33.312,632.28.574.5 × 10−52.5 × 10−2
FCER1AHigh Affinity Immunoglobulin Epsilon Receptor Subunit Alpha13.45255.28.624.8 × 10−52.5 × 10−2
PIGRPolymeric Immunoglobulin Receptor5.02332.58.874.9 × 10−52.5 × 10−2
LYG_EPICOLysozyme G352.5120,945.08.425.0 × 10−52.5 × 10−2
SAMD9Sterile Alpha Motif Domain-containing Protein 92.51271.28.996.5 × 10−52.8 × 10−2
IRF4Interferon Regulatory Factor 410.02811.38.141.1 × 10−43.6 × 10−2
GVINP1Interferon-induced Very Large GTPase 153.311,420.67.741.4 × 10−44.5 × 10−2
TMEM173Stimulator of Interferon Genes Protein23.34917.57.721.6 × 10−44.8 × 10−2
MPEG1Macrophage-expressed Gene 1 Protein13.32433.87.512.4 × 10−46.0 × 10−2
HSP30Heat Shock Protein 301.7572.58.422.4 × 10−46.0 × 10−2
GZMAGranzyme A9.21761.47.572.4 × 10−46.0 × 10−2
IRF3Interferon Regulatory Factor 35.81155.67.632.8 × 10−46.6 × 10−2
EIF2AK2Interferon-induced, Double-stranded RNA-activated Protein Kinase15.62438.57.293.3 × 10−47.3 × 10−2
IRF1Interferon Regulatory Factor 1349.222,350.86.001.5 × 10−32.0 × 10−1
PSMB6L-BProteasome Subunit Beta Type-6-b Like Protein131.78506.16.011.5× 10−32.0 × 10−1
CASP3Caspase-324.21688.86.131.5 × 10−32.1 × 10−1
PSME1Proteasome Activator Complex Subunit 1220.812,675.85.841.8 × 10−32.4 × 10−1
IFIH1Interferon-induced Helicase C Domain-containing Protein 196.75280.55.772.1 × 10−32.6 × 10−1
PSMB8Proteasome Subunit Beta Type-886.73997.55.533.0 × 10−33.2 × 10−1
GRNGranulins275.011,720.05.413.3 × 10−33.3 × 10−1
MR1Major Histocompatibility Complex Class I-related Gene Protein153.66162.85.333.8 × 10−33.6 × 10−1
SOCS3Suppressor Of Cytokine Signaling 3125.74607.55.204.6 × 10−34.2 × 10−1
IGSF3Immunoglobulin Superfamily Member 311.7408.05.138.3 × 10−36.1 × 10−1
IRGCInterferon-inducible GTPase 511.9346.94.861.2 × 10−27.7 × 10−1
Note, FPKM: fragments per kilobase of transcript per million mapped reads; Log FC: Log value of fold changes, FDR: false discovery rate.

Share and Cite

MDPI and ACS Style

Kim, J.-O.; Kim, J.-O.; Kim, W.-S.; Oh, M.-J. Characterization of the Transcriptome and Gene Expression of Brain Tissue in Sevenband Grouper (Hyporthodus septemfasciatus) in Response to NNV Infection. Genes 2017, 8, 31. https://doi.org/10.3390/genes8010031

AMA Style

Kim J-O, Kim J-O, Kim W-S, Oh M-J. Characterization of the Transcriptome and Gene Expression of Brain Tissue in Sevenband Grouper (Hyporthodus septemfasciatus) in Response to NNV Infection. Genes. 2017; 8(1):31. https://doi.org/10.3390/genes8010031

Chicago/Turabian Style

Kim, Jong-Oh, Jae-Ok Kim, Wi-Sik Kim, and Myung-Joo Oh. 2017. "Characterization of the Transcriptome and Gene Expression of Brain Tissue in Sevenband Grouper (Hyporthodus septemfasciatus) in Response to NNV Infection" Genes 8, no. 1: 31. https://doi.org/10.3390/genes8010031

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop