Fish and Shell sh Immunology 157 (2025) 110092 Contents lists available at ScienceDirect Fish and Shellfish Immunology journal homepage: www.elsevier.com/locate/fsi CRISPR/Cas9-induced knockout of tumor necrosis factor-alpha-type I augments viral infection in zebrafish Arthika Kalaichelvan a,1 , Kishanthini Nadarajapillai a,1, Sarithaa Raguvaran Sellaththurai a , U.P.E. Arachchi a , Myoung-Jin Kim c , Sumi Jung a,b,** , Jehee Lee a,b,* a b c Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea Marine Life Research Institute, Gidang Marine Research Institute, Jeju National University, Jeju, 63333, Republic of Korea Nakdonggang National Institute of Biological Resources, Sangju-si, Gyeongsangbuk-do, 37242, Republic of Korea A R T I C L E I N F O A B S T R A C T Keywords: TNF-α1 Zebrafish CRISPR/Cas9 VHSV Antiviral immunity Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine with critical roles in inflammation, cell survival, and defense. As a member of the TNF superfamily, it exerts its effects by binding to transmembrane receptors and triggering various downstream signaling pathways. Although TNF-α′s involvement in antiviral responses in mammals is well-established, its role in teleost remains poorly understood. This study investigated the contri­ bution of TNF-α1 to antiviral immunity in zebrafish using a tnf-α1(− /− ) knockout (KO) line. We challenged both wild-type and tnf-α1(− /− ) zebrafish with viral hemorrhagic septicemia virus (VHSV) at both embryonic and adult stages. Mortality was observed at 4 days post-infection (dpi) in tnf-α1-deficient adult fish challenged with 5 × 106 TCID50 (VHSV) and at 5 dpi in adult wild fish challenged with the same concentration. In addition, tnf-α1(− /− ) KO adult fish reached the maximum mortality of 100 % at 20 dpi, whereas wild adult fish reached 54 % mortality at the same time point. This increased susceptibility to early mortality was associated with a higher viral burden and altered expression of key immune genes, including the pro-inflammatory cytokines il-6 and il-1β, the antiinflammatory cytokine il-10, and interferon-related genes such as irf1 and ifn-γ. Our findings demonstrate the crucial role of TNF-α1 in antiviral defense mechanisms in zebrafish and provide valuable insights into the functional conservation of TNF-α signaling across vertebrate species. This knowledge may contribute to the development of strategies to combat viral diseases in fish. 1. Introduction Tumor necrosis factor-alpha (TNF-α) is a pro-inflammatory cytokine that is mainly produced by macrophages, monocytes, T cells, and nat­ ural killer cells in response to pathogen invasions [1]. It was initially identified for its cytotoxic effects on tumor cells in rabbits and mice following endotoxin injection [2,3]. TNF-α, a member of the TNF su­ perfamily, plays a pivotal role in regulating a multitude of cellular functions, including cell proliferation, differentiation, survival, and death [4]. Its production depends on the involvement of multiple re­ ceptors, including Toll-like and T-cell receptors, amongst others [5]. It is normally produced as a transmembrane protein (tmTNF) and converted to a soluble form (sTNF) by TNF-converting enzyme (TACE) [6]. Once sTNF binds to TNF receptor type 1 (TNFR1), it activates key signaling pathways such as nuclear factor kappa B (NF-κB), nuclear factor of activated T-cells (NFAT), and activator protein 1 (including c-Jun and Fos) [7]. These signaling pathways contribute to the diverse functions of TNFα, including its role in antiviral immunity. Compelling evidence in­ dicates that TNF-α is crucial for combating a range of viruses in mam­ mals. For instance, human TNF-α has been shown to inhibit the replication of RNA viruses such as encephalomyocarditis virus (EMCV) and vesicular stomatitis virus (VSV) [8]. Furthermore, TNF-α exhibits antiviral activity against VSV and herpes simplex virus (HSV) infections in human cell lines [9]. Additionally, TNF-α inhibition facilitates the advancement or reactivation of several significant viral infections, including human immunodeficiency, varicella-zoster and Epstein-Barr viruses, cytomegalovirus, and human papillomavirus, in patients * Corresponding author. Marine Molecular Genetics Lab, Jeju National University, 102 Jejudaehakno, Jeju, 63243, Republic of Korea. ** Corresponding author. Marine Molecular Genetics Lab, Jeju National University, 102 Jejudaehakno, Jeju, 63243, Republic of Korea. E-mail addresses: tnal1004u@jejunu.ac.kr (S. Jung), jehee@jejunu.ac.kr (J. Lee). 1 These authors contributed equally to this work. https://doi.org/10.1016/j.fsi.2024.110092 Received 9 September 2024; Received in revised form 2 December 2024; Accepted 20 December 2024 Available online 21 December 2024 1050-4648/© 2024 Elsevier Ltd. All rights are reserved, including those for text and data mining, AI training, and similar technologies. A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 2.3. VHSV challenge experiment to WT and tnf-α1(− /− ) larvae undergoing TNF-α blocking therapy [10]. Moreover, TNF-α has demonstrated robust antiviral efficacy against avian, swine, and human influenza viruses [11]. Conversely, some studies have proposed that TNF-α may play a role in viral pathogenesis, whereby viruses can manipulate cellular ma­ chinery to evade the immune response and facilitate infection dissemi­ nation [12]. Notably, a previous study on zebrafish indicated that TNF-α is involved in the in vitro replication of the spring viremia of carp virus (SVCV) and increases the in vivo susceptibility of zebrafish to the virus while simultaneously increasing mRNA expression of antiviral genes [13]. Furthermore, although TNF-α produced by dendritic cells has been shown to regulate influenza virus infection, its excessive production can result in pathogenesis [14]. Thus, the dual role of TNF-α in antiviral immunity appears to either support viral clearance or exacerbate immunopathology, depending on the context [15]. Consequently, further research is required to elucidate its precise function in the context of viral pathogenesis. Viral hemorrhagic septicemia virus (VHSV) is a negative-stranded virus of the genus Novirhabdovirus that is highly infectious to a wide range of freshwater and marine fish species [16]. To date, VHSV isolates have been obtained from more than 82 different freshwater and marine species in Europe, North America, and North Asia [17]. Therefore, research on VHSV is crucial for developing effective defense mecha­ nisms against viral threats in aquaculture. Investigating antiviral genes in fish can provide insights into immunity and disease resistance, which is vital for enhancing fish health and optimizing aquaculture practices. This knowledge could also significantly mitigate the economic impact of disease outbreaks in the industry. We have previously successfully generated a tnf-α1(− /− ) zebrafish model and demonstrated its effect on Edwardsiella piscicida infection [18]. To further investigate the role of TNF-α in antiviral immunity, particularly in teleosts, this study investigated the impact of TNF-α1 deficiency in a tnf-α1(− /− ) zebrafish model challenged with VHSV. By examining the effects of TNF-α1 deletion at embryonic and adult stages, we aimed to elucidate its role in antiviral defense and enhance our un­ derstanding of immune responses in fish. In total, 100 embryos were selected and incubated at 28 ◦ C for 3 days to evaluate the antiviral immune response in WT and tnf-α1(− /− ) larvae. At 4 days post fertilization (dpf), the larvae were separated into 2 groups. The rVHSV and 1 X PBS were prepared with dextran, tetrame­ thylrhodamine (Invitrogen, Thermo Fisher Scientific). One group was injected with a concentration of ~300 TCID50 rVHSV/larva using a micro-injector, transferred to an incubator, and maintained at 18 ◦ C. The control group was injected with the same volume of 1 X PBS. Larval viability was recorded every 12 h post-infection (hpi). Fluorescence microscopy (40X, Leica Microsystems, Germany) was used to capture images, which were processed using Leica Application Suite X software (version 3.3). Larvae from each group were collected at 24 and 48 hpi, washed three times with 1 X PBS, snap-frozen in liquid nitrogen, and stored at − 80 ◦ C for RNA extraction. 2.4. Cumulative mortality percentage of WT and tnf-α1(− /− ) adult zebrafish upon VHSV injection WT and tnf-α1(− /− ) zebrafish were acclimatized at 15 ◦ C for 7 days prior to the analysis of mortality rates [21]. Three-month-old WT and mutant zebrafish, with an average standard length of 3 cm, were divided into three groups of 22 fish each. Group 1 received 5 μL 1 X PBS, whereas groups 2 and 3 received 5 μL VHSV (5 × 106 TCID50 and 1 × 106 TCID50, respectively) by intraperitoneal injection using a 36-gauge needle (HAMILTON, USA). Fish were maintained at 15 ◦ C after injection, and daily mortality was recorded. 2.5. VHSV copy number analysis To quantify VHSV replication, we measured the expression of the viral nucleocapsid (N) protein gene using qPCR. RNA was extracted from the internal organs of VHSV-infected zebrafish using TRIzol reagent (Thermo Fisher Scientific, USA) and reverse-transcribed into cDNA with random hexamer primers (TaKaRa, Japan). The resulting cDNA was diluted five-fold before qPCR analysis. N gene expression of VHSV was quantified by quantitative PCR (qPCR) using a Dice™ TP950 Real-Time Thermal Cycler System (Takara, Japan) using TB Green Premix Ex TaqII (Takara Bio Inc.) and specific primers. The qPCR mixture (20 μL) was prepared in nuclease-free water, consisting of 10 μL of TB Green Premix Ex TaqII (Takara Bio Inc.), 3 μL cDNA, and 0.3 μL of each forward and reverse primer. The VHSV copy number was calculated in 1 μg of total RNA using a standard curve as previously described [22]. The mean VHSV copy number for five individuals at each time point was then plotted. 2. Methodology 2.1. Zebrafish husbandry Wild-type (WT) and tnf-α1(− /− ) zebrafish were maintained as described previously [19]. The water used in the tanks was maintained at a constant pH (pH 6.8–7.5), conductivity (500–800 μS), and tem­ perature (28 ± 0.5 ◦ C) throughout the rearing period. The light/dark cycle of the zebrafish facility was 14:10 h. The fish were provided with the artemia diet at three intervals throughout the day. This study was reviewed and approved by the Animal Ethics Committee of Jeju Na­ tional University, Republic of Korea. 2.6. Cytokine and regulatory gene dynamics in WT and tnf-α1(− /− ) adult zebrafish upon VHSV injection To analyze the transcriptional regulation of downstream genes, 3month-old WT and tnf-α1(− /− ) zebrafish were acclimatized at 15 ◦ C and subjected to the following injections. The WT and mutant zebrafish were divided into three groups. Group 1 was maintained as an unin­ fected control, whereas groups 2 and 3 were injected with 5 μL 1 X PBS and 5 μL VHSV (5 × 105 TCID50), respectively. Internal organs (spleen, intestine, liver and kidney) were collected from each group (7 fish per group) at 1, 3, 5, 7, and 9 dpi. Tissues from each fish were immediately snap-frozen in liquid nitrogen and stored at − 80 ◦ C until RNA extraction. 2.2. Virus stock preparation The genotype IVa VHSV (FWando05, NCBI- FJ811900. 2) from infected olive flounder kidney tissue and recombinant VHSV-ΔNV-EGFP (donated by the Department of Aquatic Life Medicine and the Depart­ ment of Marine Biomaterials & Aquaculture, Pukyong National Uni­ versity, Busan, Republic of Korea) was cultured in fathead minnow (FHM) cells using Leibovitz L-15 medium (L-15; Sigma, USA) supple­ mented with 10 % fetal bovine serum (FBS) and 1 % penicillin/strep­ tomycin (Gibco, USA). Virus-treated cells were maintained at 20 ◦ C until cytopathic effects (CPE) reached approximately 80–90 %. Subsequently, the cell supernatant was collected, subjected to three cycles of freezethawing, and filtered through a 20 μm filter. The virus titer was deter­ mined using the TCID50 technique described by Lei et al. [20]. Virus stocks were then stored at − 80 ◦ C until required. 2.7. RNA extraction, cDNA synthesis, and RT-qPCR RNA was extracted from frozen tissues and larvae using TRIzol re­ agent (Thermo Fisher Scientific, USA). RNA concentrations were measured using a Multiskan GO microplate spectrophotometer (Thermo 2 Fish and Shell sh Immunology 157 (2025) 110092 A. Kalaichelvan et al. Fisher Scientific, USA) at 260 nm. cDNA from each sample was prepared using 3 μg of RNA and a PrimeScript™ II 1st Strand cDNA Synthesis Kit (Takara, Japan). The cDNA samples were diluted 30-fold and stored at − 20 ◦ C for PCR and qPCR analysis. The qPCR primers (Table 1) for gene expression analysis were designed according to the Guidelines for the Publication of Quantitative Real-Time PCR Experiments (MIQE) [23]. A 10 μL reaction mixture was prepared, containing 4 μL of cDNA template, 1 μL of a primer mixture with forward and reverse primers (10 pmol/μL each), and 5 μL of TaKaRa Ex Taq SYBR premix (2 × ). The RT-qPCR Dice™ TP950 Real-Time Thermal Cycler System (Takara, Japan) was used to analyze gene expression patterns. The qPCR consisted of initial denaturation at 95 ◦ C for 10 s, followed by 45 PCR cycles of denaturation at 95 ◦ C for 5 s, annealing at 58 ◦ C for 10 s, and extension at 72 ◦ C for 20 s. A melting curve analysis was performed with a cycle of 95 ◦ C for 15 s, 60 ◦ C for 30 s, and 95 ◦ C for 15 s. Gene expression data were analyzed using the Livak (2− ΔΔCt) method [24]. VHSV migration was examined by fluorescence microscopy at 36, 42, and 48 h post-injection in groups injected with rVHSV-ΔNV-EGFP (Fig. 1C). No significant differences were observed between groups at 24 h post-injection (data not shown). The observed mortality corre­ sponded to a significantly high rVHSV replication in the tnf-α1(− /− ) group at 42 and 48 h post-injection. GFP fluorescence was detected in the spleen, kidney, heart, gastrointestinal tract, and ocular mucosa of both WT and tnf-α1(− /− ) larvae. Morphological changes were also observed at later times. 3.2. Difference in cumulative percentage mortality of WT and tnf-α1(− /− ) adult zebrafish following VHSV injection WT and tnf-α1(− /− ) mutant fish were injected intraperitoneally with different concentrations of VHSV, and mortality was recorded daily to investigate the role of Tnf-α1 during viral infection. As shown in Fig. 2, the tnf-α1(− /− ) mutant and WT fish began to die at 4 and 5 dpi, respectively, when a high concentration (5 × 106 TCID50) of VHSV was injected intraperitoneally. At the high concentration of VHSV at 20 dpi, tnf-α1(− /− ) and WT fish achieved 100 % and 54 % mortality, respec­ tively. The maximum mortality rate at 1 × 106 TCID50 VHSV was 54 % for the tnf-α1(− /− ) mutant fish and 27 % for WT fish. However, the mortality rate of the tnf-α1(− /− ) mutant with a low concentration of VHSV was higher than the mortality rate of the WT with a high con­ centration of VHSV at 18 dpi, which persisted in later days as well. Both WT and tnf-α1(− /− ) fish survived when injected with 1 X PBS as a negative control. Although both types of fish showed no symptoms up to 3 dpi after VHSV injection, on 4 dpi, the VHSV-infected fish began to die and showed characteristic signs of VHSV infection such as bleeding and protruding eyes. The severity of the infection was greater in the tnf-α1(− / − ) mutant than in the WT, as evidenced by the hemorrhaging. This may be because the tnf-α1(− /− ) mutant fish were unable to clear the virus as quickly as the WT fish. This finding is consistent with a previous study in which TNF-α-deficient mice infected with an adenovirus (Ad) gene transfer vector showed a reduced humoral immune response to viral infection [25]. Similarly, another study using TNF-α-deficient mice concluded that TNF-α is essential for the expression of adhesion mole­ cules and the recruitment of leukocytes to sites of inflammation during viral myocarditis and that its absence results in an inability to effectively clear infectious agents [26]. 2.8. Statistical analysis Results are presented as the means of triplicate experiments. Stu­ dent’s t-tests were used to assess statistical significance in the down­ stream gene analysis. One-way analysis of variance (ANOVA) was used to analyze tissue-specific expressions. Significance was determined using a threshold of p < 0.05. Graphs were generated using GraphPad Prism software (version 8.0.2, GraphPad Software, Inc., CA, USA). 3. Results and discussion 3.1. VHSV injection into WT and tnf-α1(− /− ) zebrafish larvae The lack of an adaptive immune response in zebrafish larvae makes them an ideal model for studying innate immunity. Therefore, we administered 300 TCID50 rVHSV/larva into the yolk sacs of WT and tnfα1(− /− ) zebrafish larvae and monitored mortality every 12 h. No deaths occurred in the PBS-injected control groups, whereas both WT and tnfα1(− /− ) larvae started to show mortality at 60 h after rVHSV injection. However, tnf-α1(− /− ) mutants exhibited significantly higher mortality, reaching 80 % compared to 58 % in WT larvae by the end of the experiment (Fig. 1A). Viral copy numbers were determined 24 and 48 h after injection. The tnf-α1(− /− ) mutants had significantly higher viral copy numbers than WT larvae (Fig. 1B). These results are consistent with the increased mortality observed in the tnf-α1(− /− ) mutants, suggesting a correlation between increased viral replication and increased mortality in the absence of Tnf-α mediated immunity. 3.3. VHSV copy number analysis in WT and tnf-α1(− /− ) adult zebrafish upon VHSV injection To assess the impact of Tnf-α1 on VHSV infection in adult zebrafish, we measured VHSV copy numbers in tnf-α1(− /− ) and WT adult zebrafish following the VHSV challenge. The results demonstrated a significant difference in VHSV copy numbers between the groups (Fig. 3). The VHSV copy number in WT fish remained relatively stable from 3 dpi to 9 dpi and was consistently lower than that in tnf-α1(− /− ) mutants throughout this period. In contrast, the VHSV copy number in tnf-α1(− / − ) fish increased steadily from 1 dpi to 7 dpi. A significant peak in VHSV copy number was observed on 9 dpi in the tnf-α1(− /− ) group, indicating a substantial increase in viral replication at this later stage. Table 1 Primer sequences used in this study. Gene (Accession No) Primer Sequence (5′-3′) Application il-6 (NP_001248378.1) GTGAAGACACTCAGAGACGAGCAG GGTTTGAGGAGAGGAGTGCTGATC qPCR- F qPCR-R il-1β (AAQ16563.1) TGGACTTCGCAGCACAAAATG GTTCACTTCACGCTCTTGGATG qPCR- F qPCR-R il-10 (NP_001018621.2) GCGGGATATGGTGAAATGCAAGAGG TGAGGTGTGCCATAACGCAGTAGA qPCR- F qPCR-R irf-1 (NM_001040352.1) CTGCAGCACAAACACAGAGAAACTCC AGTCCTTCATCAGTCCAGCAGACG qPCR- F qPCR-R ifn-γ (NM_001040352.1) AGAGCTCAGGACGTATGCAGAAACG TATAGACACGCTTCAGCTCAAACAAAGCC qPCR- F qPCR-R VHSV nucleocapsid protein (KF477302) TGTCTCAGATCAGTGGGAAGTACGC GGACCTCAGCGACAAGTTCGG qPCR- F qPCR- R ef-1α (NP_571338.1) CTCCTCTTGGTCGCTTTGCT CCGATTTTCTTCTCAACGCTCT qPCR- F qPCR-R 3.4. Cytokine and regulatory gene dynamics in WT and tnf-α1(− /− ) adult zebrafish upon VHSV injection To assess the impact of TNF-α1 on the immune response to VHSV, we analyzed cytokine and regulatory gene dynamics in WT and tnf-α1(− /− ) zebrafish (Fig. 4). Our findings indicate that TNF-α1 knockout dysre­ gulates the immune response, leading to altered expression of pro- and anti-inflammatory cytokines and interferon-related genes. This dysre­ gulation likely contributes to the increased susceptibility to VHSV infection observed in the tnf-α1(− /− ) fish. Focusing on pro-inflammatory cytokines, we observed dynamic 3 A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 Fig. 1. Injection of VHSV into zebrafish larvae. (A) Zebrafish larvae at 4 dpf were injected with VHSV at York, and mortality was observed at 12 h intervals after injection. (B) VHSV copy number was measured in infected WT and tnf-α1(− /− ) larvae at different time points after injection. (C) A-I, A-II, and A-III represent different time points to observe virus tropism in WT larvae (36, 42, and 48 hpi, respectively). B-I, B-II, and B-III represent different time points for the observation of virus tropism in tnf-α1(− /− ) larvae (36, 42, and 48 hpi, respectively). Red indicates the distribution of dextran tetramethylrhodamine, and green indicates the migration of rVHSV. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.) changes in the expression of il-6 and il-1β. While both cytokines were upregulated following VHSV infection in the mutant fish, their temporal expression patterns differed significantly from those in WT fish. In WT zebrafish, il-6 was upregulated from 3 days post-infection (dpi) and peaked at 7 dpi. In contrast, tnf-α1(− /− ) mutants exhibited an earlier and continuous upregulation of il-6, starting at 1 dpi. This suggests a compensatory mechanism attempting to counteract the absence of TNFα. However, this compensatory response may be inadequate because of the lack of TNF-α′s priming effect on other immune pathways essential for effective antiviral defense. This observation highlights the multi­ faceted role of TNF-α in not only triggering an acute inflammatory response but also priming the immune system for a robust antiviral response. Furthermore, the interplay between TNF-α and IL-6 is welldocumented, as they induce each other’s production, thereby ampli­ fying the expression of pro-inflammatory genes [27,28]. The expression of the pro-inflammatory cytokine il-1β also showed distinct patterns in WT and tnf-α1(− /− ) fish. In WT fish, il-1β was upre­ gulated at 3 and 7 dpi but downregulated at 5 and 9 dpi, suggesting a dynamic regulation of its expression during VHSV infection. In contrast, tnf-α1(− /− ) mutants exhibited a continuous upregulation of il-1β from 1 dpi, reaching its peak at 9 dpi. This sustained upregulation may result from an altered signaling environment in the absence of TNF-α1, potentially favoring the activation of inflammasomes, particularly the NLRP3 inflammasome, which processes IL-1β into its mature form [29]. It is important to note that IL-1β signaling plays a complex role in the immune response. Although it contributes to host protection against viral infections, excessive or prolonged IL-1β signaling can also promote inflammation and immunopathology. Low levels of IL-1β signaling may favor viral persistence, whereas high levels can enhance pathogenic IL-17A-producing CD4+ T helper cell (Th17) responses [30]. For instance, research has demonstrated that IL-1β is a critical mediator of lung inflammation and damage during influenza infection in mice [31]. Therefore, the persistent upregulation of il-1β observed in tnf-α1(− /− ) mutants may exacerbate inflammation and contribute to their increased mortality during VHSV infection. To further investigate the potential imbalance in immune regulation 4 A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 Fig. 2. Percentage mortality of WT and tnf-α1(− /− ) adult zebrafish during VHSV infection. After intraperitoneal injection of WT and tnf-α1(− /− ) zebrafish with VHSV at two different doses (5 × 106 and 1 × 106 TCID50), the death rates were examined up to 20 dpi. To confirm that the VHSV spread caused the mortality of the WT and tnf-α1(− /− ) zebrafish, PBS-injected control groups were maintained. Fig. 3. VHSV copy number in WT and tnf-α1(− /− ) adult zebrafish following VHSV infection. An experimental setup in which WT and tnf-α1(− /− ) adult zebrafish were injected with 5 μL of VHSV at a concentration of 5 × 105 TCID50 per fish. VHSV N-protein was quantified by qPCR, and VHSV copy number was calculated for 1 μg of RNA isolated from each VHSV-injected zebrafish. Error bars represent the standard deviation of VHSV copy numbers of five zebrafish from the same group at the same time point. suggested by the pro-inflammatory cytokine data, we examined the expression of the anti-inflammatory cytokine il-10. Its expression in WT zebrafish was significantly upregulated from 3 to 5 dpi, indicating a response to viral infection. However, il-10 upregulation was observed in tnf-α1(− /− ) mutants as early as 1-day post-infection, with a pronounced increase at 9 dpi. The abrupt increase in il-10 expression in the mutants at 9 dpi may represent a compensatory mechanism to control inflam­ mation, consistent with the well-established anti-inflammatory role of IL-10. Although IL-10 can help control inflammation and prevent excessive tissue damage, it can also create an environment conducive to the survival of pathogens. This duality is highlighted by a previous study suggesting that elevated levels of IL-10 support mycobacterial survival in the host [32]. In addition, our study found an abrupt increase in virus copy number in tnf-α1(− /− ) mutants at 9 dpi, which correlates with the observed increase in IL-10 expression. This correlation suggests a potential link between elevated il-10 levels and viral replication in the absence of Tnf-α1 signaling. Given the interplay between TNF-α1 and interferon signaling, we subsequently analyzed the expression of irf1, a critical transcription factor in the antiviral response. irf1 expression was upregulated from 1 dpi and reached its maximum level at 9 dpi in both WT and mutants. However, at 9 dpi, irf1 expression levels were higher in WT fish compared to the tnf-α1(− /− ) mutants, indicating a more robust response in the presence of Tnf-α1. Additionally, irf1 expression in tnf-α1(− /− ) mutants remained at its basal level from 1 to 5 dpi. The limited IRF1 induction in tnf-α1(− /− ) mutants likely reflects the lack of TNF-α1 signaling. This could lead to a suboptimal antiviral response, as IRF1 is a crucial transcription factor that regulates interferon production and other immune responses [33]. Following our analysis of irf1, we next examined the expression of 5 A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 Fig. 4. Transcriptional modulation of antiviral genes following VHSV challenge in WT and tnf-α1(− /− ) adult zebrafish. The relative transcription of each gene was normalized to the respective PBS-injected control group and ef-1α of zebrafish. The relative transcription of genes following VHSV injection was represented as the fold value of the respective non-injected group. The error bars show the SD (n = 3) of three triplicates. ifn-γ, another crucial component of the antiviral response. Throughout the experiment, WT fish exhibited a fluctuating upregulation of ifn-γ. This reached a 50-fold increase in expression at 9 dpi compared to basal levels. Conversely, upregulation was observed in tnf-α1(− /− ) mutants from 3 dpi, reaching its maximum level at 9 dpi, albeit with a 10-fold expression compared to basal levels. In addition, no significant differ­ ence was observed in basal expression between WT and tnf-α1(− /− ) mutants (data not shown). These results indicate that ifn-γ expression is significantly higher and sustained in WT fish compared to tnf-α1(− /− ) mutants. Antigen-presenting cells, such as dendritic cells and macrophages, process viral antigens and present them to T cells [34,35]. This process may stimulate T cells to produce IFN-gamma, contributing to the increased expression observed at later stages in WT fish. Conse­ quently, this sustained high expression of ifn-γ in WT fish may contribute to a more effective antiviral response compared to mutants lacking tnf-α1 signaling. The reduced production of IFN-γ in the absence of Tnf-α1 is consis­ tent with previous findings that TNF-α and IFN-γ can synergistically enhance antiviral responses, such as by upregulating the expression of interferon-stimulated genes [9]. A previous study has highlighted that 6 A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 TNF-α and IFN-γ together can activate the signaling molecule, signal transducer, and activator of transcription 1 (STAT1) and increase the expression, DNA binding, and activity of the transcription factor – interferon regulatory factor 1 (IRF1) [36]. This synergy results in increased expression of interferon-stimulated genes, contributing to a more robust antiviral response. Taken together, these findings provide valuable insights into the immune response and the role of specific cy­ tokines and interferons in the context of VHSV infection in zebrafish, particularly in the absence of tnf-α1 signaling. This could have broader implications for understanding the patho­ genesis of viral diseases and the development of therapeutic strategies targeting TNF-α1 pathways. However, additional research is necessary to clarify the exact role of TNF-α1, especially considering the high mortality observed in tnf-α1(− /− ) mutant fish despite the activation of various immune-related genes in zebrafish. Further studies are also needed to explore the potential interplay between TNF-α1 and other immune pathways in the context of viral infections. [3] N. Matthews, J.F. Watkins, Tumour-necrosis factor from the rabbit. I. Mode of action, specificity and physicochemical properties, Br. J. Cancer 38 (1978) 302–309, https://doi.org/10.1038/bjc.1978.202. [4] M.S. Kim, K.H. Kim, The role of viral hemorrhagic septicemia virus (VHSV) NV gene in TNF-α- and VHSV infection-mediated NF-κB activation, Fish Shellfish Immunol. 34 (2013) 1315–1319, https://doi.org/10.1016/j.fsi.2013.02.026. [5] T. Duan, Y. Du, C. Xing, H.Y. Wang, R.-F. Wang, Toll-like receptor signaling and its role in cell-mediated immunity, Front. Immunol. 13 (2022) 812774, https://doi. org/10.3389/fimmu.2022.812774. [6] D.-I. Jang, A.-H. Lee, H.-Y. Shin, H.-R. Song, J.-H. Park, T.-B. Kang, S.-R. Lee, S.H. Yang, The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics, Int. J. Mol. Sci. 22 (2021) 2719, https://doi.org/10.3390/ijms22052719. [7] A. Yarilina, K. Xu, J. Chen, L.B. Ivashkiv, TNF activates calcium–nuclear factor of activated T cells (NFAT)c1 signaling pathways in human macrophages, Proc. Natl. Acad. Sci. 108 (2011) 1573–1578, https://doi.org/10.1073/pnas.1010030108. [8] G.H.W. Wong, D.V. Goeddel, Tumour necrosis factors α and β inhibit virus replication and synergize with interferons, Nature 323 (1986) 819–822, https:// doi.org/10.1038/323819a0. [9] E. Bartee, M.R. Mohamed, G. McFadden, Tumor necrosis factor and interferon: cytokines in harmony, Curr. Opin. Microbiol. 11 (2008) 378–383, https://doi.org/ 10.1016/j.mib.2008.05.015. [10] S.Y. Kim, D.H. Solomon, Tumor necrosis factor blockade and the risk of viral infection, Nat. Rev. Rheumatol. 6 (2010) 165–174, https://doi.org/10.1038/ nrrheum.2009.279. [11] S.H. Seo, R.G. Webster, Tumor necrosis factor alpha exerts powerful anti-influenza virus effects in lung epithelial cells, J. Virol. 76 (2002) 1071–1076, https://doi. org/10.1128/JVI.76.3.1071-1076.2002. [12] C.A. Benedict, C.F. Ware, Virus targeting of the tumor necrosis factor superfamily, Virology 289 (2001) 1–5, https://doi.org/10.1006/viro.2001.1109. [13] F.J. Roca, I. Mulero, A. López-Muñoz, M.P. Sepulcre, S.A. Renshaw, J. Meseguer, V. Mulero, Evolution of the inflammatory response in vertebrates: fish TNF-α is a powerful activator of endothelial cells but hardly activates phagocytes, J. Immunol. 181 (2008) 5071–5081, https://doi.org/10.4049/ jimmunol.181.7.5071. [14] J.R. Aldridge Jr., C.E. Moseley, D.A. Boltz, N.J. Negovetich, C. Reynolds, J. Franks, S.A. Brown, P.C. Doherty, R.G. Webster, P.G. Thomas, TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection, Proc. Natl. Acad. Sci. 106 (2009) 5306–5311, https://doi.org/10.1073/pnas.0900655106. [15] B.T. Rouse, S. Sehrawat, Immunity and immunopathology to viruses: what decides the outcome? Nat. Rev. Immunol. 10 (2010) 514–526, https://doi.org/10.1038/ nri2802. [16] N. Lorenzen, N.J. Olesen, C. Koch, Immunity to VHS virus in rainbow trout, Aquaculture 172 (1999) 41–61, https://doi.org/10.1016/S0044-8486(98)004438. [17] M. Cieslak, S.S. Mikkelsen, H.F. Skall, M. Baud, N. Diserens, M.Y. Engelsma, O.L. M. Haenen, S. Mousakhani, V. Panzarin, T. Wahli, N.J. Olesen, H. Schütze, Phylogeny of the viral hemorrhagic septicemia virus in European aquaculture, PLoS One 11 (2016) e0164475, https://doi.org/10.1371/journal.pone.0164475. [18] K. Nadarajapillai, S. Jung, S. Sellaththurai, S. Ganeshalingam, M.-J. Kim, J. Lee, CRISPR/Cas9-mediated knockout of tnf-α1 in zebrafish reduces disease resistance after Edwardsiella piscicida bacterial infection, Fish Shellfish Immunol. 144 (2024) 109249, https://doi.org/10.1016/j.fsi.2023.109249. [19] A. Avdesh, M. Chen, M.T. Martin-Iverson, A. Mondal, D. Ong, S. Rainey-Smith, K. Taddei, M. Lardelli, D.M. Groth, G. Verdile, R.N. Martins, Regular care and maintenance of a zebrafish (Danio rerio) laboratory: an introduction, J. Vis. Exp. 18 (2012) e4196, https://doi.org/10.3791/4196. [20] C. Lei, J. Yang, J. Hu, X. Sun, On the calculation of TCID50 for quantitation of virus infectivity, Virol. Sin. 36 (2021) 141–144, https://doi.org/10.1007/s12250-02000230-5. [21] B. Novoa, A. Romero, V. Mulero, I. Rodríguez, I. Fernández, A. Figueras, Zebrafish (Danio rerio) as a model for the study of vaccination against viral haemorrhagic septicemia virus (VHSV), Vaccine 24 (2006) 5806–5816, https://doi.org/10.1016/ j.vaccine.2006.05.015. [22] J.-O. Kim, W.-S. Kim, S.-W. Kim, H.-J. Han, J.W. Kim, M.A. Park, M.-J. Oh, Development and application of quantitative detection method for viral hemorrhagic septicemia virus (VHSV) genogroup IVa, Viruses 6 (2014) 2204–2213, https://doi.org/10.3390/v6052204. [23] S.A. Bustin, V. Benes, J.A. Garson, J. Hellemans, J. Huggett, M. Kubista, R. Mueller, T. Nolan, M.W. Pfaffl, G.L. Shipley, J. Vandesompele, C.T. Wittwer, The MIQE Guidelines: minimum information for publication of quantitative real-time PCR experiments, Clin. Chem. 55 (2009) 611–622, https://doi.org/10.1373/ clinchem.2008.112797. [24] K.J. Livak, T.D. Schmittgen, Analysis of relative gene expression data using realtime quantitative PCR and the 2− ΔΔCT method, Methods 25 (2001) 402–408, https://doi.org/10.1006/meth.2001.1262. [25] K.B. Elkon, C.-C. Liu, J.G. Gall, J. Trevejo, M.W. Marino, K.A. Abrahamsen, X. Song, J.-L. Zhou, L.J. Old, R.G. Crystal, E. Falck-Pedersen, Tumor necrosis factor α plays a central role in immune-mediated clearance of adenoviral vectors, Proc. Natl. Acad. Sci. 94 (1997) 9814–9819, https://doi.org/10.1073/pnas.94.18.9814. [26] H. Wada, K. Saito, T. Kanda, I. Kobayashi, H. Fujii, S. Fujigaki, N. Maekawa, H. Takatsu, H. Fujiwara, K. Sekikawa, M. Seishima, Tumor necrosis factor-α (TNFα) plays a protective role in acute viral myocarditis in mice: a study using mice lacking TNF-alpha, Circulation 103 (2001) 743–749, https://doi.org/10.1161/01. CIR.103.5.743. 4. Conclusion This study investigated the role of zebrafish Tnf-α1 during VHSV infection. In vivo experiments showed that Tnf-α1 is critical for survival, as tnf-α1(− /− ) fish had significantly lower survival rates as early as 4 dpi and a higher viral load compared to WT fish. Therefore, tnf-α1(− /− ) fish experienced more severe inflammation and mortality, likely due to impaired viral clearance. Additionally, the expression patterns of downstream genes following viral infection varied between WT and tnfα1(− /− ) fish. These findings highlight the critical role of Tnf-α1 in the immune response to viral infections and underscore its conserved function across vertebrate species. This knowledge not only enhances our understanding of TNF-α signaling in vertebrate immunity but also provides valuable insights for developing therapeutic strategies in aquaculture to combat infectious diseases. CRediT authorship contribution statement Arthika Kalaichelvan: Writing – original draft. Kishanthini Nadarajapillai: Conceptualization, Methodology, Investigation. Sar­ ithaa Raguvaran Sellaththurai: Methodology, Investigation. U.P.E. Arachchi: Writing – review & editing. Myoung-Jin Kim: Methodology, Investigation, Conceptualization. Sumi Jung: Writing – review & edit­ ing, Methodology, Investigation, Conceptualization. Jehee Lee: Writing – review & editing, Supervision, Resources, Project administration, Funding acquisition. Acknowledgement This research was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF), funded by the Ministry of Education (2019R1A6A1A03033553), and the Korea Insti­ tute of Marine Science and Technology Promotion (KIMST) funded by the Ministry of Oceans and Fisheries (RS-2022-KS221670). Data availability Data will be made available on request. References [1] B. Beutler, D. Greenwald, J.D. Hulmes, M. Chang, Y.C. Pan, J. Mathison, R. Ulevitch, A. Cerami, Identity of tumor necrosis factor and the macrophagesecreted factor cachectin, Nature 316 (1985) 552–554, https://doi.org/10.1038/ 316552a0. [2] E.A. Carswell, L.J. Old, R.L. Kassel, S. Green, N. Fiore, B. Williamson, An endotoxin-induced serum factor that causes necrosis of tumors, Proc. Natl. Acad. Sci. 72 (1975) 3666–3670, https://doi.org/10.1073/pnas.72.9.3666. 7 A. Kalaichelvan et al. Fish and Shell sh Immunology 157 (2025) 110092 central orchestrators of cytokine amplification during influenza virus infection, Cell 146 (2011) 980–991, https://doi.org/10.1016/j.cell.2011.08.015. [32] Y.V.N. Cavalcanti, M.C.A. Brelaz, J.K. de A. Lemoine Neves, J.C. Ferraz, V.R. A. Pereira, Role of TNF-alpha, IFN-gamma, and IL-10 in the development of pulmonary tuberculosis, pulm, Med 2012 (2012) 745483, https://doi.org/ 10.1155/2012/745483. [33] K. Honda, T. Taniguchi, IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors, Nat. Rev. Immunol. 6 (2006) 644–658, https://doi.org/10.1038/nri1900. [34] R.M. Steinman, Linking innate to adaptive immunity through dendritic cells, in: Innate Immun. To Pulm. Infect., 2006, pp. 101–113, https://doi.org/10.1002/ 9780470035399.ch9. [35] S. Yona, S. Gordon, From the reticuloendothelial to mononuclear phagocyte system – the unaccounted years, Front. Immunol. 6 (2015) 328, https://doi.org/10.3389/ fimmu.2015.00328. [36] C.M. Robinson, K.A. Shirey, J.M. Carlin, Synergistic transcriptional activation of indoleamine dioxygenase by IFN-γ and tumor necrosis factor-α, J. Interf. Cytokine Res 23 (2003) 413–421, https://doi.org/10.1089/107999003322277829. [27] C. Lee, J.-I. Oh, J. Park, J.-H. Choi, E.-K. Bae, H.J. Lee, W.J. Jung, D.S. Lee, K.S. Ahn, S.-S. Yoon, TNF α mediated IL-6 secretion is regulated by JAK/STAT pathway but not by MEK phosphorylation and AKT phosphorylation in U266 multiple myeloma cells, BioMed Res. Int. 2013 (2013) 580135, https://doi.org/ 10.1155/2013/580135. [28] G.K. Wollenberg, L.E. DeForge, G. Bolgos, D.G. Remick, Differential expression of tumor necrosis factor and interleukin-6 by peritoneal macrophages in vivo and in culture, Am. J. Pathol. 143 (1993) 1121–1130. [29] P. Halfmann, L. Hill-Batorski, Y. Kawaoka, The induction of IL-1β secretion through the NLRP3 inflammasome during ebola virus infection, J. Infect. Dis. 218 (2018) S504–S507, https://doi.org/10.1093/infdis/jiy433. [30] B.S. Kim, Y.-H. Jin, L. Meng, W. Hou, H.S. Kang, H.S. Park, C.-S. Koh, IL-1 signal affects both protection and pathogenesis of virus-induced chronic CNS demyelinating disease, J. Neuroinflammation 9 (2012) 217, https://doi.org/ 10.1186/1742-2094-9-217. [31] J.R. Teijaro, K.B. Walsh, S. Cahalan, D.M. Fremgen, E. Roberts, F. Scott, E. Martinborough, R. Peach, M.B.A. Oldstone, H. Rosen, Endothelial cells are 8