Next Article in Journal
Microecological Shifts in the Rhizosphere of Perennial Large Trees and Seedlings in Continuous Cropping of Poplar
Next Article in Special Issue
A Review: Microbes and Their Effect on Growth Performance of Litopenaeus vannamei (White Leg Shrimps) during Culture in Biofloc Technology System
Previous Article in Journal
Genetic Engineering of Agrobacterium Increases Curdlan Production through Increased Expression of the crdASC Genes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptome Profiling of Oncorhynchus mykiss Infected with Low or Highly Pathogenic Viral Hemorrhagic Septicemia Virus (VHSV)

Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, PD, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2024, 12(1), 57; https://doi.org/10.3390/microorganisms12010057
Submission received: 28 November 2023 / Revised: 20 December 2023 / Accepted: 22 December 2023 / Published: 28 December 2023
(This article belongs to the Special Issue Microbial Ecology and Sustainable Aquaculture)

Abstract

:
The rainbow trout (Oncorhynchus mykiss) is the most important produced species in freshwater within the European Union, usually reared in intensive farming systems. This species is highly susceptible to viral hemorrhagic septicemia (VHS), a severe systemic disease widespread globally throughout the world. Viral hemorrhagic septicemia virus (VHSV) is the etiological agent and, recently, three classes of VHSV virulence (high, moderate, and low) have been proposed based on the mortality rates, which are strictly dependent on the viral strain. The molecular mechanisms that regulate VHSV virulence and the stimulated gene responses in the host during infection are not completely unveiled. While some preliminary transcriptomic studies have been reported in other fish species, to date there are no publications on rainbow trout. Herein, we report the first time-course RNA sequencing analysis on rainbow trout juveniles experimentally infected with high and low VHSV pathogenic Italian strains. Transcriptome analysis was performed on head kidney samples collected at different time points (1, 2, and 5 days post infection). A large set of notable genes were found to be differentially expressed (DEGs) in all the challenged groups (e.s. trim63a, acod1, cox-2, skia, hipk1, cx35.4, ins, mtnr1a, tlr3, tlr7, mda5, lgp2). Moreover, the number of DEGs progressively increased especially during time with a greater amount found in the group infected with the high VHSV virulent strain. The gene ontology (GO) enrichment analysis highlighted that functions related to inflammation were modulated in rainbow trout during the first days of VHSV infection, regardless of the pathogenicity of the strain. While some functions showed slight differences in enrichments between the two infected groups, others appeared more exclusively modulated in the group challenged with the highly pathogenic strain.

1. Introduction

Viral hemorrhagic septicemia (VHS) is one of the WOAH-listed diseases responsible for a serious systemic disease in fish. It is caused by a bullet-shaped, enveloped, negative sense single stranded RNA virus belonging to the family Rhabdoviridae, genus Novirhabdovirus [1].
Viral hemorrhagic septicemia virus (VHSV) genome consists of six genes, ordered 3′-N-P-M-G-NV-L-5′, which encode five structural proteins with conserved functions among Rhabdoviruses [2,3]. The major structural protein is the nucleoprotein (N) that, together with the phosphoprotein (P) and the viral polymerase (L), forms the ribonucleoprotein complex (RNP) responsible for the transcription and replication of the viral genome. Finally, the matrix protein (M) spirals the large RNP complex [4]. The external glycoprotein (G) is the main antigenic protein of VHSV that allows the virus attachment and entry [5,6]. The non-virion protein (NV) is exclusive to the Novirhabdovirus genus and completes the protein asset of VHSV. This peculiar non-structural protein has been shown to influence the in vivo pathogenicity of the different VHSV [7,8].
Among the several species susceptible to the infection, the rainbow trout (Oncorhynchus mykiss) is one of the most severely affected by VHS, with a high level of mortality especially in intensive farming system. The disease is present all over the world, especially in the Northern hemisphere, and in Europe it is endemic in several countries, Italy included [9,10] The phylogenetic studies undertaken so far have described four genotypes (I, II, III, and IV), further divided in several subtypes and clades [11,12,13,14]. In Italy, the disease caused by the VHSV is widespread amongst rainbow trout grow on farms and is responsible for severe economic losses due to fish mortality and commercial constraints [9,15].
It has been demonstrated that virulence in VHSV can mainly depend on the viral genetic makeup [16,17,18]. Indeed, Panzarin et al. [16] have identified an extensive list of polymorphic sites (SAPs), scattered throughout VHSV coding regions, obtained using statistical association analyses that combined in vivo virulence records and associated whole genome sequences of known VHSV genotypes. Reported SAPs varied significantly with virulence and, as such, potentially associated with the high and low virulent phenotype. Interestingly, the listed SAPs also included virulence markers located in the N and P protein coding regions, recently suggested to be major virulence determinants for VHSV in rainbow trout [17,19,20]. Moreover, the existence of three different virulence categories of VHSV for rainbow trout, namely low, moderate, and highly virulence, have also been described [16]. Consequently, from this latter study we have selected two VHSV isolates, with distinct virulence classifications, in order to perform the herein presented experiment. Furthermore, in vitro studies suggest that virulence can mainly be determined by the capacity of the virus to penetrate the host and spread to target organs, and is independent from replication fitness during the infectious process [21].
Even if the obtained data highlight that VHSV virulence has strong genetic bases, we cannot exclude that the host response to the virus may play a crucial role in determining the outcome of the infection. The aim of the present study is to unravel this aspect, which deserves to be investigated further. In particular, our work wanted to investigate whether rainbow trout transcriptome can vary over time in response to the infection with VHSV strains belonging to two different virulence classes (namely highly and low virulence), as very little information is currently available in the literature on this matter [22,23,24].

2. Materials and Methods

2.1. VHSV Strains, Isolation and Propagation

Two VHSVs, namely VHSV/O.mykiss/I/TN/80/Mar10 and VHSV/O.mykiss/I/TN/480/Oct96 (GenBank accession numbers MK829682 and MK829677, respectively), stored at −80 °C at the Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe) repository, were propagated in epithelioma papulosum cyprini (EPC) cell monolayers for 24 h, according to standard procedures [16]. For each strain, the replication was performed in 75 cm2 flasks (CytoOne®, STARLAB International GmbH, Hamburg, Germany) at 15 °C and the cell culture supernatant was collected at the appearance of the complete cytopathic effect, then pooled and clarified for 10 min at 2800× g at 4 °C. The RNA was subsequently extracted as described in Section 2.3 and qualitatively checked with TaqMan® reverse transcription real-time PCR (rRT-PCR) for the presence of VHSV as described by Jonstrup et al. [25]; the details of the conditions are summarized in Tables S1 and S2. Furthermore, the same samples were also tested for the absence of infectious hematopoietic necrosis virus (IHNV) with methodology and oligonucleotides described by Overturf et al. [26] and infectious pancreatic necrosis virus (IPNV) according to Ørpetveit et al. [27]. Viral titres of cell culture supernatant were calculated and expressed as TCID50/mL according to Reed and Müench’s formula [28]. Viral batches were aliquoted and stored at −80 °C until use.
Both the VHS viruses used for the present study were isolated from Italian diseased rainbow trout (Oncorhynchus mykiss). Their phenotypes were characterized in a previous work as being highly virulent for VHSV/O.mykiss/I/TN/80/Mar10 and moderate/low virulent for VHSV/O.mykiss/I/TN/480/Oct96 [16]. From now on, we will refer to these viruses as VHSV-H and VHSV-L, respectively, for VHSV/O.mykiss/I/TN/80/Mar10 and VHSV/O.mykiss/I/TN/480/Oct96. These two viruses, together with several others, were also characterized for virulence determinant and intracellular replication by reverse genetics and in vitro studies, respectively [21,29].

2.2. Mortality and Transcriptomic Experimental Challenges

Female rainbow trout juveniles (n = 330) of approximately 13 ± 1 g were purchased from an Italian commercial farm classified within Category I (EU Directive 88/2006), acclimated for a period of 10 days, and tested for main pathogens. During this period, the presence of external parasites was excluded by gills and skin fresh smears observations. Main pathogenic bacteria, such as Yersinia Rukerii, Lactococcus garvie, and Aeromonas salmonicidae, were tested by plating on blood agar according to standard procedures samples from kidney and spleen of ten randomly selected fish. Viruses (infectious pancreatic necrosis, salmonid alphavirus, infectious hematopoietic necrosis, and VHS) were tested by rRT-PCR on internal organs pools (kidney, spleen, and heart) according to already published methods [25,26,27,30]. A commercial feed was supplied for the entire duration of the challenges. Fish were divided in six groups into individual tanks: 3 groups of 55 animals each (for mortality recording) and 3 groups of 25 animals each (for transcriptomic analyses). Groups for mortality and transcriptomic study were paired and challenged by immersion as follows: (i) two groups with VHSV-H, (ii) two groups with VHSV-L, and (iii) two mock-infected groups used for comparison with the infected fish. Exposure with 103.05 TCID50/mL of each viral strain was performed for 3 h in 20 L static water, supplying additional aeration. At the end of the challenge, the level of the water was restored to 80 L by adding clean freshwater and the water flow was turned on. For each viral strain, the first group (mortality recording) was monitored regularly for a 4-week period and daily mortality was recorded in order to elaborate survival curves. Spleen (SPL) and head kidney (HK) were collected both from the dead and survivor fish, euthanized by anaesthetic overdose (Tricaine, Pharmaq, Overhalla, Norway) at the end of the observation period. In the second group (transcriptomic challenge), five fish per tank were euthanized by anaesthetic overdose for SPL and HK collection in order to quantify viral genomic copy number (at 0.5, 1, 2, 3, and 5 days post infection (dpi)) and to perform transcriptomic analyses (at 1, 2, and 5 dpi). The third group of both challenges was mock infected with sterile E- Minimum Essential Medium (E-MEM) (Merck KGaA, Darmstadt, Germany) and the same sampling scheme applied to the mortality and transcriptomic groups was adopted.

2.3. RNA Extraction, Quality Check and Normalization

Aliquots of the viral batches produced for experimental trials and the organs (SPL and HK pooled for each analyzed fish) of the mortality challenge group were tested for VHSV detection. Total nucleic acids were purified starting directly from 250 μL of viral isolate, while the organs were first subjected to a homogenization step. The specimens were manually grinded in vials using a sterile small potter and quartz sand adding E-MEM to obtain a 1:3 dilution. Homogenized samples were centrifuged at 8000× g for 2 min and the clarified supernatants collected. Total nucleic acids were extracted using QIAsymphony DSP Virus/Pathogen Midi kit (Qiagen, Hilden, Germany) in combination with the automated system QIAsymphony SP (Qiagen, Hilden, Germany). Isolation of the nucleic acids was performed following the manufacturer’s recommendations. The RNAs were used undiluted to perform qualitative real time PCR for VHSV diagnosis in order to confirm the cause of mortality, as described below (See Section 2.4).
In the transcriptomic challenge, total RNA was isolated from the spleen and head kidney of each rainbow trout specimen using an RNeasy Mini kit (QIAGEN, Hilden, Germany). The purification protocol was followed according to the manufacturer’s instructions, as described in Toffan et al. [31]. The quantity and quality of all the isolated RNA samples were checked, respectively, using a Qubit® RNA Broad Range (BR) Assay kit (ThermoFisher Scientific, Waltham, MA, USA) and an RNA 6000 Nano kit in Agilent 2100 Bioanalyzer (Agilent Technology, Santa Clara, CA, USA). All the samples with RNA integrity number (RIN) > 9.00 were normalized at 20 ng/µL and subjected to molecular and transcriptomic analysis.

2.4. VHSV Detection by Real-Time RT-PCR

In order to confirm the success of infection and determine the cause of death in the mortality recording group, all dead specimens collected during the observation period (n = 53 in VHSV-H group; n = 2 in VHSV-L group) were tested for the presence of VHSV by a qualitative real-time RT-PCR (rRT-PCR) [25]; for details see Tables S1 and S2. Viral positivity was also investigated in survivor fish (n = 2 in VHSV-H group; n = 10/53 VHSV-L; n = 10/55 in mock infected group). All fish samples were analyzed singularly.

2.5. In Vitro VHSV RNA Synthesis for Standards Preparation

Reference strain DK-F1 [11] (GenBank accession number MT162452) was used to produce synthetic RNA. The partial nucleocapsid gene was amplified using the Qiagen® OneStep RT-PCR Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions, while the primer set used for the RT-PCR is reported in Table S1. In order to allow subsequent RNA in vitro transcription, T7 RNA polymerase promoter sequence was added at the 5′ end of the forward primer. The RT-PCR was performed as described in Table S2. The PCR product of 470 bp was analyzed for size and purity on 1% agarose gel electrophoresis. Amplicon was then excised from the agarose gel and purified using the QIAquick Gel Extraction Kit (Qiagen, Hilden, Germany), according to the manufacturer’s protocol. In vitro transcription reaction was carried out at 37 °C for 4 h using the MEGAshortscript™ T7 Transcription Kit (ThermoFisher Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. The reaction was finished by incubation for 15 min at 37 °C with 1 μL of TURBO DNase. VHSV synthetic RNA was finally purified with the MEGAclear™ Transcription Clean-Up Kit (ThermoFisher Scientific, Waltham, MA, USA) following the manufacturer’s recommendations. The obtained in vitro transcribed RNA (size 453 nt) was therefore quantitated with the Qubit® RNA BR Assay kit (ThermoFisher Scientific, Waltham, MA, USA), supplemented with 40 U of RNAsin Plus RNase Inhibitor (Promega, Woods Hollow, Fitchburg, WI, USA) and preserved at −80 °C until use. The total number of synthetic RNA molecules in the standard stock solution was calculated as previously described [31].

2.6. Determining the Limits of Detection and Quantification of the qRT-PCR Protocol

Ten-fold serial dilutions of VHSV in vitro transcribed RNA, at known concentrations, were used to determine the limit of detection (LoD) and limit of quantification (LoQ) of the quantitative real-time RT-PCR (qRT-PCR) protocol. The concentration of RNA ranged between 10−1 and 108 copy number per µL. Each dilution was tested in triplicate, together with a no-template control. The employed primers and the probe, reported in Jonstrup et al. [25] and targeting a conserved region within the N gene, are described in Table S1, while the amplification reaction conditions are detailed in Table S2. The LoD was defined as the lowest sample dilution for which all the replicates tested positive. The LoQ was determined by the lowest sample dilution for which all the replicates tested positive with a coefficient of variation (CV) of the estimated copy numbers < 35 per cent [32].The developed VHSV qRT-PCR protocol provided a LoD and LoQ of 0.5 viral copy numbers (CN) detected in one ng of total RNA.

2.7. Quantification of VHSV Genomic Copies

Intra-host viral replication was assessed in fish target organs of the transcriptomic groups. Purified RNAs (n = 330) were used to detect the VHSV gene copy numbers by applying a new qRT-PCR protocol. The assay was developed starting from the reference qualitative diagnostic protocol [25] modified with an absolute quantification approach, as described below. Quantitative real-time RT-PCR reactions were carried out in a final volume of 25 μL using 5 μL of template (normalized RNA sample or VHSV synthetic RNA). Amplification reactions were performed in a CFX96 Touch™ Real-Time PCR Detection System (BioRad, Hercules, CA, USA) under the conditions reported in Table S2. Amplification data were analyzed with the Bio-Rad CFX Manager software V3.1.
The amount of VHSV genome copy numbers was determined by interpolation of the quantification cycle (Cq) values obtained with the standard curve developed with ten-fold serial dilutions of in vitro transcribed RNA tested in triplicate. In each run, VHSV synthetic RNA dilutions were used and ranged from 102 and 107 copy numbers per μL. All specimens were analyzed individually (n = 150). Each qRT-PCR reaction contained 100 ng of template RNA and the results are reported hereinafter as Log10 of copy numbers detected in one ng of total RNA (LCN/ng).

2.8. RNA Sequencing (RNA-Seq)

Considering the high costs of the RNA-Seq technique, it was not possible to test all the samples collected from the transcriptomic challenge group. The specific times post-infection (namely 1, 2, 5 dpi), the target organ (namely the HK), and the number of animals (n = 4 out of 5) in each of the three groups (challenged with VHSV-H, VHSV-L, and mock-infected) were selected based on the results obtained by qRT-PCR (See Section 3.2 and Table S3). The transcriptome library of each sample (n = 36) was prepared starting from 500 ng of total RNA using the TruSeq Stranded mRNA Library Prep kit (Illumina Inc., San Diego, CA, USA) following the company’s protocols. Each library was run on an Agilent 2100 Bioanalyzer using Agilent High Sensitivity DNA kit to ensure the proper range of cDNA length distribution. Sequencing was performed on an Illumina NextSeq instrument with NextSeq® 500/550 High Output Kit v2 (75 cycles) in single-read mode (Illumina, San Diego, CA, USA) in order to produce about 30 million reads per sample. Raw sequencing data are available at SRA (NCBI) under accession number PRJNA1024374.

2.9. Bioinformatic Analysis

Raw data were quality filtered by clipping library adaptors and trimming low quality ends with trimmomatic v0.38 [33] using standard parameters. Passing filter reads shorter than 50 bp were removed. High quality data were aligned against Oncorhynchus mykiss genome (Omyk_1.0, Ensembl release GCA_002163495.1) with STAR v2.7.6a [34] using standard parameters. Gene count was generated with htseq-count v0.11.0 [35] using the “union” mode and “exon” feature type. Differential expression analysis was performed with DeSeq2 package v1.20.0 [36]. Differentially expressed genes (DEGs) were defined using FDR < 0.05 and |log2FC| ≥ 1 (FDR: False Discovery Rate; FC: Fold Change). Gene Ontology (GO) terms were assigned to each gene by using Blast2GO v5.2.5 [37] and merged with those downloaded from Ensembl through BioMart [38]. GO enrichment was computed by Fisher’s exact test and p-values were adjusted through Benjamini–Hochberg correction. An FDR < 0.05 was considered significant. Enrichment scores were computed as -log(FDR). Child-father relationships belonging to GO graph were reconstructed using the OBO file downloaded from [39]. To compute expression levels of viral genes, we recovered not mapping reads and mapped them to VHSV-H and VHSV-L genomes with STAR v2.7.6a using standard parameters. Gene count was generated with htseq-count v0.11 as previously described. Viral expression levels were computed as RPKM (Reads Per Kilobase of transcript per Million mapped reads.). Expression levels of gene N were used for a comparison with the genomic copy number computed by qRT-PCR protocol. Correlation value was computed as a linear regression value.

2.10. Statistical Analysis

To estimate the survival function from lifetime data, the non-parametric Kaplan–Meier method was used, which allowed the survival curve to be drawn as a step curve for each experimental group, measuring the length of time the fish survives the infection [40]. To compare survival curves under different experimental conditions, the Wilcoxon non-parametric test was used. Chi square test was employed to compare cumulative mortality at the end of the study period between independent groups.
The viral quantification data of (five) biological replicates are expressed as mean genomic copy numbers on one nanogram of total RNA. To streamline reading, numbers are reported in the text as logarithmic values of copy number (LCN). For pair comparison of CN from each organ between groups at each time point, Mann–Whitney U test was applied. The level for accepted statistical significance was p-value <0.05. Statistical analyses were performed using STATISTICA 13 (Tibco Software, Palo Alto, CA, USA). Reported graphs were generated and edited using GraphPad Prism Software (v10.0.2 GraphPad Software, San Diego, CA, USA).

2.11. Ethics Statement

The experimental protocol for fish challenges was designed in compliance with Directive 2010/63/EU and the National Legislative Decree n° 26/2014. The experimental design was evaluated by the IZSVe Animal Welfare Body and Ethics Committee and finally approved by the Italian Ministry of Health with authorization n° 735/2016-PR 22/7/2016.

3. Results

3.1. Cumulative Mortality and Survivorship Curves

To confirm the difference in terms of virulence of the VHSV isolates in experimentally challenged rainbow trout, cumulative mortality was calculated for each strain over the observation period. Results confirmed the previously established virulence phenotype of the two employed VHSV strains [16], although, in general, a slightly lower mortality was observed. Indeed, at the end of the observation period, cumulative mortality was 96.4% for VHSV-H (n = 53 dead; n = 2 survivor) and 3.6% for VHSV-L (n = 2 dead; n = 53 survivor), which confirms the different phenotype of the two VHSVs. All the mock infected fish survived. Kaplan–Meier curves developed by plotting the survival rates during the observation period (Figure 1a) represent the daily mortality data recorded in all the challenged experimental groups. All tested fish resulted positive for VHSV by rRT-PCR.

3.2. Determination of Viral Replication

The quantification of VHSV copy numbers was assessed applying the qRT-PCR protocol to the SPL and HK samples from five fish per time point, collected from the VHSV-L and VHSV-H treated groups as well as from the mock group. All detailed values are available in Table S3. In summary, rainbow trout infected with the VHSV-L strain started to show quantifiable viral loads from 2 dpi (Mean LCN/ng of 1.57 in SPL and 1.61 in HK). Mean LCN/ng values then rapidly started to increase and reached the maximum peak at 3 dpi with values of 2.91 in SPL and 2.70 in HK. At 5 dpi, the mean values slightly dropped to 2.17 and 2.38 in SPL and HK, respectively. On the other hand, quantifiable viral loads in rainbow trout infected with VHSV-H were already detectable starting from 1 dpi, when an increasing trend in viral loads over time was detected. Indeed, at 2 dpi mean LCN/ng values were 4.33 and 4.58 for SPL and HK, respectively, reaching the maximum level of viral genomic copies at 3 dpi (6.23 for SPL; 6.19 for HK). At 5 dpi, the viral loads reached a plateau with observed values of 6.13 in SPL and 6.06 in HK (Figure 1b). As expected, the mock samples analyzed at all time points tested negative for VHSV.
Interestingly, the LCN/ng values detected in VHSV-H infected fish were higher than those observed in fish infected with VHSV-L and exhibited significant differences (p < 0.001) at 2, 3, and 5 dpi in both target organs. Moreover, the number of detected viral genomic copies was almost equivalent in the SPL and HK of all the challenged fish. Hence, transcriptome analyses were performed only on HK samples as the head kidney is considered the dominant hematopoietic organ [41,42]. Expression levels for gene N computed with RNA-Seq data showed high level of correlation (R2 = 0.9937) with CN values.

3.3. Data Quality, Mapping and Normalization

Transcriptome changes in the HK of rainbow trout infected with two different VHSV strains were investigated using RNA-Seq technology. As previously stated, only head kidneys were tested for transcriptome analyses. Sequencing produced a grand total of 1,642,117,765 single-end 75 bp reads for 36 samples, accounting for about 45 million reads per sample (Table S4). Quality filtering removed 8.4% of the generated data, allowing the recovery of 41,799,829 high-quality reads per sample on average. After alignment of generated high-quality data against O. mykiss genome, 79.6% of the reads resulted as uniquely mapping and the intersection with annotation revealed that 22,543,860 high-quality reads per sample on average were assigned to genes.
The principal component analysis (PCA) of normalized counts of host genes confirmed viral infection progression induced by both VHSV strains (Figure 2). In general, samples from infected fish moved along the first PCA component (PC1), which accounts for 46% of the total variance. Transcriptomes from fish infected with a highly pathogenic strain resulted separated from those belonging to fish from the mock infected group, starting from early time points in comparison to what observed for the low pathogenic strain. In detail, transcriptomes from both VHSV-H and -L infected fish at 1 dpi overlapped with those from mocks while, on the second day of infection, a well separation from the mock infected fish was observable only for VHSV-H samples. This trend was further strengthened at 5 dpi. Differently, even if transcriptomes from VSHV-L samples reached the same separation from mocks at 5 dpi, at previous time points (1 and 2 dpi) they showed little to no separation from the mock samples. While looking at the second PCA component (PC2), accounting for 28% of the total variance, no peculiar results were highlighted except for a clear separation of four samples from the remaining ones. More specifically, those samples belonged to various conditions, i.e., one replicate from different time points of mock, low, and high VHSV infected groups. PC2 seemed to capture some aspects of the experiments that are not correlated with the infections.

3.4. Transcriptome Analysis over Time

The characterization of host response to the two VHSV strains at the different time points (1, 2, 5 dpi) was performed by comparing infected with mock fish at each time point separately. At 5 dpi, a similar number of differentially expressed genes (DEGs) was detected in both infection challenges, reaching 5338 and 7448 DEGs for VHSV-L and VHSV-H conditions, respectively (Figure 3). However, the latter challenge group also prompted a strong host response at the previous time point, with 2003 DEGs at 2 dpi, and even a mild response at 1 dpi with 866 DEGs. On the contrary, in the VHSV-L challenge group, at 1 and 2 dpi only 106 and 403 DEGs were found, respectively. Such numbers are probably not biologically relevant because they are lower than the variation observed when comparing specific time point mock replicates against each other.
While looking at the gene sharing level within each challenge group, results revealed that 45 and 51% of found DEGs, at 1 dpi, were specific to such time point for VHSV-L and VHSV-H, respectively (Figure 4a,b). When considering 2 dpi, the percentages of specific DEGs were 40 and 27% for VHSV-L and VHSV-H group, respectively. However, at the latest time point (4 dpi), such percentages increased up to 95% for the VHSV-L group, while for the VHSV-H group the percentage reached 76%. Additionally, when considering the VHSV-H specimens, it is worth noting that days 2 and 5 post infection shared 18% of DEGs.
Since pairwise differential expression analysis revealed that during the latest time points the host mounted the largest response, we checked how many DEGs were commonly shared between the two groups (Figure 4c). Overall, the majority of the DEGs were unique to each condition, in particular at 5 dpi, which included 1608 regulated genes (16.5%) for VHSV-L and 3555 DEGs (36.4%) for VHSV-H. About 138 DEGs were shared between all crossed time points of both challenges. Moreover, it is worth mentioning that 2388 (24.5%) DEGs were shared between the two groups at 5 dpi, while 990 (10.1%) DEGs were shared between the three conditions with the highest number of DEGs (2 and 5 dpi VHSV-H, 5 dpi VHSV-L).
For details of all the genes found as DEGs see Table S5.

3.5. Top Differentially Expressed Genes

In order to focus on the genes with a biologically continuous and strong impact in the course of infection, amongst all the DEGs we selected those simultaneously present in all the three sampling time points for each challenge group.
After the removal of the genes without known function, within the VHSV-H group there were globally 39 DEGs, 5 of which were upregulated, 3 downregulated, and 31 displayed a fluctuating expression trend at different time points (see Table 1). The most upregulated gene was trim63a (or MuRF1) that had a zebrafish orthologous encoding for a RING-type E3 ubiquitin-protein with transferase activity likely involved in positive regulation of DNA-templated transcription. The second over-expressed gene in the VHSV-H group was acod1 (or irg1), predicted to have an aconitate decarboxylase activity resulting in itaconate production. Another upregulated gene in the VHSV-H infected group was cox-2, also called ptgs2, an enzyme with a peroxidase activity involved in prostaglandin biosynthesis and predominating in inflammation sites.
Moreover, skia was found among the upregulated genes. This gene is predicted to have a SMAD binding activity and a transcription corepressor activity.
Lastly, results showed a slight upregulation of a gene that is orthologous to hipk1 encoding a serine/threonine-protein kinase involved in transcription regulation and TNF-mediated cellular apoptosis.
Considering the downregulation genes, within the VHSV-H group, gene cx35.4 (or gjb3) was found to be the most downregulated. In zebrafish, it encodes a predicted gap junction involved in cell communication. One gap junction consists of a cluster of closely packed pairs of transmembrane channels, the connexons, through which materials of low molecular weight diffuse from one cell to a neighboring cell.
The second downregulated gene was the non-coding gene mir-142, a microRNA with a potential regulatory function in eukaryotic cells.
The zinc finger protein 572 gene, likely involved in the transcriptional regulation as reported for its hypothetical human orthologous, was also slightly under-expressed in the VHSV-H group.
Finally, of the 31 DEGs found as differentially expressed with a fluctuating trend at different time points, we will mention only those which were also found as DEGs in at least two time points of the VHSV-L group. In detail, downregulated genes at 1 dpi but upregulated at subsequent sampling time points were (i) sox19b, which encodes a transcription factor DNA-binding protein involved in the development of the central nervous system, while its orthologous SOX15 evolved through neofunctionalization is required for skeletal muscle regeneration, (ii) col2a1a, encoding the alpha-1 chain of type II collagen, and (iii) tdrd7a, which encodes a protein with predicted mRNA binding activity. In humans, the Tudor domain-containing TDRD proteins are involved in the post-transcriptional regulation of specific genes required during lens development and also appear to have antiviral activity against herpes simplex virus HSV-1, (iv) Avd, encoding for a protein with peculiar biophysical characteristics and highly expressed in the gonads of both male and female zebrafish and in the gills of male fish, (v) lhx8a, with an involvement in basal forebrain cholinergic neurons development and in the regulation of transcription by RNA polymerase II, (vi) kcnk10a, likely involved in potassium ion transmembrane transport and stabilization of membrane potential, (vii) tgfa in zebrafish and human orthologues encodes a ligand involved in epidermal growth factor receptor (EGFR) signaling pathway; it is also involved in positive regulation of cell proliferation, in differentiation, and in development, (viii) rapgefl1, in zebrafish and humans, this is a probable guanine nucleotide exchange factor predicted to be involved in G protein-coupled receptors mediated signal transduction and in the development of the nervous system.
Epyc (or PG-Lb, dspg3) was also a DEG with a fluctuating trend, but in a different way (downregulated gene at 1 and 2 dpi that changed to upregulated at 5 dpi). This gene encodes a member of the small leucine-rich repeat proteoglycan family involved in fibrillogenesis by interacting with collagenous fibrils and other extracellular matrix proteins.
The only upregulated gene at 1 dpi but downregulated at subsequent sampling time points was uox. This gene, in zebrafish and mice, exhibits a urate oxidase activity with an involvement in protein homotetramerization and urate catabolic process.
Within the VHSV-L group, only 10 genes with the same criteria applied to the VHSV-H group were found. One gene was upregulated, two were downregulated, and seven were fluctuant-expressed genes (Table 2). The most upregulated gene was preproinsulin (named ins), predicted to have a hormone activity. After post-translational modifications, insulin acts upstream of or within several processes, including regulation of feeding behavior, especially in regulation of carbohydrate and lipid metabolism.
The two downregulated genes were trim63a, already described above, and mtnr1a. Mammalian orthologue of the latter mentioned gene is melatonin receptor 1A, encoding an integral membrane protein predicted to be involved in G protein-coupled receptor signaling pathway. This receptor likely mediates the major neurobiological effects and circadian actions of melatonin.
Among the differentially fluctuant-expressed genes, at the three time points in the VHSV-L group, the downregulated genes at 1 dpi that were upregulated at subsequent time points were: col2a1a, encoding alpha-1 chain of type II collagen, previously described above and nkx2.5, predicted to have a DNA-binding transcription factor activity and an essential role during cardiomyogenesis in humans. Moreover, downregulated genes at 1 and 2 dpi but upregulated at 5 dpi were: klhl41a likely involved in skeletal muscle development and differentiation and epyc, a small leucine-rich repeat proteoglycan. On the contrary, the upregulated gene at 1 dpi that was downregulated at subsequent time points was uox, encoding for a urate oxidase protein (see above). Surprisingly, the upregulated genes at 1 dpi and 5 dpi that downregulated at 2 dpi were: a slightly different paralogue of trim63a, previously mentioned, and a dok5-like gene which in humans encodes for an enzymatically inert adaptor that provides a docking platform for the assembly of multimolecular signalling complexes. Generally, this gene plays a role in activating the MAP kinase pathway and is involved in neuronal differentiation. Moreover, we decided to observe the most up- and down-regulated DEGs of each time point and for each experimental group, regardless of their co-presence in the other time points (Table S6). Interestingly, we observed that a good amount of those DEGs were already present in the analyses previously described.

3.6. Gene Ontology Enrichment Analysis

To better understand the host response to VHSV infection, we examined the enriched biological processes in the differentially expressed gene set using gene ontology (GO) databases (Table S7). In general, the number of enriched GO terms followed a similar trend to the number of DEGs found at each condition, with both challenges reaching a peak at the last time point (180 and 312 enriched GO terms for VHSV-L and VHSV-H challenge, respectively). The only deviation from such a trend was found at 2 dpi for both challenges. Indeed, at this time point the enriched GO terms accounted for VHSV-L and VHSV-H trials were 135 and 258, respectively, a considerable level of enrichment despite the low number of detected DEGs. It is interesting to note how the low number of detected DEGs (403 in total) at 2 dpi for the VHSV-L challenge boosted a relatively strong enrichment, indicating a probable activation of the host response already at such a time point. At the earliest time point (1 dpi), the VHSV-L group showed no enrichment, while the VHSV-H group displayed only 40 enriched GO terms, thus reinforcing the idea that the host answer mounts later.
Overall, the host answer to VHSV-H infection started early, at 2 dpi, showing an already high number of enriched GO terms (258), which only slightly increased to 312 at the following time point. Considering altogether 2 and 5 dpi, the enrichment scores displayed a clear increasing trend of their values over time, with most general terms represented by “immune system process”, “cell death”, “signaling”, “locomotion”, and “developmental process”. In detail, enrichment scores ranged from 1.3 to 32.2 at 2 dpi and from 1.3 to 55.1 at 5 dpi, averaging, respectively, 5.2 and 5.7. Interestingly, at 5 dpi, specific terms appeared among the other enriched GO terms and included “biological adhesion”, “cytolysis”, “regulation of catalytic activity”, and “lipid metabolic process”.
Surprisingly, also in the VHSV-L challenge, the host seemed to be mounting a response to the infection starting at 2 dpi considering the number of observed DEGs. Most general terms were the same as the ones detected in the VHSV-H challenge, with a notable exception represented by “growth” term, which appeared exclusively enriched at 2 dpi for the VHSV-L group. However, enrichment scores followed a trend similar to the one observed for the VHSV-H challenge, with values that increased from 2 to 5 dpi. In particular, enrichment scores ranged from 1.3 to 12.6 at 2 dpi and from 1.3 to 21.5 at 5 dpi, averaging, respectively, 2.9 and 2.7. At 5 dpi, specific terms appeared among the enriched GO, including “regulation of molecular function”, “immune effector process”, “cell death”, “regulation of catalytic activity”, “intermediate filament-based process”, and “cell adhesion”.

3.7. Analysis of Enriched GO Terms as a Graph

Considering the high number of significant GO terms according to the enrichment analysis, we decided to consider the graph structure on the Ontology (Table S8). Each GO term is a node, and the relationships between the terms are edges between the nodes. In general, a node always has at least one parent term and may have one or more child terms. The relation among GO terms is somehow hierarchical, with ‘child’ terms being more specialized than their ‘parent’ terms, but unlike a strict hierarchy, a term may have more than one parent term. We started from the most specific GO term among the enriched ones and listed all the enriched GO terms that were fathers to it. Such a list of enriched GO terms was considered a “group”. Then we proceeded to select the most specific GO term among the remaining enriched ones, and so on. In this way, we identified 95 groups of enriched GO terms that allowed us to distinguish between shared and virus-specific enriched functions.
Considering the most specific GO terms (Figure 5) resulted enriched at 2 and 5 dpi for both VHSV-L and VHSV-H, many are clearly relative to immune response, e.g., “response to virus”, “regulation of defense response”, “inflammatory response”, “tumor necrosis factor-mediated signaling pathway”, “regulation of apoptotic process”, “cytokine-mediated signaling pathway”, “regulation of cytokine production”, and “regulation of interferon-gamma-mediated signaling pathway”. Other notable GO terms enriched in a similar pattern are “negative regulation of receptor signaling pathway via JAK-STAT”, “regulation of leukocyte migration”, “T cell activation involved in immune response”, “tissue regeneration”, “NAD biosynthetic process”, “regulation of protein kinase A signaling”, “serine family amino acid metabolic process”, and “toll−like receptor signaling pathway”. At 1 dpi, and only for the VHSV-H challenge group, we found enriched “fatty acid biosynthetic process” and “negative regulation of endopeptidase activity”. However, all of them reappeared as enriched at 5 dpi for both challenges.
Conversely, a certain number of GO terms resulted as enriched specifically for only one of the two challenged groups. The enriched biological processes exhibited in the VHSV-H group at both 2 and 5 dpi were “regulation of transcription by RNA polymerase II”, “regulation of histone H3-K4 methylation”, and “protein ubiquitination”, whereas at 2 dpi “antigen processing and presentation of exogenous peptide antigen via MHC class I”, “activation of NF-kappaB-inducing kinase activity”, and “positive regulation of JNK cascade” were enriched. Finally, at 5 dpi, we found ten enriched GO terms: “antigen processing and presentation of peptide antigen via MHC class I”, “peptidyl-tyrosine phosphorylation/dephosphorylation”, “regulation of Wnt signaling pathway”, “glycogen metabolic process”, “angiogenesis”, “regulation of blood pressure”, and three GO involving the “transforming growth factor signaling pathway”.
On the other hand, for VHSV-L challenge, we only found as enriched “negative regulation of NF-kappaB transcription factor activity” at 2 dpi and “CAAX-box protein processing” at 5 dpi.

4. Discussion

The purpose of our study was to analyze, at transcriptomic level, the response of rainbow trout infected with high and low virulent VHSV strains. According to the virulence classification proposed by Panzarin et al. [16], VHSVs can be classified as highly virulent when the cumulative percent of mortality (CPM) is >42%, moderate virulent with CPM values ranging from 42 to 14%, and low virulent when CPM is <14%. Indeed, the two VHSVs selected for the present study were characterized as possessing the most diverse phenotype, with the VHSV/O.mykiss/I/TN/80/Mar10 proving to be highly virulent (therefore called VHSV-H) and the VHSV/O.mykiss/I/TN/480/Oct96 low virulent (referred to as VHVS-L) [16,29]. To further confirm the different phenotype of the selected viruses, cumulative mortality experiments were repeated and the obtained CPM, namely 3% for VHSV-L and 96% for the VHSV-H, were as expected.
In our study, we followed the time-course infection of both low and high virulent VHSV by measuring the number of viral genomic copies at different time points (0.5, 1, 2, 3, and 5 dpi) in two important hematopoietic organs, such as the head kidney and spleen [42,43]. No differences in terms of genomic copy numbers between the two target organs at each time point were detected. Such a result allowed to choose a single organ—namely the head kidney as it is considered the dominant hematopoietic organ [43]—for the subsequent transcriptome analyses. Furthermore, genomic copy numbers in both target organs of trout infected with VHSV-H were significantly higher in comparison to the ones found in the group infected with VHSV-L. This could suggest a probable difference in the absorption capacity of the two viruses, as already reported in different in vitro studies [21], even if we cannot exclude that other steps in the viral cycle could have been affected by the genomic modifications present in the viral strain used for this study. Indeed, both VHSV infections induced massive changes (10,039 overall annotated genes found to differentially expressed in at least one comparison) in the rainbow trout transcriptome. GO term enrichment analysis showed that they were enriched in 512 GO terms, indicating that VHSV infection modulates the expression of genes involved in a broad variety of functions, mostly related to immune response and metabolism, as expected during a viral infection.
Furthermore, replication experiments highlighted that both VHSV strains increased their viral copies between 1 to 3 dpi and then stabilized between 3 to 5 dpi. However, the VHSV-L viral load increased more slowly, becoming detectable only at 2 dpi, and even less with respect to VHSV-H. Despite the fact that viral replication analysis revealed a similar trend to the infection kinetics, although with different timing among strains, such a trend differed if compared to the transcriptomic analysis. In fact, host response to VSHV-H started at 1 dpi, consistent with viral replication, increased until 3 dpi, and reached its peak at 5 dpi. Differently, VSHV-L stimulated a weaker response until 3 dpi and then increased at 5 dpi, but without reaching the VSHV-H levels regarding the quantity of DEGs, number, and scores of enriched GO terms. Notably, such difference is consistent with the KM survival curve observed for both strains.
Looking at the analyses, we observed that the majority of DEGs always expressed at all time points (n = 39) were related to the VHSV-H infection, while only a few (n = 10) were specific to the VHSV-L challenge. Worthy of note was the TRIM63 gene encoding an E3 ubiquitin ligase, which was found to be over-expressed in several vertebrates with a detrimental role during skeletal muscle atrophy [44]. Furthermore, TRIM63 is also probably involved in other processes (e.g., targeting pro-anabolic factors, intracellular trafficking) and, as other members of its family, seems to have emerging roles in innate immunity, since it is induced by IFNs [45,46]. Interestingly, Kim et al. found TRIM25, a protein belonging to the same family as the above mentioned one, among up-regulated DEGs in rainbow trout infected with a highly virulent strain of infectious hematopoietic necrosis virus (IHNV; family Rhabdoviridae) [47].
The acod1 gene was involved in the inhibition of the inflammatory response and acted as a negative regulator of the Toll-like receptors (TLRs)-mediated inflammatory innate response by stimulating the tumor necrosis factor alpha-induced protein TNFAIP3 expression via reactive oxygen species in lipopolysaccharide-tolerized macrophages [48,49]. In zebrafish and humans, acod1 orthologous genes were involved in the defense response to pathogens and in regulating reactive oxygen species biosynthetic process; however, the implication of these orthologues in promoting viral replication was also demonstrated [49,50]. Interestingly, acod1 was overexpressed in different hosts, in vivo or in vitro, following infections with various pathogens (bacteria, virus, or parasite) [51]. In particular, while in some cases an over-expression of acod1 seemed to promote an antiviral action, in other studies involving vesicular stomatitis virus (VSV, another virus of the family Rhabdoviridae) and human alphaherpesvirus 1 (family Herpesviridae), acod1 was associated with the promotion of viral replication [51]. Moreover, it was also observed that the over-expression of acod1 in organs of mice infected with VSV favored the entry and overgrowth of this viral species by producing itaconate, which promoted protein prenylation via an IFN-I independent mechanism [52]. Acod1 was also found among the over-expressed genes in a recent study on RNA-Seq conducted on avian influenza virus (Family Orthomixoviridae). Interestingly, a transcriptional increase of acod1 was observed only in the groups infected with precursor strains of highly pathogenic avian influenza viruses [53].
Cyclooxygenase 2 (cox-2) had a pivotal role in the establishment and maintenance of inflammatory conditions, as well as in the aetiology of some tumour processes. This gene is also involved in numerous metabolic, signalling, and pathogenic pathways [54]. The over-expression of cox-2 was considered as an indicator of invasiveness, aggressiveness, and metastatic potential in different malignancies, including carcinomas linked to human papilloma virus (HPV, family Papillomaviridae) infection [55,56]. Cox-2 protein was also involved in viral replication in cytomegalovirus (HCMV, family Herpesviridae) infections [57]. In Kaposi’s sarcoma-associated herpesvirus (KSHV, family Herpesviridae) infection, cox-2 was upregulated and involved in the progression of viral infection [58].
Skia is another upregulated gene described in our study, probably involved in different processes including transcription, cardiac conduction, cartilage development, and heart looping. Its orthologous in zebrafish played a crucial role in dorsal-ventral patterning embryogenesis [59]. Most importantly, the human orthologous (SKI) was described as a TGF-β signaling negative regulator [60,61] and was found to be differentially expressed in various diseases, as well as in vitro studies, which highlighted its upregulation during viral infection [62].
Finally, hipk1 showed a mild upregulation in the VHSV-H infected group. Interestingly, its human orthologous appeared to interact in p53 regulation during cell apoptosis in acute and chronic kidney diseases [63,64]. As a matter of fact, the kidney is one of the target organs of VHSV infections.
In the VHSV-H group, cx35.4 turned out to be the most under-expressed gene. Unfortunately, cx35.4 is not so well characterized; however, other human connexins like genes (Cx31 and Cx43) are widely studied. Interestingly, the downregulation of these latter mentioned genes was observed during a range of human viral infections associated with tumors development [65].
Additionally, the non-coding microRNA-142 was found to be downregulated in the VHSV-H group. Interestingly, its orthologe was observed as downregulated in zebrafish larvae exposed to environmental pollutants [66]. It has recently been established that miR-142 is a specific negative regulator of TIM-1, which is one of the factors involved in the viral internalization in different endothelial disease [67].
During VHVS-L infection the most up-regulated DEG was ins, whose role still remains unclear. Conversely, the downregulated genes were (i) trim63a belonging to the large family of tripartite motif-containing (TRIM) proteins, known to be crucial for most aspects related to pathogens resistance [45], and (ii) mtnr1a, which was actually downregulated only at 2 dpi.
Notably, based on a review regarding the fish innate immunity against Novirhabdoviruses [68], the TLR3, TLR7, MDA5, and LGP2 (or dhx58) genes were reported as transcriptional up-regulated receptors involved in the recognition pattern in response to VHSV infection. Similarly, we found all the above-mentioned genes over-expressed in fish of both experimental trials, even if at different timings (at 2 dpi for VHSV-H; at 5 dpi for VHSV-L). This observation further strengthened the hypothesis that the infection with the VHSV-L strain induced a slower viral kinetic in comparison to VHSV-H strain.
Looking at the GO terms, in both challenges and as expected during an infection, the host response was mainly represented by the immune system with the activation of biological functions such as “response to virus”, “regulation of defence response”, and “inflammatory response”. Such enriched GO terms appeared while we were performing our experiments during the second part of the study, meaning at 2 and 5 dpi, even though for VSHV-H the enrichment was stronger than the one observed for VSHV-L, as expressed by the enrichment scores (Figure 5). Indeed, functions like “inflammatory response”, “cell chemotaxis”, “response to stress”, and “cytokine-mediated signalling pathway” had almost double the enrichment scores in VHSV-H when compared to VSHV-L infection, and such a result indicated a stronger and earlier immune response induced by the highly virulent strain in comparison to the low virulent one. Notably, some GO terms related to the immune system appeared identical in both strains in terms of timing and score, e.g., “tumour necrosis factor-mediated signalling pathway” and “regulation of interferon-gamma-mediated signalling pathway”. Regarding the latter GO, Kim et al. [47] reported its enrichment at 3 dpi during a similar experimental trial performed using the other salmonid Novirhadbovirus (IHNV). Tumour necrosis factor (TNF) is a type II transmembrane protein expressed in the plasma [69]. TNF is a multifunctional cytokine that plays a crucial role in several cellular events, such as cell survival, proliferation, differentiation, and death. This protein exerts its biological functions through the activation of distinct signalling pathways, for example nuclear factor κB (NF-κB) and c-Jun N-terminal kinase (JNK). NF-κB is a major anti-apoptotic cell survival signal, while sustained JNK activation contributes to cell death. The crosstalk between the NF-κB and JNK determines cellular outcomes in response to TNF [63]. The virus ability to modulate TNF-mediated NF-kB activity is generally a strategy to promote viral replication. It has already been described that VHS virus suppresses the early activation of NF-κB in host cells, through the action of NV protein [70].
Immune response is always accompanied by other host processes, directly or indirectly linked to the viral infection. One example could be the need to repair any tissue damage provoked by the viral replication itself or by the immune system activity in an attempt to fight the virus. Indeed, “tissue regeneration” was observed as enriched, with the same score for both strains at 2 and 5 dpi, thus indicating a similar level and timing of tissue repair processes activation. Simultaneously, the GO term “regulation of apoptotic process”, which is a regulated mechanism of cellular death employed by the immune system to counter an infection, together with the “negative regulation of protein serine/threonine kinase activity” appeared enriched for both viruses, although with a higher score during VSHV-H rather than VHSV-L infection. Indeed, serine/threonine kinases are involved in several biological process, including apoptosis. Moreover, other biological functions arose with different timing between the two strains, all of them appearing earlier for VSHV-H compared to VHSV-L. Such functions included some immune system processes, e.g., “toll-like receptor signalling pathway” [71], but were mainly represented by terms such as “negative regulation of endopeptidase activity”, “fatty acid biosynthetic process”, and “NAD biosynthetic process”, all tied to viral needs such as viral entry into the cell, metabolism, and protein modification. In particular, fatty acid metabolism is generally a central process during viral infections. For instance, glycerophospholipid species yields increase during the Influenza virus infection (IVI) in mammalian species [72]. Tanner et al. [73] assumed there was a principal correlation between influenza replication and choline lipids metabolism. As a matter of fact, they found an IVI-mediated reduction in ester-linked phosphatidylcholine (PC) species, as well as an increased level of sphingomyelin (SM) [73] probably connected with expending cellular choline stores for SM synthesis. Such modifications led to an increase in SM species within the infected cell. Higher amounts of SM and short-chain fatty acid-containing ether-linked PC (ePC) species were found in both infected cells and virions and therefore appeared to be involved in viral morphogenesis. On the other hand, it was observed that the levels of long-chain fatty acid-containing ePC had increased in infected cells, while they were lower in the structure of the virion, highlighting the role of this phospholipid in replication of the virus. The increased biosynthesis of fatty acids and membrane lipids may reflect the fact that the virus stores structural lipids for the production of infectious particles. In olive flounder (Paralichthys olivaceus), challenged by intraperitoneal infection with VHSV and observed for 7 days (40% cumulative mortality), post infection transcriptome analysis showed that fatty acid degradation was suppressed [74]. This observation led us to speculate that the process of entry and replication of VHSV-H into host cells was completed a few hours after infection, and that the cell machinery was rapidly diverted to favour the pathogen replication. This provoked a high viral genome replication and, as a consequence, impeded the recovery of infected trout despite the strong triggered immune response.
Nevertheless, our analyses revealed the existence of functions that were VHSV-strain specific or displayed an opposite behaviour between high and low virulent strains. In particular, the nuclear factor kappaB (NF-κB) signaling appeared to be regulated differently in the two viral strains: activated for VHSV-H at 2 dpi, as expressed by enriched GO term “activation of NF-kappaB inducing kinase activity”, while inhibited for VHSV-L at the same time point, as expressed by enriched GO term “negative regulation of NF-kappaB transcription factor activity”. Interestingly, the NF-κB is the master transcription factor that controls the cell proliferation, apoptosis, and the expression of interferons, cytokine, and the proinflammatory factors [75]. As NF-κB has a key role in controlling host defense processes, viruses have evolved strategies to modulate NF-κB signaling to either evade host surveillance or exploit it for viral gene expression. In this work, we observed that NF-κB was activated by VHSV-H but negatively regulated by VHSV-L, confirming once again the strongest ability of the VHSV-H strain to infect the host. Moreover, a plethora of functions appeared specifically for VHSV-H in the last time points of infection. However, only some were linked to the immune response, while the majority were related to metabolism, viral entry into the cell, transcription regulation, and cytoskeleton. Such results further confirmed the capacity of VHSV-H in taking control of the cell machinery to promote its own replication. Furthermore, transcription appeared to be regulated at both 2 and 5 dpi, as indicated by GO terms “regulation of transcription by RNA polymerase II” and “regulation of histone H3-K4 methylation”. The latter is an important function to promote transcription, as histone methylation is critical for gene expression through gene accessibility. The transcriptional machinery, RNA polymerase II, is involved not only in the host’s transcriptional cellular processes but also in the viral mRNA capping machinery, as reported for influenza virus [76]. Perhaps a similar involvement of the eukaryotic polymerase could occur during a VHSV infection. Other important biological functions specific to VHSV-H are the de- and phosphorylation of peptidyl-tyrosine, both observed to be associated to cell entry for the herpes simplex virus [77]. Lastly, some GO terms related to blood, such as “angiogenesis”, “vascular endothelial growth factor signaling pathway”, “vascular endothelial growth factor receptor signaling pathway” and “regulation of blood pressure”, emerged as enriched specifically for VHSV-H at 5 dpi. Given that among the major clinical signs of VHSV infection, as indicated by the disease name itself, are hemorrhages caused by the active replication within the vascular endothelial cells [78,79,80], it is not surprising that such GO terms emerged. Similarly, the absence of such hemorrhages, and consequently no mortality, in VHVS-L infected fish is confirmed by the non-presence of these GO terms.
Finally, the only enriched GO term specific to VHSV-L was the “CAAX-box protein processing”. The presence of a CAAX box is known to be a necessary process for the post-translational modification of proteins called prenylation. Briefly, this process involves a lipid addition to nascent proteins that imparts new hydrophobic properties to the mature proteins for targeting towards cell membranes or organellar membranes [81]. It has also been shown that prenylation plays a key role in the life cycle of numerous pathogens, both bacteria and viruses [82,83]. Indeed, the prenylation mechanism of the eukaryotic host cell is exploited to the advantage of the pathogen. This observation could further explain the different virulence of the VHSV strains compared in the present study: the highly virulent virus enters immediately into the host cells, subdues the metabolism machinery to its need and replicates uncontrollably by the immune response. Conversely, the low virulent virus causes infection with a much slower course, encountering more obstacles during cell entry and replication, thus giving the host the opportunity to mount an adequate immune response and resolve the infection in most cases.

5. Conclusions

This in vivo study investigated the transcriptomic profile modulations in rainbow trout challenged with two VHSV strains belonging to different virulent classes. The abundant amount of DEGs and GO terms confirmed previously published results obtained from in vitro experiments or conducted in other fish species. Besides, it has been observed that strains with high or low pathogenicity are responsible for the modulation of many common biological mechanisms but also of some different host responses. Overall, we demonstrated that the processes of inflammatory response, the cytokine-mediated signalling pathway and many genes and processes involved in immune response are altered in rainbow trout during the first days of infection with VHSV. Therefore, we can assert that the achieved results have shed light on the biological and cellular mechanisms activated by VHSV in the early stages of infection; furthermore, they will be of help to future studies whose aim will be to deepen the comprehension and to better clarify the patterns of molecular responses induced by VHSV infection.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms12010057/s1, Table S1: Oligonucleotides used in the present study; Table S2: Reaction conditions of molecular amplification viral assay applied in this study; Table S3: qRT-PCR detailed Results; Table S4: Overall statistics; Table S5: DESeq2 output for all DEGs; Table S6: Top Up and Down-regulated DEGs at each time point for each of the VHSV-H and VHSV-L groups; Table S7: GO terms enrichment results; Table S8: GO terms on graph structure.

Author Contributions

Conceptualization, V.P. and A.T.; methodology, V.P., F.P. and A.T.; software, G.Z.; formal analysis, M.A., L.B., A.B. and G.Z.; data curation, M.A., L.B. and G.Z.; writing—original draft preparation, L.B. and G.Z.; writing—review and editing, A.T., M.A., F.P. and A.M.; supervision, A.T.; funding acquisition, V.P. and A.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Animal Health and Welfare (Anihwa) ERA-Net Consortium, within the framework of the Novimark Project, grant number G88F13000660001. The APC was funded by IZSVe.

Data Availability Statement

RNA sequencing raw data generated in the present study are available at SRA (NCBI) under accession number PRJNA1024374.

Acknowledgments

The authors thank Francesca Ellero for editing the manuscript. This work was also developed following the results obtained from the Novimark Project (G88F13000660001) of the ERA network. We thank the ERA-Net Consortium and point out that the content of this article reflects the author’s point of view only and that the Consortium is not responsible for any use that may be made of the information contained therein.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. WOAH. Infection with viral haemorrhagic septicemia virus. In Manual of Diagnostic Tests for Aquatic Animals; WOAH: Paris, France, 2021; ISBN 978-92-95115-59-0. [Google Scholar]
  2. Schutze, H.; Enzmann, P.J.; Kuchling, R.; Mundt, E.; Niemann, H.; Mettenleiter, T.C. Complete genomic sequence of the fish rhabdovirus infectious haematopoietic necrosis virus. J. Gen. Virol. 1995, 76, 2519–2527. [Google Scholar] [CrossRef] [PubMed]
  3. Dietzgen, R.G.; Kondo, H.; Goodin, M.M.; Kurath, G.; Vasilakis, N. The family Rhabdoviridae: Mono- and bipartite negative-sense RNA viruses with diverse genome organization and common evolutionary origins. Virus Res. 2017, 227, 158–170. [Google Scholar] [CrossRef] [PubMed]
  4. Ke, Q.; Weaver, W.; Pore, A.; Gorgoglione, B.; Wildschutte, J.H.; Xiao, P.; Shepherd, B.S.; Spear, A.; Malathi, K.; Stepien, C.A.; et al. Role of Viral Hemorrhagic Septicemia Virus Matrix (M) Protein in Suppressing Host Transcription. J. Virol. 2017, 91, 2020. [Google Scholar] [CrossRef] [PubMed]
  5. Béarzotti, M.; Monnier, A.F.; Vende, P.; Grosclaude, J.; de Kinkelin, P.; Benmansour, A. The glycoprotein of viral hemorrhagic septicemia virus (VHSV): Antigenicity and role in virulence. Vet. Res. 1995, 26, 413–422. [Google Scholar] [PubMed]
  6. Lorenzen, N.; Lorenzen, E.; Einer-Jensen, K.; Heppell, J.; Davis, H.L. Genetic vaccination of rainbow trout against viral haemorrhagic septicaemia virus: Small amounts of plasmid DNA protect against a heterologous serotype. Virus Res. 1999, 63, 19–25. [Google Scholar] [CrossRef] [PubMed]
  7. Biacchesi, S.; Lamoureux, A.; Mérour, E.; Bernard, J.; Brémont, M. Limited Interference at the Early Stage of Infection between Two Recombinant Novirhabdoviruses: Viral Hemorrhagic Septicemia Virus and Infectious Hematopoietic Necrosis Virus. J. Virol. 2010, 84, 10038–10050. [Google Scholar] [CrossRef] [PubMed]
  8. Ammayappan, A.; Vakharia, V.N. Nonvirion Protein of Novirhabdovirus Suppresses Apoptosis at the Early Stage of Virus Infection. J. Virol. 2011, 85, 8393–8402. [Google Scholar] [CrossRef]
  9. Abbadi, M.; Fusaro, A.; Ceolin, C.; Casarotto, C.; Quartesan, R.; Dalla Pozza, M.; Cattoli, G.; Toffan, A.; Holmes, E.C.; Panzarin, V. Molecular evolution and phylogeography of co-circulating IHNV and VHSV in Italy. Front. Microbiol. 2016, 7, 1306. [Google Scholar] [CrossRef]
  10. Cieslak, M.; Mikkelsen, S.S.; Skall, H.F.; Baud, M.; Diserens, N.; Engelsma, M.Y.; Haenen, O.L.M.; Mousakhani, S.; Panzarin, V.; Wahli, T.; et al. Phylogeny of the Viral Hemorrhagic Septicemia Virus in European Aquaculture. PLoS ONE 2016, 11, e0164475. [Google Scholar] [CrossRef]
  11. Einer-Jensen, K.; Ahrens, P.; Forsberg, R.; Lorenzen, N. Evolution of the fish rhabdovirus viral haemorrhagic septicaemia virus. J. Gen. Virol. 2004, 85, 1167–1179. [Google Scholar] [CrossRef]
  12. Kahns, S.; Skall, H.; Kaas, R.; Korsholm, H.; Bang Jensen, B.; Jonstrup, S.; Dodge, M.; Einer-Jensen, K.; Stone, D.; Olesen, N. European freshwater VHSV genotype Ia isolates divide into two distinct subpopulations. Dis. Aquat. Organ. 2012, 99, 23–35. [Google Scholar] [CrossRef] [PubMed]
  13. Stepien, C.A.; Pierce, L.R.; Leaman, D.W.; Niner, M.D.; Shepherd, B.S.; Chen, T.Y. Gene Diversification of an Emerging Pathogen: A Decade of Mutation in a Novel Fish Viral Hemorrhagic Septicemia (VHS) Substrain since Its First Appearance in the Laurentian Great Lakes. PLoS ONE 2015, 10, e0135146. [Google Scholar] [CrossRef] [PubMed]
  14. Guðmundsdóttir, S.; Vendramin, N.; Cuenca, A.; Sigurðardóttir, H.; Kristmundsson, A.; Iburg, T.M.; Olesen, N.J. Outbreak of viral haemorrhagic septicaemia (VHS) in lumpfish (Cyclopterus lumpus) in Iceland caused by VHS virus genotype IV. J. Fish Dis. 2019, 42, 47–62. [Google Scholar] [CrossRef] [PubMed]
  15. Marsella, A.; Pascoli, F.; Pretto, T.; Buratin, A.; Biasini, L.; Abbadi, M.; Cortinovis, L.; Berto, P.; Manfrin, A.; Vanelli, M.; et al. Efficacy of DNA Vaccines in Protecting Rainbow Trout against VHS and IHN under Intensive Farming Conditions. Vaccines 2022, 10, 2062. [Google Scholar] [CrossRef] [PubMed]
  16. Panzarin, V.; Cuenca, A.; Gastaldelli, M.; Alencar, A.L.F.; Pascoli, F.; Morin, T.; Blanchard, Y.; Cabon, J.; Louboutin, L.; Ryder, D.; et al. VHSV Single Amino Acid Polymorphisms (SAPs) Associated With Virulence in Rainbow Trout. Front. Microbiol. 2020, 11, 573993. [Google Scholar] [CrossRef] [PubMed]
  17. Ito, T.; Kurita, J.; Mori, K.-i.; Skall, H.F.; Lorenzen, N.; Vendramin, N.; Andersen, N.G.; Einer-Jensen, K.; Olesen, N.J. Virulence marker candidates in N-protein of viral haemorrhagic septicaemia virus (VHSV): Virulence variability within VHSV Ib clones. Dis. Aquat. Organ. 2018, 128, 51–62. [Google Scholar] [CrossRef] [PubMed]
  18. Kim, S.-H.; Thu, B.J.; Skall, H.F.; Vendramin, N.; Evensen, O. A Single Amino Acid Mutation (I1012F) of the RNA Polymerase of Marine Viral Hemorrhagic Septicemia Virus Changes In Vitro Virulence to Rainbow Trout Gill Epithelial Cells. J. Virol. 2014, 88, 7189–7198. [Google Scholar] [CrossRef] [PubMed]
  19. Ito, T.; Kurita, J.; Mori, K.I.; Olesen, N.J. Virulence of viral haemorrhagic septicaemia virus (VHSV) genotype III in rainbow trout. Vet. Res. 2016, 47, 4. [Google Scholar] [CrossRef]
  20. Vakharia, V.N.; Li, J.; McKenney, D.G.; Kurath, G. The Nucleoprotein and Phosphoprotein are Major Determinants of Virulence of Viral Hemorrhagic Septicemia Virus in Rainbow Trout. J. Virol. 2019, 93, e00382-19. [Google Scholar] [CrossRef]
  21. López-Vázquez, C.; Bandín, I.; Panzarin, V.; Toffan, A.; Cuenca, A.; Olesen, N.J.; Dopazo, C.P. Steps of the Replication Cycle of the Viral Haemorrhagic Septicaemia Virus (VHSV) Affecting Its Virulence on Fish. Animals 2020, 10, 2264. [Google Scholar] [CrossRef]
  22. Purcell, M.K.; Marjara, I.S.; Batts, W.; Kurath, G.; Hansen, J.D. Transcriptome analysis of rainbow trout infected with high and low virulence strains of infectious hematopoietic necrosis virus. Fish Shellfish Immunol. 2011, 30, 84–93. [Google Scholar] [CrossRef] [PubMed]
  23. Aquilino, C.; Castro, R.; Fischer, U.; Tafalla, C. Transcriptomic responses in rainbow trout gills upon infection with viral hemorrhagic septicemia virus (VHSV). Dev. Comp. Immunol. 2014, 44, 12–20. [Google Scholar] [CrossRef] [PubMed]
  24. Hwang, J.Y.; Markkandan, K.; Kwon, M.G.; Seo, J.S.; Yoo, S.i.; Hwang, S.D.; Son, M.H.; Park, J. Transcriptome analysis of olive flounder (Paralichthys olivaceus) head kidney infected with moderate and high virulent strains of infectious viral hemorrhagic septicaemia virus (VHSV). Fish Shellfish Immunol. 2018, 76, 293–304. [Google Scholar] [CrossRef] [PubMed]
  25. Jonstrup, S.P.; Kahns, S.; Skall, H.F.; Boutrup, T.S.; Olesen, N.J. Development and validation of a novel Taqman-based real-time RT-PCR assay suitable for demonstrating freedom from viral haemorrhagic septicaemia virus. J. Fish Dis. 2013, 36, 9–23. [Google Scholar] [CrossRef]
  26. Overturf, K.; Lapatra, S.; Powell, M. Real-time PCR for the detection and quantitative analysis of IHNV in salmonids. J. Fish Dis. 2001, 24, 325–333. [Google Scholar] [CrossRef]
  27. Ørpetveit, I.; Mikalsen, A.B.; Sindre, H.; Evensen, Ø.; Dannevig, B.H.; Midtlyng, P.J. Detection of Infectious pancreatic necrosis virus in sub clinically infected atlantic salmon by virus isolation in cell culture or real-time reverse transcription polymerase chain reaction: Influence of sample preservation and storage. J. Vet. Diagnostic Investig. 2010, 22, 886–895. [Google Scholar] [CrossRef]
  28. Reed, L.J.; Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 1938, 27, 493–497. [Google Scholar] [CrossRef]
  29. Baillon, L.; Mérour, E.; Cabon, J.; Louboutin, L.; Vigouroux, E.; Alencar, A.L.F.; Cuenca, A.; Blanchard, Y.; Olesen, N.J.; Panzarin, V.; et al. The Viral Hemorrhagic Septicemia Virus (VHSV) Markers of Virulence in Rainbow Trout (Oncorhynchus mykiss). Front. Microbiol. 2020, 11, 574231. [Google Scholar] [CrossRef]
  30. Hodneland, K.; Endresen, C. Sensitive and specific detection of Salmonid alphavirus using real-time PCR (TaqMan®). J. Virol. Methods 2006, 131, 184–192. [Google Scholar] [CrossRef]
  31. Toffan, A.; Biasini, L.; Pretto, T.; Abbadi, M.; Buratin, A.; Franch, R.; Dalla Rovere, G.; Panzarin, V.M.; Marsella, A.; Bargelloni, L.; et al. Age dependency of RGNNV/SJNNV viral encephalo-retinopathy in Gilthead Sea Bream (Sparus aurata). Aquaculture 2021, 539, 736605. [Google Scholar] [CrossRef]
  32. Bustin, S.A.; Benes, V.; Garson, J.A.; Hellemans, J.; Huggett, J.; Kubista, M.; Mueller, R.; Nolan, T.; Pfaffl, M.W.; Shipley, G.L.; et al. The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin. Chem. 2009, 55, 611–622. [Google Scholar] [CrossRef]
  33. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 2014, 30, 2114. [Google Scholar] [CrossRef] [PubMed]
  34. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15. [Google Scholar] [CrossRef] [PubMed]
  35. Anders, S.; Pyl, P.T.; Huber, W. HTSeq—A Python framework to work with high-throughput sequencing data. Bioinformatics 2015, 31, 166–169. [Google Scholar] [CrossRef] [PubMed]
  36. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed]
  37. Conesa, A.; Götz, S. Blast2GO: A comprehensive suite for functional analysis in plant genomics. Int. J. Plant Genom. 2008, 2008, 619832. [Google Scholar] [CrossRef] [PubMed]
  38. Ensembl. Available online: https://www.ensembl.org/index.html (accessed on 7 January 2021).
  39. Geneontology. Available online: http://geneontology.org/ (accessed on 7 January 2021).
  40. Van Belle, G.; Fisher, L.; Heagerty, P.J.; Lumley, T. Biostatistics: A Methodology for the Health Sciences, 2nd ed.; John Wiley & Sons: New York, NY, USA, 2004; ISBN 978-0-471-03185-7. [Google Scholar]
  41. Zapata, A.; Diez, B.; Cejalvo, T.; Gutiérrez-De Frías, C.; Cortés, A. Ontogeny of the immune system of fish. Fish Shellfish Immunol. 2006, 20, 126–136. [Google Scholar] [CrossRef]
  42. Bjørgen, H.; Koppang, E.O. Anatomy of teleost fish immune structures and organs. Immunogenetics 2021, 73, 53–63. [Google Scholar] [CrossRef]
  43. Kondera, E. Haematopoiesis and haematopoietic organs in fish. Anim. Sci. Genet. 2019, 15, 9–16. [Google Scholar] [CrossRef]
  44. Peris-Moreno, D.; Taillandier, D.; Polge, C. MuRF1/TRIM63, Master Regulator of Muscle Mass. Int. J. Mol. Sci. 2020, 21, 6663. [Google Scholar] [CrossRef]
  45. Ozato, K.; Shin, D.M.; Chang, T.H.; Morse, H.C. TRIM family proteins and their emerging roles in innate immunity. Nat. Rev. Immunol. 2008, 8, 849–860. [Google Scholar] [CrossRef] [PubMed]
  46. Li, K.; Pan, W.; Ma, Y.; Xu, X.; Gao, Y.; He, Y.; Wei, L.; Zhang, J. A novel oncogene TRIM63 promotes cell proliferation and migration via activating Wnt/β-catenin signaling pathway in breast cancer. Pathol. Res. Pract. 2019, 215, 152573. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, J.; Cho, M.; Lim, J.; Choi, H.; Hong, S. Pathogenic Mechanism of a Highly Virulent Infectious Hematopoietic Necrosis Virus in Head Kidney of Rainbow Trout (Oncorhynchus mykiss) Analyzed by RNA-Seq Transcriptome Profiling. Viruses 2022, 14, 859. [Google Scholar] [CrossRef] [PubMed]
  48. Li, Y.; Zhang, P.; Wang, C.; Han, C.; Meng, J.; Liu, X.; Xu, S.; Li, N.; Wang, Q.; Shi, X.; et al. Immune responsive gene 1 (IRG1) promotes endotoxin tolerance by increasing A20 expression in macrophages through reactive oxygen species. J. Biol. Chem. 2013, 288, 16225–16234. [Google Scholar] [CrossRef] [PubMed]
  49. Michelucci, A.; Cordes, T.; Ghelfi, J.; Pailot, A.; Reiling, N.; Goldmann, O.; Binz, T.; Wegner, A.; Tallam, A.; Rausell, A.; et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc. Natl. Acad. Sci. USA 2013, 110, 7820–7825. [Google Scholar] [CrossRef]
  50. Hall, C.J.; Boyle, R.H.; Astin, J.W.; Flores, M.V.; Oehlers, S.H.; Sanderson, L.E.; Ellett, F.; Lieschke, G.J.; Crosier, K.E.; Crosier, P.S. Immunoresponsive gene 1 augments bactericidal activity of macrophage-lineage cells by regulating β-oxidation-dependent mitochondrial ROS production. Cell Metab. 2013, 18, 265–278. [Google Scholar] [CrossRef]
  51. Wu, R.; Chen, F.; Wang, N.; Tang, D.; Kang, R. ACOD1 in immunometabolism and disease. Cell. Mol. Immunol. 2020, 17, 822–833. [Google Scholar] [CrossRef]
  52. Yin, S.; Han, D.; Tao, Y.; Xu, S.; Li, T.; Li, J.; Li, Y.; Huang, J.; Cao, X.; Yu, Y. The IRG1-Itaconate Axis Promotes Viral Replication Via Metabolic Reprogramming and Protein Prenylation. SSRN Electron. J. 2019, 19-00910, 54. [Google Scholar] [CrossRef]
  53. Zamperin, G.; Bianco, A.; Smith, J.; Bortolami, A.; Vervelde, L.; Schivo, A.; Fortin, A.; Marciano, S.; Panzarin, V.; Mazzetto, E.; et al. Heterogeneity of early host response to infection with four low-pathogenic H7 viruses with a different evolutionary history in the field. Viruses 2021, 13, 2323. [Google Scholar] [CrossRef]
  54. Park, J.Y.; Pillinger, M.H.; Abramson, S.B. Prostaglandin E2 synthesis and secretion: The role of PGE2 synthases. Clin. Immunol. 2006, 119, 229–240. [Google Scholar] [CrossRef]
  55. Gadducci, A.; Guerrieri, M.E.; Greco, C. Tissue biomarkers as prognostic variables of cervical cancer. Crit. Rev. Oncol. Hematol. 2013, 86, 104–129. [Google Scholar] [CrossRef] [PubMed]
  56. Macedo, J.; Silva, E.; Nogueira, L.; Coelho, R.; da Silva, J.; dos Santos, A.; Teixeira-Júnior, A.A.; Belfort, M.; Silva, G.; Khayat, A.; et al. Genomic profiling reveals the pivotal role of hrHPV driving copy number and gene expression alterations, including mRNA downregulation of TP53 and RB1 in penile cancer. Mol. Carcinog. 2020, 59, 604–617. [Google Scholar] [CrossRef] [PubMed]
  57. Amsler, L.; Verweij, M.C.; Defilippis, V.R. The Tiers and Dimensions of Evasion of the Type I Interferon Response by Human Cytomegalovirus. J. Mol. Biol. 2013, 425, 4857–4871. [Google Scholar] [CrossRef] [PubMed]
  58. Chandrasekharan, J.A.; Sharma-Walia, N. Arachidonic Acid Derived Lipid Mediators Influence Kaposi’s Sarcoma-Associated Herpesvirus Infection and Pathogenesis. Front. Microbiol. 2019, 10, 358. [Google Scholar] [CrossRef] [PubMed]
  59. Cui, Z.; Clark, K.J.; Kaufman, C.D.; Hackett, P.B. Inhibition of skiA and skiB gene expression ventralizes zebrafish embryos. Genesis 2001, 30, 149–153. [Google Scholar] [CrossRef] [PubMed]
  60. Takahata, M.; Inoue, Y.; Tsuda, H.; Imoto, I.; Koinuma, D.; Hayashi, M.; Ichikura, T.; Yamori, T.; Nagasaki, K.; Yoshida, M.; et al. SKI and MEL1 Cooperate to Inhibit Transforming Growth Factor-β Signal in Gastric Cancer Cells. J. Biol. Chem. 2009, 284, 3334–3344. [Google Scholar] [CrossRef] [PubMed]
  61. Deheuninck, J.; Luo, K. Ski and SnoN, potent negative regulators of TGF-β signaling. Cell Res. 2008, 19, 47–57. [Google Scholar] [CrossRef]
  62. Chen, Y.; Pirisi, L.; Creek, K.E. Ski protein levels increase during in vitro progression of HPV16-immortalized human keratinocytes and in cervical cancer. Virology 2013, 444, 100–108. [Google Scholar] [CrossRef]
  63. Li, X.; Zhang, R.; Luo, D.; Park, S.J.; Wang, Q.; Kim, Y.; Min, W. Tumor necrosis factor alpha-induced desumoylation and cytoplasmic translocation of homeodomain-interacting protein kinase 1 are critical for apoptosis signal-regulating kinase 1-JNK/p38 activation. J. Biol. Chem. 2005, 280, 15061–15070. [Google Scholar] [CrossRef]
  64. Overstreet, J.M.; Gifford, C.C.; Tang, J.; Higgins, P.J.; Samarakoon, R. Emerging role of tumor suppressor p53 in acute and chronic kidney diseases. Cell. Mol. Life Sci. 2022, 79, 474. [Google Scholar] [CrossRef]
  65. Aasen, T.; Leithe, E.; Graham, S.V.; Kameritsch, P.; Mayán, M.D.; Mesnil, M.; Pogoda, K.; Tabernero, A. Connexins in cancer: Bridging the gap to the clinic. Oncogene 2019, 38, 4429–4451. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, J.; Xu, T.; Yin, D.; Zhang, B.; Bai, J. The Regulatory Roles of MicroRNA in Effects of 2,2′4,4′-Tetrabromodiphenyl Ether (BDE47) on the Transcriptome of Zebrafish Larvae. PLoS ONE 2017, 12, e0169599. [Google Scholar] [CrossRef]
  67. Kansakar, U.; Gambardella, J.; Varzideh, F.; Avvisato, R.; Jankauskas, S.S.; Mone, P.; Matarese, A.; Santulli, G. miR-142 Targets TIM-1 in Human Endothelial Cells: Potential Implications for Stroke, COVID-19, Zika, Ebola, Dengue, and Other Viral Infections. Int. J. Mol. Sci. 2022, 23, 10242. [Google Scholar] [CrossRef] [PubMed]
  68. He, M.; Ding, N.Z.; He, C.Q. Novirhabdoviruses versus fish innate immunity: A review. Virus Res. 2021, 304, 198525. [Google Scholar] [CrossRef] [PubMed]
  69. Webster, J.D.; Vucic, D. The Balance of TNF Mediated Pathways Regulates Inflammatory Cell Death Signaling in Healthy and Diseased Tissues. Front. Cell Dev. Biol. 2020, 8, 543956. [Google Scholar] [CrossRef] [PubMed]
  70. Kim, M.S.; Kim, K.H. The role of viral hemorrhagic septicemia virus (VHSV) NV gene in TNF-α- and VHSV infection-mediated NF-κB activation. Fish Shellfish Immunol. 2013, 34, 1315–1319. [Google Scholar] [CrossRef] [PubMed]
  71. Kawai, T.; Akira, S. Toll-like Receptors and Their Crosstalk with Other Innate Receptors in Infection and Immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef]
  72. Keshavarz, M.; Solaymani-Mohammadi, F.; Namdari, H.; Arjeini, Y.; Mousavi, M.J.; Rezaei, F. Metabolic host response and therapeutic approaches to influenza infection. Cell. Mol. Biol. Lett. 2020, 25, 15. [Google Scholar] [CrossRef]
  73. Tanner, L.B.; Chng, C.; Guan, X.L.; Lei, Z.; Rozen, S.G.; Wenk, M.R. Lipidomics identifies a requirement for peroxisomal function during influenza virus replication. J. Lipid Res. 2014, 55, 1357–1365. [Google Scholar] [CrossRef]
  74. Cho, S.Y.; Kwon, Y.K.; Nam, M.; Vaidya, B.; Kim, S.R.; Lee, S.; Kwon, J.; Kim, D.; Hwang, G.S. Integrated profiling of global metabolomic and transcriptomic responses to viral hemorrhagic septicemia virus infection in olive flounder. Fish Shellfish Immunol. 2017, 71, 220–229. [Google Scholar] [CrossRef]
  75. Song, K.; Li, S. The Role of Ubiquitination in NF-κB Signaling during Virus Infection. Viruses 2021, 13, 145. [Google Scholar] [CrossRef] [PubMed]
  76. Walker, A.P.; Fodor, E. Interplay between Influenza Virus and the Host RNA Polymerase II Transcriptional Machinery. Trends Microbiol. 2019, 27, 398–407. [Google Scholar] [CrossRef] [PubMed]
  77. Qie, L.; Marcellino, D.; Herold, B.C. Herpes Simplex Virus Entry Is Associated with Tyrosine Phosphorylation of Cellular Proteins. Virology 1999, 256, 220–227. [Google Scholar] [CrossRef] [PubMed]
  78. Smail, D.A.; Snow, M. Viral haemorrhagic septicaemia. In Fish Diseases and Disorders; Viral, Bacterial and Fungal Infections; CABI: Wallingford, UK, 2011; Volume 3, pp. 110–142. [Google Scholar] [CrossRef]
  79. Luque, A.; González Granja, A.; González, L.; Tafalla, C. Establishment and characterization of a rainbow trout heart endothelial cell line with susceptibility to viral hemorrhagic septicemia virus (VHSV). Fish Shellfish Immunol. 2014, 38, 255–264. [Google Scholar] [CrossRef] [PubMed]
  80. Marana, M.H.; Schmidt, J.G.; Biacchesi, S.; Lorenzen, N.; Jørgensen, L.v.G. Zebrafish (Danio rerio) larvae as a model for real-time studies of propagating VHS virus infection, tissue tropism and neutrophil activity. J. Fish Dis. 2021, 44, 563–571. [Google Scholar] [CrossRef]
  81. Amaya, M.; Baranova, A.; Van Hoek, M.L. Protein prenylation: A new mode of host–pathogen interaction. Biochem. Biophys. Res. Commun. 2011, 416, 1–6. [Google Scholar] [CrossRef]
  82. Pronin, A.V.; Narovlyansky, A.N.; Sanin, A.V. New Approaches to the Prevention and Treatment of Viral Diseases. Arch. Immunol. Ther. Exp. 2021, 69, 10. [Google Scholar] [CrossRef]
  83. Marakasova, E.S.; Eisenhaber, B.; Maurer-Stroh, S.; Eisenhaber, F.; Baranova, A. Prenylation of viral proteins by enzymes of the host: Virus-driven rationale for therapy with statins and FT/GGT1 inhibitors. BioEssays 2017, 39, 1700014. [Google Scholar] [CrossRef]
Figure 1. Kaplan–Meier survival curves and qRT-PCR results. (a) Mortality challenge: survival curves of VHSV-L e VHSV-H in rainbow trout. Trial carried out by bath immersion. The y-axis reports the survival rate; the x-axis reports the observation period expressed as days post infection (dpi). Step curves represent the survival rate of challenged fish in each experimental group. (b) Transcriptomic challenge: qRT-PCR results. In the x-axis the results are grouped according to the sampling day post-infection (dpi). The y-axis shows viral target gene copy numbers (CN) detected in SPL or HK and reported on logarithmic scale. Values are expressed as means of normalized CN in 1 ng of total RNA ± SEM (n = 5). * = p-value < 0.001.
Figure 1. Kaplan–Meier survival curves and qRT-PCR results. (a) Mortality challenge: survival curves of VHSV-L e VHSV-H in rainbow trout. Trial carried out by bath immersion. The y-axis reports the survival rate; the x-axis reports the observation period expressed as days post infection (dpi). Step curves represent the survival rate of challenged fish in each experimental group. (b) Transcriptomic challenge: qRT-PCR results. In the x-axis the results are grouped according to the sampling day post-infection (dpi). The y-axis shows viral target gene copy numbers (CN) detected in SPL or HK and reported on logarithmic scale. Values are expressed as means of normalized CN in 1 ng of total RNA ± SEM (n = 5). * = p-value < 0.001.
Microorganisms 12 00057 g001
Figure 2. Principal component analysis of the normalized gene counts of the host genes. The x- and y-axes show the two dimensions that explain the overall amount of variance related to gene expression levels. Each replicate is represented by a colored numerical code based on the challenge group and the time elapsed since infection.
Figure 2. Principal component analysis of the normalized gene counts of the host genes. The x- and y-axes show the two dimensions that explain the overall amount of variance related to gene expression levels. Each replicate is represented by a colored numerical code based on the challenge group and the time elapsed since infection.
Microorganisms 12 00057 g002
Figure 3. Differentially expressed gene numbers in head kidney of rainbow trout infected with VHSV High or Low virulent strains. At each time point, the total number of genes is shown in a stacked bar chart. Red and blue colors represent up- and down-regulated genes, respectively.
Figure 3. Differentially expressed gene numbers in head kidney of rainbow trout infected with VHSV High or Low virulent strains. At each time point, the total number of genes is shown in a stacked bar chart. Red and blue colors represent up- and down-regulated genes, respectively.
Microorganisms 12 00057 g003
Figure 4. Number of common and specific genes observed after infection. Venn diagrams show: (a) the common and specific differentially expressed genes (DEGs) between different time points of VHSV-L challenged group; (b) VHSV-H challenged group; (c) between the important time points of both challenged groups. Percentages of common and specific genes reported in (a,b) are computed relative to the total number of DEGs in each time point, while in (c) they refer to the total number of DEGs overall the two infections at both time points considered.
Figure 4. Number of common and specific genes observed after infection. Venn diagrams show: (a) the common and specific differentially expressed genes (DEGs) between different time points of VHSV-L challenged group; (b) VHSV-H challenged group; (c) between the important time points of both challenged groups. Percentages of common and specific genes reported in (a,b) are computed relative to the total number of DEGs in each time point, while in (c) they refer to the total number of DEGs overall the two infections at both time points considered.
Microorganisms 12 00057 g004
Figure 5. GO enrichment for the L and H VHSV challenges. Dotplot representing the most specific enriched GO terms for all the analyzed time points in both infections. L1, L2, L5 refer to time points 1, 2, 5 dpi in the VHSV-L challenge; similarly, H1, H2, H5 refer to time points 1, 2, 5 dpi in the VHSV-H challenge. Statistically significant enrichments (FDR < 0.05) are presented, and the -Log FDR is shown.
Figure 5. GO enrichment for the L and H VHSV challenges. Dotplot representing the most specific enriched GO terms for all the analyzed time points in both infections. L1, L2, L5 refer to time points 1, 2, 5 dpi in the VHSV-L challenge; similarly, H1, H2, H5 refer to time points 1, 2, 5 dpi in the VHSV-H challenge. Statistically significant enrichments (FDR < 0.05) are presented, and the -Log FDR is shown.
Microorganisms 12 00057 g005
Table 1. Differentially expressed genes of rainbow trout head kidney in VHSV high virulent strain infected group.
Table 1. Differentially expressed genes of rainbow trout head kidney in VHSV high virulent strain infected group.
VHSV-H
Differential Gene Expression TrendGene NameGene DescriptionDatabase Identifier Number 1 dpi
log2FC
2 dpi
log2FC
5 dpi
log2FC
ZFIN: ZDB-GENE-IdUniprotNCBI: Gene IDHGNCmiRBase
Up-regulatedtrim63atripartite motif containing 63a040625-139Q6IQH8
Q969Q1
20.4418.4615.09
acod1aconitate decarboxylase 1081104-428B0UYM1
A6NK06
110501238 3.426.508.17
cox-2Oncorhynchus mykiss cyclooxygenase-2020530-2Q9W715
Q8JH43
P35354
100136025 2.626.673.44
skiav-ski avian sarcoma viral oncogene homolog a990715-9Q1ECW8
P12755
1.701.411.88
Hipk1bhomeodomain interacting protein kinase 1b140324-1A0A0R4INQ1
Q86Z02
1.071.091.45
Down-regulatedcx35.4
(or gjb3)
connexin 35.4 (or junction protein beta 3)050417-174 Q567J2
O75712
−17.21−14.68−14.43
mir142microRNA 142 31529MI0000458−3.12−2.84−5.14
ZN572-likezinc finger protein 572-like Q7Z3I7110537993 −1.14−1.34−1.11
Fluctuating-
regulated
hbbe2Oncorhynchus mykiss embryonic beta-type globin2 Q9YGF4100135792 −30.6924.9528.27
degs2-likesphingolipiddelta(4)desaturase/C4-monooxygenase DES2-like Q6QHC5110533418 −25.74−14.9624.86
vtg3vitellogenin-like991019-2 110521946 −23.89−24.7623.14
sox19btranscription factor Sox-19b-like010111-1Q9DDD7110500375 −23.8514.9922.09
s100a1S100 calcium binding protein A1040916-1A3KH13110487342 −22.0615.9218.80
gata4GATA binding protein 4980526-476 110522338 −21.7219.7618.52
QRFPRpyroglutamylated RFamide peptide receptor 15565 −21.0614.9019.12
epycepiphycan041008-9Q99645 −17.28−15.6614.58
zgc:110045poly(rC)-binding protein 3-like050706-149 110537993 −17.2314.87−14.61
col2a1acollagen, type II, alpha 1a980526-192Q2LDA1
B3DLK0
−14.7614.5116.17
tdrd7atudor domain containing 7 a040426-2103A6NAF9
Q8NHU6
406379 −10.8221.9324.81
avdavidin140205-1A0A8M1P1N5 −10.4418.229.14
si:dkeyp-46h3.6L-rhamnose- binding lectin CSL3-like060503-90A3KQ75110528627 −9.6117.009.69
lhx8aLIM/homeobox protein Lhx8-like031008-2Q6BDC3110508831 −8.8016.197.41
kcnk10apotassium channel subfamily K member 10-like041210-291A3QJX1110522533 −7.2616.806.92
tgfaprotransforming growth factor alpha-like040724-208Q7T011
P01135
110494101 −6.6013.765.39
lama3laminin subunit alpha-3-like050208-475A0A8M9Q7C5110521524 −6.114.814.63
anxa13lannexin A13-like050522-310 110509080 −4.183.013.30
rasl11bras-like protein family member 11B040426-793Q6P0U3110521663 −3.462.824.58
stap2bsignal-transducing adaptor protein 2-like040426-2540Q6NYF8110524344 −3.092.443.47
chgachromogranin A 110529746 −2.772.381.65
ptpn3protein tyrosine phosphatase non-receptor type 3030131-2934F1QLN8 −2.533.023.84
RND3rho-related GTP-binding protein RhoE P61587110501874671 −2.362.562.96
rapgefl1rap guanine nucleotide exchange factor-like 1131121-404E7FGN2
Q9UHV5
110486183 −2.313.976.18
exoc3l4exocyst complex component 3-like protein 4 Q17RC7110497811 −2.114.324.61
tmem106btransmembrane protein 106B-like Q9NUM4110486388 −1.651.562.98
Fluctuating-
regulated
spsb4aSPRY domain-containing SOCS box protein 4-like070911-3B0S5W3110508890 −1.341.712.70
mycbpC-Myc-binding protein-like 110496688 −1.291.862.41
rnf217probable E3 ubiquitin-protein ligase RNF217060503-400Q1LUI6110529563 1.43−1.46−2.80
tgm2lprotein-glutamine gamma-glutamyltransferase2-like050420-97A3KQ87110520123 17.71−19.2120.16
uoxuricase-like030826-24Q6DG85110521955 26.18−27.02−26.79
Table 2. Differentially expressed genes of rainbow trout head kidney in VHSV low virulent strain infected group.
Table 2. Differentially expressed genes of rainbow trout head kidney in VHSV low virulent strain infected group.
VHSV-L
Differential Gene
Expression Trend
Gene NameGene DescriptionDatabase Identifier Number1 dpi
log2FC
2 dpi
log2FC
5 dpi
log2FC
ZFIN: ZDB-GENE-IdUniprotNCBI: Gene ID
Up-regulatedinspreproinsulin980526-110O73727
P01308
3026220.3816.2716.99
Down-regulatedmtnr1aamelatonin receptor Mel1A a990415-155P51046
P48039
100135888−16.57−16.20−15.24
trim63aOncorhynchus mykiss muscle RING finger 1040625-139Q6IQH8
Q969Q1
100505417−15.47−17.53−14.95
Fluctuating-
regulated
uoxuricase-like030826-24Q6DG8511052195526.71−27.11−27.43
trim63atripartite motif containing 63a040625-139Q6IQH8
Q969Q1
17.84−18.3516.48
dok5docking protein 5-like Q9P10411052856514.46−14.7914.63
klhl41akelch-like family member 41a081105-22E9QIN8
O60662
100148315−16.59−15.3614.91
epycepiphycan041008-9Q99645 −16.99−15.7516.37
col2a1acollagen, type II, alpha 1a980526-192Q2LDA1
B3DLK0
−14.6714.5617.42
nkx2.5homeobox protein Nkx-2.5-like980526-321Q98872
P52952
110489113−14.7115.0415.91
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Biasini, L.; Zamperin, G.; Pascoli, F.; Abbadi, M.; Buratin, A.; Marsella, A.; Panzarin, V.; Toffan, A. Transcriptome Profiling of Oncorhynchus mykiss Infected with Low or Highly Pathogenic Viral Hemorrhagic Septicemia Virus (VHSV). Microorganisms 2024, 12, 57. https://doi.org/10.3390/microorganisms12010057

AMA Style

Biasini L, Zamperin G, Pascoli F, Abbadi M, Buratin A, Marsella A, Panzarin V, Toffan A. Transcriptome Profiling of Oncorhynchus mykiss Infected with Low or Highly Pathogenic Viral Hemorrhagic Septicemia Virus (VHSV). Microorganisms. 2024; 12(1):57. https://doi.org/10.3390/microorganisms12010057

Chicago/Turabian Style

Biasini, Lorena, Gianpiero Zamperin, Francesco Pascoli, Miriam Abbadi, Alessandra Buratin, Andrea Marsella, Valentina Panzarin, and Anna Toffan. 2024. "Transcriptome Profiling of Oncorhynchus mykiss Infected with Low or Highly Pathogenic Viral Hemorrhagic Septicemia Virus (VHSV)" Microorganisms 12, no. 1: 57. https://doi.org/10.3390/microorganisms12010057

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