Next Article in Journal
The Roles of SPOP in DNA Damage Response and DNA Replication
Next Article in Special Issue
Multi-Omics Analysis Provides Novel Insight into Immuno-Physiological Pathways and Development of Thermal Resistance in Rainbow Trout Exposed to Acute Thermal Stress
Previous Article in Journal
Diverse Physiological Functions and Regulatory Mechanisms for Signal-Transducing Small GTPases
Previous Article in Special Issue
Role of Macrophages and Microglia in Zebrafish Regeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cytosolic Sensors for Pathogenic Viral and Bacterial Nucleic Acids in Fish

1
Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
2
Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Gakuenkibanadai-nishi 1-1, Miyazaki 889-2192, Japan
3
Kakdwip Research Centre of ICAR-Central Institute of Brackishwater Aquaculture, Kakdwip, South 24 Parganas, West Bengal 743347, India
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(19), 7289; https://doi.org/10.3390/ijms21197289
Submission received: 22 September 2020 / Accepted: 29 September 2020 / Published: 2 October 2020
(This article belongs to the Special Issue Fish Immunology 2.0)

Abstract

:
Recognition of the non-self signature of invading pathogens is a crucial step for the initiation of the innate immune mechanisms of the host. The host response to viral and bacterial infection involves sets of pattern recognition receptors (PRRs), which bind evolutionarily conserved pathogen structures, known as pathogen-associated molecular patterns (PAMPs). Recent advances in the identification of different types of PRRs in teleost fish revealed a number of cytosolic sensors for recognition of viral and bacterial nucleic acids. These are DExD/H-box RNA helicases including a group of well-characterized retinoic acid inducible gene I (RIG-I)-like receptors (RLRs) and non-RLR DExD/H-box RNA helicases (e.g., DDX1, DDX3, DHX9, DDX21, DHX36 and DDX41) both involved in recognition of viral RNAs. Another group of PRRs includes cytosolic DNA sensors (CDSs), such as cGAS and LSm14A involved in recognition of viral and intracellular bacterial dsDNAs. Moreover, dsRNA-sensing protein kinase R (PKR), which has a role in antiviral immune responses in higher vertebrates, has been identified in fish. Additionally, fish possess a novel PKR-like protein kinase containing Z-DNA binding domain, known as PKZ. Here, we review the current knowledge concerning cytosolic sensors for recognition of viral and bacterial nucleic acids in teleosts.

1. Introduction

The innate immune response relies on the recognition of evolutionarily conserved pathogen components, termed as pathogen-associated molecular patterns (PAMPs), through the germ line-encoded pattern recognition receptors (PRRs) [1]. Till now five major groups of PRRs have been discovered: (i) Toll-like receptors (TLRs), (ii) nucleotide-binding oligomerization domain (NOD)-leucin rich repeats (LRR)-containing receptors (NLRs), (iii) retinoic acid-inducible gene 1 (RIG-1)-like receptors (RLRs or RIG-1-like helicases-RLH) which belongs to the large family of DExD/H-box RNA helicases, (iv) C-type lectin receptors (CLRs) and (v) cytosolic DNA sensors [2,3,4,5,6,7]. They can be found associated to subcellular compartments–cellular and endosomal membranes, in the cytosol, as well as extracellularly and in secreted forms present in the bloodstream and interstitial fluids [8].
Interestingly, pathogens/PAMPs of quite different biochemical composition, structure and location can be recognized by slightly different yet surprisingly similar and overlapping mechanisms by limited range of host PRRs [1]. Moreover, in specific cases, PRRs also recognize self-danger signals (DAMPs, damage/danger associated molecular patterns), which are present in abnormal locations or atypical molecular complexes as a consequence of infection/inflammation, or cellular stress [9,10].

2. Viral and Bacterial PAMPs and Their PRRs

PAMPs are molecular structures (glycoproteins, lipopolysaccharides (LPS), peptidoglycans (PGN), and nucleic acid motifs) characterized by being invariant among entire classes of pathogens, essential for the survival of the pathogen, and distinguishable from “self” [2]. Viruses possess several structurally diverse PAMPs, including surface glycoproteins, DNA, and different kinds of RNA including viral double-stranded RNA (dsRNA) and single-stranded RNA (ssRNA) [11]. They can be present in the infecting virion or may be produced during viral replication.
Main PRR family recognizing viral glycoproteins are membrane TLRs such as TLR2 and TLR4. Thus, TLR2 has been shown to recognize for example the hemagglutinin (HA) protein of measles virus [12], while TLR4 recognizes fusion protein of respiratory syncytial virus (RSV) and envelope proteins of murine leukemia virus [13,14]. In turn, ssRNA oligonucleotides containing guanosine- and uridine-rich sequences from RNA viruses are recognized by endosomal TLR7/8, while dsRNA is recognized by TLR3 which in human is localized in endosomal membrane or cell surface. In turn endosomal TLR9 binds bacterial and viral CpG DNA [11]. Moreover, dsRNA (short), 5′-triphosphate RNA and dsRNA (long, at least 2 kbp) are recognized by RLRs (RIG-I and MDA5, respectively), whereas DAI and AIM2, from cytosolic DNA sensor family of receptors, recognize viral DNA localized in cytoplasm [2]. Recently, also NLRs have been shown to recognize viral PAMPS e.g., NLRC2 was found to recognize ssRNA from RSV, influenza A virus, and parainfluenza virus [15].
One of the most well-known bacterial PAMPs is lipopolysaccharides (LPS) of Gram-negative bacteria recognized by Toll-like receptor 4 (TLR4), however many other Gram-negative bacteria PAMPs have been described. They include peptidoglycans, flagellin, porins, unmethylated CpG DNA derived from bacterial genomes and classical B-form double-stranded (ds) DNA present in the cytosol [4]. In turn, the major PAMPs of Gram-positive bacteria are: glycolipid lipoteichoic acid anchored in the cytoplasmic membrane as well as lipoproteins embedded in the bacterial cell wall, and similarly to those of Gram-negative bacteria are: peptidoglycans (e.g., peptidoglycan-derivative muramyl dipeptide, MDP) and CpG DNA [16]. Bacterial DNA is mainly recognized by endosomal TLR9, cytoplasmic NLRP3 as well as RIG-I, AIM2-like receptors and DNA helicases, while peptidoglycans bind to TLR2 and activate NLRs (NOD2 and NALP1/3). Moreover, TLR2 recognizes lipoteichoic acid and lipoproteins of Gram-positive bacteria [11,17], while flagellin of the flagellated Gram-negative bacteria, activates TLR5 [18] and NLRC4 (also known as IPAF) [19].
Interestingly many fish species possess a higher number of functional TLR genes as compared to human (10 TLRs) or mouse (12 TLRs). In case of some TLRs it is due to presence of duplicated copies of TLRs in fish genome but there are also several fish-specific TLRs not present in mammals. These are for example TLR22 which is present on the cell surface and sense dsRNA outside the cells or endosomal TLR21, present also in chicken, sensing CpG-oligodeoxynucleotides [20,21]. Since the essential immune mechanisms, receptors and pathways are often well conserved in vertebrates, teleost fish represent a relevant model for the study of the core immune mechanisms activated by viral infections. As presence, function and evolution of Toll-like receptors in fish has been well described previously [20,22,23,24], in the present review we mainly focus on DExD/H-box RNA helicases and intracellular DNA sensors that recognize viral and bacterial nucleic acids in cytosol.

3. DExD/H-Box RNA Helicases

DExD/H-box RNA helicases are conserved proteins present in all eukaryotic and prokaryotic cells but only in a minority of virus families [25]. The DExD/H-box RNA helicases consist of at least eight conserved motifs (I, Ia, Ib, II, III, IV, V, VI) which are involved in ATP binding and hydrolysis (motifs I, II and VI), nucleic acid binding (motifs Ia, Ib, IV and V) and helicase activity (motif III) [26,27,28]. Additionally, DEAD-box, but not DEAH-box helicases, have motif Q, which is necessary for the efficient binding of ssRNA as well the conformation changes that are driven by nucleotide binding and ATP hydrolysis [26].
All described conserved motifs of the DExD/H-box RNA helicases are clustered in a central core region that contains about 350 to 400 amino acids [26]. Crystal structural analysis indicated that DExD/H-box RNA helicases have two covalently linked RecA-like globular domains, each containing five β-strands surrounded by five α-helices [29]. Motifs Q, I, Ia, Ib, II and III belong to RecA-like domain 1, while motifs IV, V, and VI are present in RecA-like domain 2 [26].
DExD/H-box RNA helicases are involved in a variety of biological processes. They are implicated in all aspects of RNA metabolism, including transcription, splicing, nuclear export of mRNA, ribosome biogenesis, RNA turnover, initiation of translation, modulation of complex RNA structures and RNA degradation [30,31,32]. Many of them are multifunctional and have additional diverse roles, for example, in cell cycle regulation [33], regulation of apoptosis [34,35], cancer development/progression [36,37] and in the antiviral innate immune response. The main DExD/H-box RNA helicases involved in sensing of viral RNAs are retinoic acid-inducible gene I (RIG-I)-like receptors [38,39].

3.1. RIG-I-Like Receptors (RLR)

3.1.1. Receptor Structure

RIG-I-like receptors include three members of cytosolic receptors: (i) retinoic acid-inducible gene I product (RIG-I) also known as DEAD box polypeptide 58 (DDX58), (ii) melanoma differentiation-associated antigen 5 (MDA5) also known as interferon induced with helicase C domain 1 (IFIH1), and (iii) laboratory of genetics and physiology 2 (LGP2) also known as DExH box polypeptide 58 (DHX58) [39].
All three proteins possess a C-terminal domain (CTD) which is responsible for binding of viral RNA, however in unbound form it functions as a repressor domain (RD) to keep the receptor in an inactive conformation. The CTD of RIG-I and MDA5 possess a positively charged groove that binds RNA and is structurally different between the two proteins. This might explain the differences in binding of RNA ligands between RIG-I and MDA5 [40]. Next, there is a flexible hinge region and the DExD/H-box RNA helicase region that consists of the RecA-like Hel1 and Hel2 domains with ATP binding and hydrolyzing activity. This receptor region is connected by another flexible hinge region to two N-terminal caspase activation and recruitment domains (CARDs), which are not present in LGP2 [37,40] (Figure 1A). CARD domains are important for their signaling by interacting with a downstream adaptor molecule-mitochondrial antiviral-signaling protein (MAVS), also known as IFNβ-promoter stimulator-1 (IPS-1), virus-induced signaling adapter (VISA), or CARD adapter inducing interferon-beta (CARDIF) [41].
In the absence of viral RNA, RIG-I and MDA5 exist in cytoplasm of the cell in a phosphorylated and “auto-inhibited” conformation where CARD domains are not accessible. In this inactivated conformation, the CARD1 domain of RIG-I is phosphorylated at residue S8, whereas the CARD2 domain is phosphorylated at residue T170 by protein kinase C-α/β (PKC-α/β) [40]. Additionally, the CTD of RIG-I is phosphorylated at residues S854, S855, and T770 by casein kinase β (CKβ) [40]. The CTD of RIG-I is also acetylated at residue K909, and its deacetylation is necessary for activation of RIG-I [42]. In contrast, the MDA5 is phosphorylated only at the CTD at residue S828 by RIO kinase 3 (RIOK3) and also by different kinases that are still unknown [40].
After ligand binding, RIG-I unfolds into an open and activated state [40]. Open conformation is made due to changes in the flexible hinge regions between CARD domains and the helicase domain as well as between helicase and CTD domain [40]. In contrast, MDA5 exists in a conformational balance between open and closed states. However, closed conformation is favored during the absence of ligand [40] (Figure 1A).

3.1.2. Ligands

RLRs recognize various types of viral RNAs, and synthetic double-stranded RNA (poly(I:C); polyriboinosinic:polyribocytidylic acid) [7]. RIG-I has the highest affinity to short 5′tri-phosphorylated dsRNA [40]. It can also bind 5′di-phosphorylated dsRNA, 5′tri-phosphorylated ssRNA with a polyuridine signature and low-molecular-weight poly(I:C) (around 300 bp) [2,40,43]. The binding of 5′mono-phosphorylated dsRNA by RIG-I is uncertain since some of the studies demonstrated that 5′monophosphate dsRNA did not activate RIG-I [44] while other studies have shown that RIG-I can bind 5′monophosphate dsRNA to a certain degree [45]. MDA5 preferentially binds long dsRNA (>3 kb) and high-molecular-weight poly(I:C) (around 4–8 kbp) [40,43,46]. It has been observed that RIG-I can recognize viral nucleic acids of positive-strand RNA viruses like Japanese encephalitis virus, but also negative-stranded RNA viruses like influenza viruses, bunyaviruses, filoviruses, and rhabdoviruses [40]. On the other hand, MDA5 mainly recognizes positive-stranded RNA viruses, such as picornaviruses and arteriviruses [40]. Despite the fact that LGP2 does not contain the N-terminal CARD, it still can bind varied dsRNAs irrespective of 5′-PPP or RNA length [47].

3.1.3. Signal Transduction

RLRs are broadly expressed in most tissues where they are responsive for signal transduction and activation of antiviral response in a variety of cell types [41]. Ligand binding results in receptor dephosphorylation and ubiquitination [3]. Following dephosphorylation of the RIG-I CTD, this domain is ubiquinated at residues K849 and K851 by the E3 ubiquitin ligase RIPLET. Similarly, dephosphorylation of the RIG-I CARD domain led to polyubiquitination of this domain at residue K172 by the E3 TRIM25 ubiquitin ligase [40]. It has been shown that RIG-I complexes with ubiquitin are strong inducers of type I IFNs [48]. After activation, RIG-I receptor oligomerizes with other RIG-I/dsRNA complexes to form helical oligomers and MDA5 oligomerizes to form long RNA-associated filaments [40]. When the receptors are activated and oligomerized, the CARD domain becomes exposed and interacts with CARD-containing adaptor molecule MAVS [2]. MAVS is located on the outer membrane of the mitochondria and coordinates the activation of two signaling pathways. It can activate the kinases TBK1 and IKK-ε which phosphorylate interferon-regulatory factor 3 (IRF3) and 7 (IRF7). The phosphorylated IRF3 and IRF7 dimerize and translocate to the nucleus to initiate transcription of type I IFNs which upon secretion signal in autocrine and paracrine fashion to induce expression of the interferon-stimulated genes (ISGs) among which there are many antiviral proteins, such as Mx proteins, viperin and ISG-15. MAVS can also activate the kinase complex (IKK-α, IKK-β, IKK-γ/NEMO) which leads to the phosphorylation and subsequent degradation of IκB, which in the unphosphorylated form is coupled to NF-κB. This results in NF-κB translocation to the nucleus and activation of the expression of the genes encoding pro-inflammatory cytokines [2,3,7,49] (Figure 1A).
LGP2 does not possess signaling CARD domains and its role in antiviral response induction is unclear. Strong activation of LGP2 expression occurs upon viral infection, poly(I:C) stimulation and IFNα treatment [47]. It is intriguing that in various experiments, LGP2 receptor has been shown to function as a positive or negative regulator of RLR-mediated signaling. For example, Yoneyama and co-workers [50] reported that LPG2 acts as a negative regulator of RIG-I/MDA5-mediated signaling, by interfering with the viral RNA recognition by RIG-I and MDA5. Another report suggested that LGP2 inhibits antiviral activation by engaging in a protein complex with MAVS [51,52]. In contrast, studies on LGP2-deficient mice indicated that lack of this gene resulted in deficient type I IFN response [53,54].

4. RLRs in Fish

The orthologs of RLRs have been identified from many fish species and fish cell lines, and their constitutive expression was demonstrated [22,43] (Table 1). Interestingly, RIG-I has been identified mainly in Cypriniformes, Siluriformes, and Salmoniformes [43], but is absent in fish of superorder Acanthopterygii, such as medaka (Oryzias latipes), Japanese pufferfish (Takifugu rubripes), tetradon (Tetradon nigroviridis), three-spined stickle-back (Gasterosteus aculeatus), European seabass (Dicentrarchus labrax), gilt-head seabream (Sparus aurata), and mandarin fish (Chinese perch; Siniperca chuatsi) [43]. It is unclear why RIG-I has been lost in some fish species but this gene does not occur also in some other higher vertebrate species, for example in chicken [55], and Chinese tree shrew [56].
RLRs in fish and mammals are structurally similar at the protein level [22]. However, some differences can be found at the genetic level in the exon/intron organization. For example, Japanese flounder (Paralichthys olivaceus) LGP2 gene contains 12 exons, while human LGP2 contains 14 exons [57]. Furthermore, LGP2 of grass carp (Ctenopharyngodon idella) possesses one more α-helix domain in the RD, compared with that in human LGP2 [58].

4.1. RIG-I (DDX58)

Intracellular localization of RIG-I demonstrated its presence in the cytoplasm of fish cells [70,73]. Up-regulation of the expression of rig-I after viral infection or poly(I:C) stimulation in cell lines (in vitro studies) or various organs of different fish species (in vivo studies) has been demonstrated and is summarized in Table 1. Moreover, different RIG-I isoforms were identified in several fish species. For example, rig-I in zebrafish (Danio rerio) possesses four different transcripts: rig-Ia, rig-Ib, rig-Ic and rig-Id [108]. Zebrafish rig-Ib encodes the typical form of RIG-I, while rig-Ia encodes protein with an insertion of 38 amino acids in the second CARD domain [73], rig-Ic encodes a protein that lacks the first 189–192 amino acid region just behind the second CARD domain, while rig-Id encodes a protein that lacks 2 amino acids just behind the second CARD domain, but has an insertion of 3 amino acids in the helicase domain [108]. Comparison of the constitutive expression of zebrafish rig-Ia and rig-Ib in the ZF4 cell line indicated a higher level (of about 20 folds) of rig-Ib expression [108]. Moreover, their expression was significantly up-regulated during spring viremia of carp virus (SVCV), and Edwardsiella tarda infection, although, the up-regulation of rig-Ib was to a lesser degree [73]. Two isofoms of RIG-I: AjRIG-Ib and AjRIG-Ibv were described in the Japanese eel (Anguilla japonica) [70]. Based on the structure, AjRIG-Ib represents the classical RIG-I form containing all characteristic domains, while AjRIG-Ibv is a truncated form that lacks C-terminal domain (CTD) [70]. The transcripts of both RIG-I isoforms were detected in all studied tissues/organs of the Japanese eel, with higher level of rig-Ib expression [70]. I.p. injection of Japanese eel with poly(I:C) induced up-regulation of the expression of both rig-Ib and rig-Ibv in all examined tissues/organs with more rapid up-regulation of rig-Ibv (as early as 8 h post-injection) than rig-Ib [70]. Two isoforms of the RIG-I have also been identified in gibel carp (Carassius gibelio) (RIG-Ia and RIG-Ib) and the genes encoding both isoforms were up-regulated during Carassius auratus herpesvirus (CaHV) infection in head kidney, spleen and liver [65].
Several functional studies demonstrated that knock-down of gene encoding RIG-I impairs the antiviral response in fish. Moreover, it was demonstrated that functional blockade of RIG-I by using dominant negative mutants of this receptor (RIG-I-DN) significantly attenuated in fish SVCV-induced activation of IFN promoters, and up-regulation of IFNs and ISGs in Epithelioma Papulosum Cyprini (EPC) cells, derived from fathead minnow (Pimephales promelas) [109]. In zebrafish ZF4 cell line, morpholino-induced knock-down of RIG-I resulted in reduction of the expression of group II type I IFNs (ifnφ2 and ifnφ3), but not group I type I IFNs (ifnφ1), upon nervous necrosis virus (NNV) infection [71]. Several groups studied the function of RIG-I using overexpression approach. Biacchessi and co-workers [110] demonstrated that overexpression of N-terminal fragment (first 275 aa) of RIG-I in EPC cell line resulted in the induction of expression of genes encoding type I IFN and several ISGs and provided cell protection from viral hemorrhagic septicemia virus (VHSV) infection. Moreover, the authors were able to detect the presence of IFN in supernatant collected from EPC cells transfected with expression vector for RIG-I Nter (coding N-terminus protein) and this supernatant was able to protect fresh EPC cells against VHSV infection [110]. Overexpression of the two isoforms of RIG-I of the Japanese eel (ajrig-ib or ajrig-ibv) in the EPC cells led to the activation of type I IFN promoter [70]. In case of zebrafish, luciferase assay demonstrated that transfection of EPC cells with vector encoding rig-Ib activated the type I IFN promoter, which was not the case for rig-Ia. Furthermore, overexpression of rig-Ib, but not rig-Ia, reduced the viral titer during SVCV infection in EPC cells [73]. Overexpression of rig-I in crucian carp (Carassius carassius) blastulae embryonic cells (CABs) also induced activation of type I IFN promoter as demonstrated by luciferase assay [111]. In turn, Chen and co-workers [112], using expression vectors, demonstrated the role of different domains of grass carp RIG-I in downstream signaling pathway in Ctenopharyngodon idella kidney cell lines (CIK) cells during grass carp reovirus (GCRV) infection and stimulation with various PAMPs of viral and bacterial origin. The CARDs and helicase domains were shown to play a role in signaling cascade after GCRV infection while the CARD domains were also involved in the induction of signal transduction upon poly(I:C) stimulation. Helicase domain mediated induction of signaling pathway upon LPS and PGN stimulation and CARDs domain strengthened this induction. In all cases, RD domain inhibited activation of the signal transduction. Interestingly, lack of CARDs domain showed positive modulation in RIG-I signal transduction [112].

4.2. MDA5

Up-regulation of the expression of mda5 after viral infection or poly(I:C) stimulation in cell lines (in vitro studies) or various organs of different fish species (in vivo studies) have been demonstrated and this data are summarized in Table 1. Similar to RIG-I, intracellular localization of MDA5 demonstrated its presence in the cytoplasm of fish cells [78,79,88,92].
In zebrafish, two isoforms of MDA5 (mda5a and its shorter splicing variant mda5b) were identified [92], of which mda5a gene consists of 16 exons and 15 introns, whereas mda5b does not have part of exons 11 and 13 and complete exon 12. Mda5b has a premature stop codon in exon 11 which results in lack of C-terminal regulatory domain (RD) embedded within the C-terminal domain (CTD) [92]. Both variants were significantly up-regulated in ZF4 cells during SVCV or E. tarda infection with mda5a showing a higher level of expression in comparison to mda5b [92].
A few functional studies demonstrated that inhibition of MDA5 impairs the antiviral response in fish. Zebrafish dominant-negative mda5 mutants lacking CARD domains were more susceptible to snakehead rhabdovirus (SHRV) infection and showed reduced ifnφ1 up-regulation during SHRV infection as compared to wild-type larvae. This effect was rescued by overexpression of mda5 [113]. Functional blockade of mda5 in the EPC cells, by using dominant negative mutants of MDA5 (MDA5-DN), significantly attenuated in fish activation of IFN promoters and up-regulation of IFNs and ISGs during SVCV infection [109]. Transient silencing of mda5 in Japanese flounder gill cells (FG) by siRNA technology resulted in reduced antiviral activity upon poly(I:C) stimulation [114]. The role of MDA5 in antiviral response of fish was also studied using the overexpression approach. Overexpression of crucian carp mda5 induced activation of type I IFN promoter in CABs cells [111] while overexpression of rainbow trout (Oncorhynchus mykiss) mda5 enhanced mx gene expression in rainbow trout gonad 2 cells (RTG-2) [78]. Moreover, overexpression of mda5 in hirame natural embryo (HINAE) cell line derived from Japanese flounder embryo, resulted in the inhibition of replication of hirame rhabdovirus (HIRRV), VHSV and infectious pancreatic necrosis virus (IPNV) [85]. Furthermore, HINAE cells with mda5 overexpression showed enhanced type I IFN response during VHSV infection as compared to the cells transfected with empty vector [85]. The overexpression of zebrafish mda5a and mda5b induced strong activation of type I IFN promoter in EPC cells with mda5a exhibiting stronger induction [92]. The protective effect of mda5 overexpression against viral infection was demonstrated by reduced cytopathic effect (CPE) in zebrafish liver cells (ZFL) upon SHRV infection and EPC cells upon SVCV or GCRV infection [79,113]. Other studies indicated that overexpression of orange-spotted grouper mda5 in Grouper spleen (GS) cells delayed the CPE progression during Singapore grouper iridovirus (SGIV) or red spotted grouper nervous necrosis virus (RGNNV) infection [88]. It also induced type I IFN and IFN-stimulated response element (ISRE) promoter activities as shown by reporter gene assay and enhanced the expression of irf3 and irf7. Furthermore, GS cells with mda5 overexpression exhibited higher expression of pro-inflammatory cytokines such as tnf-α (during SGIV and RGNNV infection) and il-8 (during RGNNV infection) as compared to cells transfected with empty plasmids [88]. Wan and co-workers [115] using the stable transfected CIK cell line demonstrated that grass carp MDA5 induced a stronger type I IFN response as compared to RIG-I during GCRV infection and poly(I:C) stimulation. Interestingly, the stable overexpression of rig-I decreased the expression level of endogenous mda5 while the opposite effect was not demonstrated [115]. Gu and co-workers [116] studied the function of MDA5 domains using different overexpression vectors and demonstrated that helicase domain was essential in response to GCRV infection and PGN stimulation. Moreover, CARD domain alone was sufficient to induce signaling cascade after LPS stimulation [116].

4.3. LGP2

LGP2 was identified in many fish species and its up-regulation of the expression after viral infection or poly(I:C) stimulation in cell lines (in vitro studies) or various organs of different fish species (in vivo studies) is summarized in Table 1. The intracellular localization of LGP2 demonstrated its distribution in the cytoplasm [78,98,99] and also in nucleus [117].
Two variants of LGP2 (LGP2a and alternatively spliced LGP2b) have been identified in rainbow trout where LGP2b is 54 amino acids shorter than LGP2a due to the presence of an unspliced intron at the 3′ end region of the ORF causing early termination of translation [78]. In zebrafish, three splicing variants of the LGP2 have been described, including a full-length LGP2 (with all 12 exons and encoding a 679-aa protein), and two truncating forms: LGP2v1 (lacking the exon 9 and encoding a 575-aa protein with incomplete DExDc domain) and LGP2v2 (lacking the exons 3 and 4 and encoding a 547-aa protein with incomplete HELIc domain) [102]. All three variants were up-regulated in ZFL cells during poly(I:C) stimulation and in immune-related organs of zebrafish during SVCV infection, showing differential expression patterns and higher expression level of lgp2 than lgp2v1 and lgp2v2 [102].
Similarly to the situation observed in mammals, there are many reports indicating contrary biological activities of fish LGP2 as a positive or negative regulator of antiviral response. For instance, it was demonstrated for EPC cells that overexpression of zebrafish LGP2, but not overexpression of LGP2v1 and LGP2v2, significantly inhibited SVCV replication and expression of viral genes through induction of type I IFN and ISGs expression [102]. It was therefore concluded that LGP2 alone as well as under stimulation with low concentrations of poly(I:C) and at the early stages of SVCV infections functions as a positive regulator of IFN type I response, however, with the increasing concentrations of poly(I:C) or SVCV titer and consequent to the robust type I IFN response, it switches to negative function. LGP2v1 and LGP2v2, however, demonstrate only the inhibitory functions [102]. Ohtani and co-workers [57] demonstrated that overexpression of Japanese flounder LGP2 in HINAE cells has protective effect upon VHSV and HIRRV infection, while transfection of this cells with LGP2ΔRD failed to protect them from VHSV infection and provided only slight protection against HIRRV infection [57]. These studies also demonstrated that overexpression of full-length LGP2 in HINAE cells resulted in significant induction of type I IFN and ISGs (mx, isg15, and isg56) as compared to the cells transfected with LGP2ΔRD and empty plasmid during VHSV infection at 24 hpi [57]. In rainbow trout overexpression of lgp2a, but not lgp2b, in RTG-2 cells confers up-regulation of the expression of mx and enhances protection against VHSV [78]. Interestingly, an inhibitory effect of LGP2b on LGP2a was demonstrated [78]. It has been also shown in rainbow trout that both LGP2a and LGP2b, and MDA5 can bind poly(I:C) [78]. Chen and co-workers [118] described the contrary effect of grass carp lgp2 overexpression during GCRV infection and viral or bacterial PAMPs stimulation on endogenous rig-I and mda5 expression. Moreover, lgp2 overexpression confers its inhibitory effect on GCRV replication and therefore, positive role in anti-GCRV immune response [118]. The positive role of LGP2 in antiviral immune response was also demonstrated in miiuy croaker (Miichthys miiuy) [98] and black carp (Mylopharyngodon piceus) [79,95]. It is also suggested that LGP2 acts as a positive regulator in MDA5 mediated signaling during viral infections [79,118].
There are also various reports suggesting that fish LGP2 acts as a negative regulator in the activation of the RIG-I/MDA5 signaling pathway. Yu and co-workers [99] demonstrated that overexpression of orange-spotted grouper LGP2 in GS cells induced an enhanced CPE and increased expression of viral genes during RGNNV and SGIV infection as compared to the control cells. Overexpression of fish LGP2 caused a marked decreased expression of transcription factors (irf3, irf7, and nf-κB), IFNs and ISGs in unstimulated cells [99,119], as well as in cells stimulated with poly(I:C) [87], and during early phase of GCRV infection [119]. Furthermore, Rao and co-workers [119] demonstrated that knock-down of lgp2 increased phosphorylation levels of IRF3 and IRF7 and increased expression of type I IFNs in unstimulated cells but not in cells infected with GCRV. Interestingly LGP2 was shown to inhibit MAVS (IPS-1) activation by RIG-I and MDA5 via direct protein-protein interactions with both RLRs [119]. Moreover, in Nile tilapia (Oreochromis niloticus), LGP2 decreased MAVS-dependent NF-κB activation which suggests its negative regulatory effect on the MAVS gene [117].

5. Non-RLR DExD/H-box RNA Helicases

Besides RLR group, several non-RLR DExD/H-box RNA helicases have been described in mammals as proteins involved in sensing of viral genetic material and activation of downstream signaling pathways leading to type I interferon (IFN) production. They include: DDX1 [120], DDX3 [121,122], DHX9 [123], DHX15 [124], DDX17 [125], DDX21 [120], DDX23 [126], DHX29 [127], DHX36 [120], DDX41 [128] and DDX60 [129]. Interestingly, some of them, like DHX33, can also recognize bacterial RNA [130].
In mammals, various non-RLR DExD/H-box RNA helicases can recognize both pathogenic DNA and RNA and signal via distinct adaptor molecules (Figure 1A,B). For example, DDX1 was found to form a complex with two other DExD/H-box RNA helicases, DDX21 and DHX36, and in this complex, DDX1 directly binds to both short and long forms of poly(I:C) while DDX21 and DHX36 are responsible for signal transduction via TRIF adaptor molecule [120]. DHX9 was described as a cytoplasmic sensor for both dsRNA in murine myeloid dendritic cells [123] and CpG DNA in human plasmacytoid dendritic cells [131]. Depending on the nature of the ligand and the cell type, DHX9 induces signaling through MAVS adaptor molecule (upon dsRNA binding) or in a MyD88-dependent manner (upon DNA binding) [123,131]. Another RNA helicase, DDX3, was shown to interact with IKKε [121] or TBK1 [122] kinases and act as a signaling intermediate downstream of TBK1/IKKε in the IFN-β induction pathway. Moreover, activated by TBK1, DDX3 binds directly to the IFN-β promoter enhancer region, and functions as a transcriptional regulator [122]. Finally it was suggested that at the early stages of infection, DDX3 can also sensitize the RLR system for dsRNA ligands [132]. In mammals, DDX41 recognizes intracellular DNA derived from bacteria and induces the production of type I IFNs in myeloid dendritic cells (mDCs) in a STING-dependent manner [128,133].
Phylogenetic analysis of fish non-RLR DExD/H-box RNA helicases resulted in two main clusters: DEAD-box and DEAH-box [100,103,104,105]. Moreover, this analysis confirmed that motifs constituting these helicases were well conserved among species, however motif V was missing in DHX9 of common carp and in DHX32A and DHX32B of channel catfish [103,134]. Constitutive expression of all studied non-RLR DExD/H-box RNA helicases in different organs among studied fish species was demonstrated [100,103,104,105,107,135]. The intracellular localization of several non-RLR DExD/H-box RNA helicases was also studied. In trout RTG-2 cells, DDX3 was localized to the cytoplasm, while DHX9 was localized mainly in the nucleus [100]. DDX41 was localized in the nucleus of GS cells [107].
Compared to mammals, the role of non-RLR DExD/H-box RNA helicases in antiviral response of fish is poorly described. The expression of fish non-RLR DExD/H-box RNA helicases in response to DNA and RNA viruses, poly(I:C) stimulation as well as to bacterial infection and CpG was studied in different fish species [100,103,104,105,106,107]. The results of these studies are summarized in Table 1. Moreover, in rainbow trout, DDX3 and DHX9 were observed to bind dsRNA [100].
In fish, there are few functional studies of non-RLR DExD/H box RNA helicases. Ddx3 overexpression in GS cells resulted in enhanced type I IFN-related antiviral response and inhibition of replication of RGNNV but not SGIV [104]. Overexpression of Japanese flounder ddx41 induced activation of antiviral status of the HINAE cells upon C-di-GMP stimulation [106]. The overexpression of Nile tilapia DDX41 induced a strong activation of both zebrafish IFN1 and IFN3 promoters in EPC cells treated with poly(dA:dT) [136]. Furthermore, overexpression of ddx41 in GS cells inhibited SGIV and RGNNV replication as well as increased the gene expression of antiviral and pro-inflammatory cytokines [107]. These results suggest that DDX41 is involved in the type I IFN-mediated antiviral and inflammatory response in teleosts.

6. MAVS

So far many MAVS orthologs have been detected in teleost fish [89,91,110,117,118,137,138,139,140,141,142,143,144,145,146]. It was observed moreover, that fish MAVS contains similar domains as the mammalian one [43] and is associated with both RIG-I and MDA5 signaling pathways, and can induce type I IFN response through activation of TRAF3 and TBK1 [43]. For example, grass carp MAVS induced activation of type I IFN through IRF7 but not IRF3 [141] while zebrafish MAVS splicing variant (MAVS_tv2) suppressed activation of type I IFN by targeting IRF7 [142]. Intracellular localization of MAVS in mitochondria was demonstrated in various fish species [110,137,145,147,148]. Interestingly Xiang et al. 2011 [139] reveal that MAVS of Tetraodon nigroviridis is located near by the plasma membrane but not merged with mitochondria in fathead minnow (FHM) epithelial cell.
Up-regulation of the expression of mavs after viral infection or poly(I:C) stimulation in cell lines (in vitro studies) or in various organs of different fish species (in vivo studies) have been widely demonstrated in many reports [64,91,140,144,147,149,150,151,152,153,154]. The functional studies demonstrated that inhibition of MAVS impairs the antiviral response in fish. For example, cytosolic poly (I:C)-induced or RIG-I-induced type I IFN response was attenuated by functional blockade of crucian carp MAVS [155]. Deletion of CARD or its transmembrane domains (TM) in Atlantic salmon abolished activation of the IFNa1 promoter and the NF-κβ driven promoter [137]. Similarly, deletion of the TM in MAVS of Tetraodon nigroviridis resulted in inhibition of ISRE and NF-κβ activation while deletion of the CARD domain negatively affected MAVS function on the ISRE but not affected activation of NF-κβ [139]. The protective effect of mavs overexpression against viral infections was manifested by reduced CPE and decreased viral titer or viral gene expression in studied cell lines of different fish species [110,138,140,145,147,149,152]. Moreover overexpression of mavs induced up-regulation of the expression of several downstream signaling molecules (e.g., irf3 and irf7), type I IFNs and antiviral proteins [117,138,139,140,145,147,153,155] Interestingly, co-transfection of nile tilapia MAVS with MDA5 resulted in the slightly increased activation of MAVS-dependent NF-κB, whereas co-transfection of nile tilapia MAVS with LGP2 resulted in significantly decreased MAVS-dependent NF-κB activation [117].

7. cGAS

Cyclic guanosine monophosphare-adenosine monophosphate (GMP-AMP) synthase (cGAS) is one of the cytosolic PRRs that binds to double-stranded DNA (dsDNA) in the cytoplasm and catalyzes the synthesis of second messenger molecule 2′3′-cGAMP. cGAS belongs to the family of nucleotidyltransferase (NTase) enzymes [156].
In mammals, cGAS is composed of N-terminal and C-terminal male abnormal 21 (mab21) domains. The mab21 domain has a region that binds Zn2+. The coordinated Zn2+ (Zinc-Ribbon) binds across the dsDNA to form a cGAS-DNA complex, which excites the cGAS protein [156,157]. The Zn2+ binding region attaches to other NTase family proteins. Deletion of zinc-ribbon domain shows that it is importance in dsDNA sensing by cGAS [158]. Furthermore, the N-terminal domain of mammalian cGAS plays an important role in mounting the STING/IRF3-mediated cytosolic DNA signaling [159]. cGAS is conserved in various species spanning from fish to mammals. In teleosts, the N-terminal region of the cGAS protein is markedly conserved, while the amino acids involved in the DNA-binding surface and cGAMP synthesis in the mab21 domain are also highly conserved [105,160].
Upon binding to foreign DNA of viruses, bacteria, and parasites that invade into cells and upon detection in the cytoplasm of mislocalized endogenous DNA, cGAS catalyzes the synthesis of a second messenger molecule, cGAMP (from ATP and GTP) (Figure 1B). cGAMP binds to the protein stimulator of interferon gene (STING), which is localized in the endoplasmic reticulum (ER) membrane, and shifts translocation of STING from the ER to the Golgi apparatus. Next, STING promotes the activation of TANK binding kinase 1 (TBK1). Activated TBK1 causes phosphorylation and dimerization of the IRF3 or IRF7. These transcription factors translocate into the nucleus and induce production of type I IFNs. At the same time, STING promotes nuclear translocation of NF-κB, what induces the production of pro-inflammatory cytokines [161,162,163]. Furthermore, upon nonspecific binding of cytosolic B-form DNA, cGAS synthesizes the 2′3′-cGAMP and triggers STING-dependent signaling [159]. However, human cGAS favors DNA-length discrimination and sufficiently detects a longer DNA [164]. By transfecting THP-1 cells with dsDNA of 88 to 4003 bp at a DNA, a ligand length-dependent type I IFN response was confirmed [165].
The function of cGAS against pathogens has been extensively studied in mammals, but little is known about their functions in fish. In mammals, injection of genomic DNA of pathogens including viruses, bacteria, and parasites into monocyte-derived cells excites immune signals downstream of cGAS. The function of cGAS against pathogens including virus, bacteria and parasite in mammals and fish are summarized in Table 2.
Although, induction of type I IFN by cGAS sensing mechanism is more powerful than its induction by other DNA sensors, pathogens may inhibit cGAS activity as a part of pathogen evasion mechanisms. For example, UL41 protein of Herpes simplex virus 1 (HSV-1) may partially degrade cGAS mRNA, while VP22 directly acts on the cGAS protein to degrade it [166,167,168]. The naked relaxed-circular DNA of chronic hepatitis B virus (HBV) is sensed in a cGAS-dependent manner in human hepatocytes, however host cell recognition of viral nucleic acids is abolished during HBV infection, which suppresses cGAS expression and function in the hepatocytes [169]. In addition, cGAS senses cDNAs of retroviruses (i.e., RNA viruses), such as human immunodeficiency virus (HIV), Murine leukemia virus (MLV) and Simian immunodeficiency virus (SIV), which are synthesized by reverse transcription in the cytoplasm. Moreover, Dengue virus (DENV) cleaves STING on the ER membrane with a protease, which also blocks self-DNA detection. Then, some of the viral proteins reach the mitochondrial membrane, causing mitochondrial stress and subsequent mtDNA leakage. Upon recognition of mtDNA leaked into the cytoplasm, cGAS synthesizes the second messenger molecule cGAMP, and induces IFN production by STING activation in DENV-infected cells and signals to neighboring cells via gap junctions [170]. This functional mechanism of mammalian cGAS was also confirmed by infection with intracellular bacteria [171,172,173,174] and parasites [175,176,177] (Table 2). Sensing Mycobacterium tuberculosis (Mtb) DNA via the cGAS-STING pathway induces type I IFN and autophagy [172,173], however, these inductions do not contribute to host protection against Mtb infection in mammalian lung cells [171]. Human cGAS also senses the genomic DNAs of malaria inducer, Plasmodium falciparum and Leishmania donovani and induces type I IFN production [176,177].
On the contrary, the function of cGAS against pathogens in fish is poorly understood. Infection of HSV-1 in cGAS knock-down zebrafish had no effect on induction of ifnφ1, isg15 and viperin. Rather, double-knockdown of two zebrafish DNA sensors, DDX41 and DHX9, almost abolished the induction of above-mentioned antiviral genes. Therefore, cGAS was hardly involved in the biological defense of zebrafish, and it was considered that cGAS evolved in the animal kingdom after fish [178]. Currently, specific activities of cGAS variants were determined using in vitro assay and RP-HPLC measurements of cGAMP production. Although, the zebrafish cGAS homologue has low amino acid identity to human cGAS (less than 35%), their similar activities were determined [179]. Liu and co-workers [160] reported that overexpression of zebrafish cGASa/b in HEK293T cells and zebrafish embryos significantly activated NF-κB and type I IFN signaling pathways in a STING-dependent manner, and that cGASa, but not cGASb, was involved in immunoglobulin Z-mediated mucosal immunity in gill-associated lymphoid tissue, suggesting differential functions between the two DrcGASs [160]. Interestingly, it was revealed that the ortholog of cGAS in grass carp, cGAS-like (cGASL) by interacting with STING down-regulates transcription of type I IFN gene and functions as a negative regulator of IFN response. It suggests interaction between cGAS and MITA-TBK1 complex, which may partly hinder the phosphorylation process mediated by TBK1 [180]. Moreover, it has also been reported that infection of Japanese medaka with the intracellular bacteria, E. tarda promotes increased expression of cgas gene in the intestinal tract [105]. In summary, fish cGAS, like mammals, induces STING-mediated production of type I IFN through NF-κB activation. On the other hand, it may be involved in humoral immune response mediated by immunoglobulin Z, which has not been confirmed in mammals. It is still controversial whether fish cGAS is effective to avoid infection with pathogens.

8. LSm14A

Sm-like protein homolog A (LSm14A; also known as RAP55) belongs to the RNA-binding proteins. It is a highly conserved protein and a component of processing body (P-body) involved in mRNA metabolism. LSm14 was first discovered in the oocytes of Pleurodeles waltl and Xenopus laevis as RAP55 (mRNA-associated protein of 55kDa) [188,189]. LSm14A is composed of a N-terminal LSm domain that is necessary for RNA binding, localization to processing (P)-bodies and translational control while in a C-terminal domain there are a DFDF box, an FFD-TFG box and RCG repeats responsible for targeting the proteins to P-bodies in mammals [189,190]. Several studies of the function of LSm14A in higher vertebrates indicated that it is a sensor for viral nucleic acids and performs a pivotal role in activation of IFN-β signaling pathway [191,192,193,194,195,196]. However, the function of LSm14A in teleosts is still very poorly understood. Recently, LSm14A has been identified in Japanese medaka [105]. The Japanese medaka Cab LSm14A cDNA is a 1311 bp long ORF encoding a predicted protein of 436 amino acids. Both the LSm and FDF domains in LSm14A are highly conserved, especially the sequence identities of medaka LSm domain were 96–99% to that of 14 other vertebrates [105]. Moreover, it was found that lsm14a gene was induced in the intestine, kidney, and spleen of formalin-killed E. tarda-treated medaka, what suggests that the transcription of this CDS gene was induced by the stimulation with dsDNA of E. tarda. As there is a single report so far, the identification and function of this molecule of other fish species require to be studied for further understanding.

9. PKR and PKZ

The double stranded RNA (dsRNA)-dependent protein kinase R (PKR) (alternatively called eukaryotic translation initiation factor 2-alpha kinase 2; E2AK2 or eIF2AK2) is one of the most studied interferon-induced proteins presented in all vertebrates [197,198,199,200]. PKR contains two N-terminal dsRNA binding domains and a catalytic kinase domain in the C terminal region. Its activation is triggered by binding to viral derived dsRNA, which resulted in homodimer formation and autophosphorylation. Active PKR contributes to the anti-viral innate immune response, mainly (but not exclusively) by phosphorylation of the eukaryotic initiation factor 2 alpha (eIF-2α) and consequent shut-down of protein synthesis [201]. Interestingly, a paralogue of PKR, called PKR-like protein kinase containing Z-DNA binding domain (PKZ) [202,203], which has a closer evolutionary relationship with PKR [204], has been identified in teleost fish species [203]. The C-terminal regions of PKR and PKZ are closely related (both encode an active protein kinase domain). In contrast, the N-terminal region of PKZ contains two Zalpha domains instead of the dsRNA binding domains found in PKR. Zalpha domains of PKZ bind the left-handed dsDNA (Z DNA) [205]. Binding of PKZ to Z-DNA induces its activation through homodimerization and autophosphorylation. Once activated PKZ, like PKR, is able to phosphorylate eIF-2α thereby blocking protein synthesis [203,206,207]. In addition to PKZ, Zalpha domains have only been identified in two additional cellular proteins which play a role in regulation of interferon responses: the RNA-editing enzyme ADAR1 (described in vertebrates) [208] and the Z-DNA binding protein 1 (ZBP1), also known as DNA-dependent activator of IFN-regulatory factors (DAI) or DLM-1 [209]. On account of the fact that DAI (ZBP1/DLM-1) has not been found in the fish genome, it can be hypothetized that PKZ is a kind of compensator for the lack of DAI and function as a cytosolic DNA sensor triggering innate antiviral immune response [210]. The antiviral functions of PKR and PKZ are completely different compared to the effect of the activation of RLRs that induce expression of the gene encoded the host antiviral factors.
In the fish of order cypriniformes, such as goldfish (Carassius auratus), grass carp (Ctenopharyngodon idella) and zebrafish (Danio rerio), pkr and pkz genes are tandemly arranged in head-to-tail (parallel) orientation on the same chromosome, with respect to 5′ to 3′ alignment of the genes [199,207,210]. In contrast, in the fish of order tetraodontiformes, such as spotted green pufferfish (Tetraodon nigroviridis) and Japanese pufferfish (Takifugu rubripes), two or three pkr genes (pkr 1, pkr 2, and pkr 3) are tandemly arranged on the same chromosome [199,203], suggesting that one of the pkr genes could be rearranged to pkz or an ancestral gene could be differently duplicated by some evolutional events between cypriniformes to tetraodontiformes. Fish pkr genes have been already cloned from many fish species; goldfish [207], grass carp [211], Japanese flounder (Paralichthys olivaceus) [200], Japanese pufferfish [212], rock bream (Oplegnathus fasciatus) [213], spotted green pufferfish [199], and zebrafish [214], while pkz genes have been identified in Atlantic salmon (Salmo salar) [206], Chinese rare minnow (Gobiocypris rarus) [215], goldfish [202,207], grass carp [216], and zebrafish [203] (Table 3). Fish pkr and pkz genes are constitutively expressed at a very low level in cultured cells and tissues and their expression is up-regulated in response to stimulation with interferon, poly(I:C), poly(dA-dT), poly(dG-dC), genomic DNA, and during infections with several viruses but also bacterial Aeromonas hydrophila [199,202,203,206,207,215,216]. It was moreover found that overexpression of pkr or pkz in transfected cells significantly inhibited protein synthesis [203]. Although the importance of PKR in antiviral innate immune response has been demonstrated by an extensive studies using various virus-host models, the anti-viral role of PKZ has been investigated to the small extend. For example [207] demonstrated that PKR and PKZ can cooperate roles in antiviral innate immune response against grass carp reovirus. The antiviral functions of PKR and PKZ in fish are summarized in Table 3.

10. Conclusions

The recognition of various kinds of intracellular nucleic acids of pathogens and initiation of innate immune responses to them by host cells has yet to be fully understood. Over the past decade or so, several investigations have focused on clarifying the mechanisms underlying these innate immune responses triggered by sensors that recognize pathogen-derived components in cytosol. The findings of these studies have suggested that in higher vertebrates immune responses to viral or bacterial nucleic acids are mediated by cytosolic sensors and their adaptor molecules while our understanding of this systems/molecules in other animals is less advanced. Teleost fish form a perfect model to identify and extend our knowledge of evolutionary conservation of important ligands and receptors involved in this process. They are the first species to utilize both an innate and adaptive response with specific antibodies to combat infections. Moreover, they are found in essentially every aquatic habitat and have been successful in adapting to different environments. More than half of all extant vertebrate species belong to this group (there are an estimated 35,000 species of bony fish) and their ability to recognize and eradicate pathogens must have contributed to this success.
It is already known that fish cells, similarly to their mammalian counterparts, respond to a variety of pathogens through their PRRs, which recognize different types of nucleic acids from pathogens. As in mammals also in fish, viral RNAs are recognized by DExD/H-box RNA helicases that include a group of well-characterized RLRs (RIG-I/DDX58, MDA5 and LGP2) and non-RLRs (DDX1, DDX3, DHX9, DDX21, DHX36, and DDX41). Schematic representation of the domain topology and conserved motifs of fish and mammalian RLRs and non-RLRs DExD/H-box RNA helicases revealed that the structure of these receptors is very conserved. The same domains and motifs are present in these receptors in fish and mammals [22,46,100]. Although, all the three RLRs (RIG-I, MDA5 and LGP2) are mostly conserved in fish, RIG-I does not exist in some fish species of Acanthopterygii superorder therefore it can be hypothesized that fish rig-I gene could has been lost after the divergence occurring before the Acanthopterygian fish [22]. In turn, among non-RLRs DExD/H-box RNA helicases, DDX1, DDX3, DHX9, DDX21, DHX36 and DDX41 have been described in several fish species and their role in antiviral and antibacterial immune response was confirmed. Also another RNA-binding cytosolic PRR, LSm14A/RAP55, described to act as a sensor for viral nucleic acids in higher vertebrates, it has recently been identified in fish with a potential ability to sensing dsDNA of an intracellular bacterium. In contrast function of teleost cGAS in the recognition of viral, bacteria and parasite-derived DNAs has not yet been elucidated clearly. Intriguingly, next to exerting antiviral immune responses upon sensing dsRNA, PKR, fish possess a novel PKR-like protein kinase containing Z-DNA binding domain, named as PKZ which is involved in sensing of Z-DNA.
Although, these cytosolic PRRs recognizing pathogenic viral and bacterial nucleic acids have recently been identified from various fish species, their detailed origin and evolution, involvement of adaptor molecules, exact ligand binding specificities, downstream signaling pathways with regulation mechanism, and induction of type I IFN and inflammatory cytokine production are largely unknown and may vary compared to those of mammals. Therefore, further studies are required to reveal these aspects clearly. Regardless of existence of differences in recognition mechanisms for cytosolic viral and bacterial nucleic acids between fish and other vertebrates, there is much similarity, suggesting an evolutionarily conserved immune system that is indispensable for pathogen sensing and removal.
More detailed recognition of this molecules and pathways will extend our knowledge but will have also practical aspect to design and improve new strategies for fish health control. This is of great importance in view of the annual growth of the fish farming industry, where infectious diseases have a significant impact on productivity and profitability.

Author Contributions

Writing—original draft preparation, review and editing: M.M., K.R., M.C., K.N., G.B., M.S., J.-i.H. Figure and Tables preparation: M.M., K.R., K.N., J.-i.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Science Centre of Poland under Sonata Bis 5 project (Grant number UMO-2015/18/E/NZ6/00516). This research was also funded by a Grant-in Aid for Scientific Research (B) (KAKENHI Grant number 17H03863) from the Japan Society for the Promotion of Science, Japan.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Jensen, S.; Thomsen, A.R. Sensing of RNA viruses: A review of innate immune receptors involved in recognizing RNA virus invasion. J. Virol. 2012, 86, 2900–2910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kawasaki, T.; Kawai, T.; Akira, S. Recognition of nucleic acids by pattern-recognition receptors and its relevance in autoimmunity. Immunol. Rev. 2011, 243, 61–73. [Google Scholar] [CrossRef] [PubMed]
  4. Paludan, S.R.; Bowie, A.G. Immune sensing of DNA. Immunity 2013, 38, 870–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Walsh, D.; McCarthy, J.; O’Driscoll, C.; Melgar, S. Pattern recognition receptors-molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013, 24, 91–104. [Google Scholar] [CrossRef]
  6. Wilkins, C.; Gale, M. Recognition of viruses by cytoplasmic sensors. Curr. Opin. Immunol. 2011, 22, 41–47. [Google Scholar] [CrossRef] [Green Version]
  7. Yoneyama, M.; Fujita, T. Recognition of viral nucleic acids in innate immunity. Rev. Med. Virol. 2010, 20, 4–22. [Google Scholar] [CrossRef]
  8. Medzhitov, R.; Janeway, C.A. Innate immunity: Impact on the adaptive immune response. Curr. Opin. Immunol. 1997, 9, 4–9. [Google Scholar] [CrossRef]
  9. Beg, A.A. Endogenous ligands of Toll-like receptors: Implications for regulating inflammatory and immune responses. Trends Immunol. 2002, 23, 509–512. [Google Scholar] [CrossRef]
  10. Matzinger, P. The danger model: A renewed sense of self. Science 2002, 296, 301–305. [Google Scholar] [CrossRef] [Green Version]
  11. Mogensen, T.H.; Paludan, S.R. Reading the viral signature by Toll-like receptors and other pattern recognition receptors. J. Mol. Med. 2005, 83, 180–192. [Google Scholar] [CrossRef] [PubMed]
  12. Bieback, K.; Lien, E.; Klagge, I.M.; Avota, E.; Schneider-Schaulies, J.; Duprex, W.P.; Wagner, H.; Kirschning, C.J.; ter Meulen, V.; Schneider-Schaulies, S. Hemagglutinin protein of wild-type measles virus activates Toll-like receptor 2 signaling. J. Virol. 2002, 76, 8729–8736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Burzyn, D.; Rassa, J.C.; Kim, D.; Nepomnaschy, I.; Ross, S.R.; Piazzon, I. Toll-Like receptor 4-dependent activation of dendritic cells by a retrovirus. J. Virol. 2004, 78, 576–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rassa, J.C.; Meyers, J.L.; Zhang, Y.; Kudaravalli, R.; Ross, S.R. Murine retroviruses activate B cells via interaction with toll-like receptor 4. Proc. Natl. Acad. Sci. USA 2002, 99, 2281–2286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Sabbah, A.; Chang, T.H.; Harnack, R.; Frohlich, V.; Tominaga, K.; Dube, P.H.; Xiang, Y.; Bose, S. Activation of innate immune antiviral responses by Nod2. Nat. Immunol. 2009, 10, 1073–1080. [Google Scholar] [CrossRef]
  16. Albiger, B.; Dahlberg, S.; Henriques-Normark, B.; Normark, S. Role of the innate immune system in host defence against bacterial infections: Focus on the Toll-like receptors. J. Intern. Med. 2007, 261, 511–528. [Google Scholar] [CrossRef]
  17. Schwandner, R.; Dziarski, R.; Wesche, H.; Rothe, M.; Kirschning, C.J. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by Toll-like receptor 2. J. Biol. Chem. 1999, 274, 17406–17409. [Google Scholar] [CrossRef] [Green Version]
  18. Hayashi, F.; Smith, K.D.; Ozinsky, A.; Hawn, T.R.; Yi, E.C.; Goodlett, D.R.; Eng, J.K.; Akira, S.; Underhill, D.M.; Aderem, A. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 2001, 410, 1099–1103. [Google Scholar] [CrossRef]
  19. Miao, E.A.; Ernst, R.K.; Dors, M.; Mao, D.P.; Aderem, A. Pseudomonas aeruginosa activates caspase 1 through Ipaf. Proc. Natl. Acad. Sci. USA 2008, 105, 2562–2567. [Google Scholar] [CrossRef] [Green Version]
  20. Kanwal, Z.; Wiegertjes, G.F.; Veneman, W.J.; Meijer, A.H.; Spaink, H.P. Comparative studies of Toll-like receptor signalling using zebrafish. Dev. Comp. Immunol. 2014, 46, 35–52. [Google Scholar] [CrossRef]
  21. Quiniou, S.M.A.; Boudinot, P.; Bengtén, E. Comprehensive survey and genomic characterization of Toll-like receptors (TLRs) in channel catfish, Ictalurus punctatus: Identification of novel fish TLRs. Immunogenetics 2013, 65, 511–530. [Google Scholar] [CrossRef] [PubMed]
  22. Aoki, T.; Hikima, J.; Hwang, S.D.; Jung, T.S. Innate immunity of finfish: Primordial conservation and function of viral RNA sensors in teleosts. Fish Shellfish Immunol. 2013, 35, 1689–1702. [Google Scholar] [CrossRef] [PubMed]
  23. Palti, Y. Toll-like receptors in bony fish: From genomics to function. Dev. Comp. Immunol. 2011, 35, 1263–1272. [Google Scholar] [CrossRef] [PubMed]
  24. Rebl, A.; Goldammer, T.; Seyfert, H.M. Toll-like receptor signaling in bony fish. Vet. Immunol. Immunopathol. 2010, 134, 139–150. [Google Scholar] [CrossRef] [PubMed]
  25. Ranji, A.; Boris-Lawrie, K. RNA helicases: Emerging roles in viral replication and the host innate response. RNA Biol. 2010, 7, 775–787. [Google Scholar] [CrossRef] [Green Version]
  26. Cordin, O.; Banroques, J.; Tanner, N.K.; Linder, P. The DEAD-box protein family of RNA helicases. Gene 2006, 367, 17–37. [Google Scholar] [CrossRef]
  27. Fullam, A.; Schröder, M. DExD/H-box RNA helicases as mediators of anti-viral innate immunity and essential host factors for viral replication. Biochim. Biophys. Acta Gene Regul. Mech. 2013, 1829, 854–865. [Google Scholar] [CrossRef] [Green Version]
  28. Lee, T.; Pelletier, J. The biology of DHX9 and its potential as a therapeutic target. Oncotarget 2016, 7, 42716–42739. [Google Scholar] [CrossRef] [Green Version]
  29. Story, R.M.; Li, H.; Abelson, J.N. Crystal structure of a DEAD box protein from the hyperthermophile Methanococcus jannaschii. Proc. Natl. Acad. Sci. USA 2001, 98, 1465–1470. [Google Scholar] [CrossRef] [Green Version]
  30. Ariumi, Y. Multiple functions of DDX3 RNA helicase in gene regulation, tumorigenesis, and viral infection. Front. Genet. 2014, 5, 1–10. [Google Scholar] [CrossRef] [Green Version]
  31. Fuller-Pace, F.V. DExD/H box RNA helicases: Multifunctional proteins with important roles in transcriptional regulation. Nucleic Acids Res. 2006, 34, 4206–4215. [Google Scholar] [CrossRef] [PubMed]
  32. Linder, P. Dead-box proteins: A family affair-active and passive players in RNP-remodeling. Nucleic Acids Res. 2006, 34, 4168–4180. [Google Scholar] [CrossRef] [PubMed]
  33. Schröder, M. Human DEAD-box protein 3 has multiple functions in gene regulation and cell cycle control and is a prime target for viral manipulation. Biochem. Pharmacol. 2010, 79, 297–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Haas, T.; Poeck, H. Apoptosis induction by cytosolic RNA helicases. J. Med. Microb. Diagn. 2012, 1, 4–9. [Google Scholar] [CrossRef]
  35. Kolokoltsova, O.A.; Grant, A.M.; Huang, C.; Smith, J.K.; Poussard, A.L.; Tian, B.; Brasier, A.R.; Peters, C.J.; Tseng, C.K.; Torre, J.C.; et al. RIG-I enhanced interferon independent apoptosis upon junin virus infection. PLoS ONE 2014, 9, e99610. [Google Scholar] [CrossRef] [PubMed]
  36. Abdelhaleem, M. Over-expression of RNA helicases in cancer. Anticancer Res. 2004, 3954, 3951–3953. [Google Scholar]
  37. Fuller-Pace, F.V. DEAD box RNA helicase functions in cancer DEAD. RNA Biol. 2013, 10, 121–132. [Google Scholar] [CrossRef] [Green Version]
  38. Takeuchi, O.; Akira, S. MDA5/RIG-I and virus recognition. Curr. Opin. Immunol. 2008, 20, 17–22. [Google Scholar] [CrossRef]
  39. Yoneyama, M.; Onomoto, K.; Jogi, M.; Akaboshi, T.; Fujita, T. Viral RNA detection by RIG-I-like receptors. Curr. Opin. Immunol. 2015, 32, 48–53. [Google Scholar] [CrossRef]
  40. Brisse, M.; Ly, H. Comparative structure and function analysis of the RIG-I-like receptors: RIG-I and MDA5. Front. Immunol. 2019, 10, 1–27. [Google Scholar] [CrossRef]
  41. Loo, Y.; Gale, M. Immune signaling by RIG-I-like receptors. Immunity 2011, 34, 680–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Choi, S.J.; Lee, H.; Kim, J.; Park, S.Y.; Kim, T.; Lee, W.; Jang, D.; Yoon, J.; Choi, Y.; Kim, S.; et al. HDAC 6 regulates cellular viral RNA sensing by deacetylation of RIG. EMBO J. 2016, 35, 429–442. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, S.N.; Zou, P.F.; Nie, P. Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) in fish: Current knowledge and future perspectives. Immunology 2017, 151, 16–25. [Google Scholar] [CrossRef] [PubMed]
  44. Takahasi, K.; Yoneyama, M.; Nishihori, T.; Hirai, R.; Kumeta, H.; Narita, R.; Gale, M.; Inagaki, F.; Fujita, T. Nonself RNA-sensing mechanism of RIG-I helicase and activation of antiviral immune responses. Mol. Cell 2008, 29, 428–440. [Google Scholar] [CrossRef] [PubMed]
  45. Deddouche, S.; Goubau, D.; Rehwinkel, J.; Chakravarty, P.; Begum, S.; Maillard, P.V.; Borg, A.; Matthews, N.; Feng, Q.; van Kuppeveld, F.J.M.; et al. Identification of an LGP2-associated MDA5 agonist in picornavirus-infected cells. eLife 2014, 3, 1–20. [Google Scholar] [CrossRef]
  46. Lazarte, J.M.S.; Thompson, K.D.; Jung, T.S. Pattern recognition by melanoma differentiation-associated gene 5 (Mda5) in teleost fish: A review. Front. Immunol. 2019, 10, 1–11. [Google Scholar] [CrossRef]
  47. Bruns, A.M.; Horvath, C.M. LGP2 synergy with MDA5 in RLR-mediated RNA recognition and antiviral signaling. Cytokine 2015, 74, 198–206. [Google Scholar] [CrossRef] [Green Version]
  48. Jiang, X.; Kinch, L.N.; Brautigam, C.A.; Chen, X.; Du, F.; Grishin, N.V.; Chen, Z.J. Ubiquitin-induced oligomerization of the RNA sensors RIG-I and MDA5 activates antiviral innate immune response. Immunity 2012, 36, 959–973. [Google Scholar] [CrossRef] [Green Version]
  49. Schoggins, J.W.; Rice, C.M. Interferon-stimulated genes and their antiviral effector functions. Curr. Opin. Virol. 2011, 1, 519–525. [Google Scholar] [CrossRef]
  50. Yoneyama, M.; Kikuchi, M.; Matsumoto, K.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Foy, M.; Loo, Y.; Gale, M.; Akira, S.; et al. Shared and unique functions of the DExD/H-Box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. J. Immunol. 2005, 175, 2851–2858. [Google Scholar] [CrossRef] [Green Version]
  51. Komuro, A.; Horvath, C.M. RNA- and virus-independent inhibition of antiviral signaling by RNA helicase LGP2. J. Virol. 2006, 80, 12332–12342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Parisien, J.; Lenoir, J.J.; Mandhana, R.; Rodriguez, K.R.; Qian, K.; Bruns, A.M.; Horvath, C.M. RNA sensor LGP 2 inhibits TRAF ubiquitin ligase to negatively regulate innate immune signaling. EMBO Rep. 2018, 19, 1–14. [Google Scholar] [CrossRef] [PubMed]
  53. Venkataraman, T.; Valdes, M.; Elsby, R.; Kakuta, S.; Caceres, G.; Saijo, S.; Iwakura, Y.; Barber, G.N. Loss of DExD/H Box RNA helicase LGP2 manifests disparate antiviral responses. J. Immunol. 2007, 178, 6444–6455. [Google Scholar] [CrossRef] [PubMed]
  54. Satoh, T.; Kato, H.; Kumagai, Y.; Yoneyama, M.; Sato, S.; Matsushita, K.; Tsujimura, T.; Fujita, T.; Akira, S.; Takeuchi, O. LGP2 is a positive regulator of RIG-I- and MDA5-mediated antiviral responses. Proc. Natl. Acad. Sci. USA 2010, 107, 1512–1517. [Google Scholar] [CrossRef] [Green Version]
  55. Liniger, M.; Summerfield, A.; Zimmer, G.; McCullough, K.C.; Ruggli, N. Chicken cells sense influenza A virus infection through MDA5 and CARDIF signaling involving LGP2. J. Virol. 2012, 86, 705–717. [Google Scholar] [CrossRef] [Green Version]
  56. Xu, L.; Yu, D.; Fan, Y.; Peng, L.; Wu, Y.; Yao, Y.G. Loss of RIG-I leads to a functional replacement with MDA5 in the Chinese tree shrew. Proc. Natl. Acad. Sci. USA 2016, 113, 10950–10955. [Google Scholar] [CrossRef] [Green Version]
  57. Ohtani, M.; Hikima, J.; Kondo, H.; Hirono, I.; Jung, T.; Aoki, T. Evolutional conservation of molecular structure and antiviral function of a viral RNA receptor, LGP2, in Japanese flounder, Paralichthys olivaceus. J. Immunol. 2010, 185, 7507–7517. [Google Scholar] [CrossRef] [Green Version]
  58. Huang, T.; Su, J.; Heng, J.; Dong, J.; Zhang, R.; Zhu, H. Identifcation and expression profiling analysis of grass carp Ctenopharyngodon idella LGP2 cDNA. Fish Shellfish Immunol. 2010, 29, 349–355. [Google Scholar] [CrossRef]
  59. Xu, C.; Evensen, Ø.; Munang’andu, H.M. De Novo transcriptome analysis shows that SAV-3 infection upregulates pattern recognition receptors of the endosomal Toll-Like and RIG-I-like receptor signaling pathways in macrophage/dendritic like TO cells. Viruses 2016, 8, 114. [Google Scholar] [CrossRef]
  60. Nerbøvik, I.G.; Solheim, M.A.; Eggestøl, H.Ø.; Rønneseth, A.; Jakobsen, R.A.; Wergeland, H.I.; Haugland, G.T. Molecular cloning of MDA5, phylogenetic analysis of RIG-I-like receptors (RLRs) and differential gene expression of RLRs, interferons and proinflammatory cytokines after in vitro challenge with IPNV, ISAV and SAV in the salmonid cell line TO. J. Fish Dis. 2017, 40, 1529–1544. [Google Scholar] [CrossRef]
  61. Wessel, Ø.; Olsen, C.M.; Rimstad, E.; Dahle, M.K. Piscine orthoreovirus (PRV) replicates in Atlantic salmon (Salmo salar L.) erythrocytes ex vivo. Vet. Res. 2015, 26, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Skjesol, A.; Skjæveland, I.; Elnæs, M.; Timmerhaus, G.; Fredriksen, B.N.; Jørgensen, S.M.; Krasnov, A.; Jørgensen, J.B. IPNV with high and low virulence: Host immune responses and viral mutations during infection. Virol. J. 2011, 8, 396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Rajendran, K.V.; Zhang, J.; Liu, S.; Peatman, E.; Kucuktas, H.; Wang, X.; Liu, H.; Wood, T.; Terhune, J.; Liu, Z. Pathogen recognition receptors in channel catfish: II. Identification, phylogeny and expression of retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs). Dev. Comp. Immunol. 2012, 37, 381–389. [Google Scholar] [CrossRef] [PubMed]
  64. Feng, H.; Liu, H.; Kong, R.; Wang, L.; Wang, Y.; Hu, W.; Guo, Q. Expression profiles of carp IRF-3/-7 correlate with the up-regulation of RIG-I/MAVS/TRAF3/TBK1, four pivotal molecules in RIG-I signaling pathway. Fish Shellfish Immunol. 2011, 30, 1159–1169. [Google Scholar] [CrossRef] [PubMed]
  65. Mou, C.; Wang, Y.; Zhang, Q.; Gao, F.; Li, Z.; Tong, J.; Zhou, L.; Gui, J. Differential interferon system gene expression profiles in susceptible and resistant gynogenetic clones of gibel carp challenged with herpesvirus CaHV. Dev. Comp. Immunol. 2018, 86, 52–64. [Google Scholar] [CrossRef]
  66. Yang, C.; Su, J.; Huang, T.; Zhang, R.; Peng, L. Identification of a retinoic acid-inducible gene I from grass carp (Ctenopharyngodon idella) and expression analysis in vivo and in vitro. Fish Shellfish Immunol. 2011, 30, 936–943. [Google Scholar] [CrossRef]
  67. Chen, L.; Li, Q.; Su, J.; Yang, C.; Li, Y.; Rao, Y. Trunk kidney of grass carp (Ctenopharyngodon idella) mediates immune responses against GCRV and viral/bacterial PAMPs in vivo and in vitro. Fish Shellfish Immunol. 2013, 34, 909–919. [Google Scholar] [CrossRef]
  68. Liao, Z.; Wan, Q.; Su, H.; Wu, C.; Su, J. Pattern recognition receptors in grass carp Ctenopharyngodon idella: I. Organization and expression analysis of TLRs and RLRs. Dev. Comp. Immunol. 2017, 76, 93–104. [Google Scholar] [CrossRef]
  69. Feng, J.; Guo, S.; Lin, P.; Wang, Y.; Zhang, Z.; Zhang, Z.; Yu, L. Identification of a retinoic acid-inducible gene I from Japanese eel (Anguilla japonica) and expression analysis in vivo and in vitro. Fish Shellfish Immunol. 2016, 55, 249–256. [Google Scholar] [CrossRef]
  70. Huang, B.; Wang, Z.X.; Zhang, C.; Zhai, S.W.; Han, Y.S.; Huang, W.S.; Nie, P. Identification of a novel RIG-I isoform and its truncating variant in Japanese eel, Anguilla japonica. Fish Shellfish Immunol. 2019, 94, 373–380. [Google Scholar] [CrossRef]
  71. Chen, H.; Liu, W.; Wu, S.; Chiou, P.P.; Li, Y.; Chen, Y.; Lin, G.; Lu, M.; Wu, J. RIG-I specifically mediates group II type I IFN activation in nervous necrosis virus infected zebrafish cells. Fish Shellfish Immunol. 2015, 43, 427–435. [Google Scholar] [CrossRef]
  72. Wang, W.; Asim, M.; Yi, L.; Hegazy, A.M.; Hu, X.; Zhou, Y.; Ai, T.; Lin, L. Abortive infection of snakehead fish vesiculovirus in ZF4 cells was associated with the RLRs pathway activation by viral replicative intermediates. Int. J. Mol. Sci. 2015, 16, 6235–6250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zou, P.F.; Chang, M.X.; Li, Y.; Zhang, S.H.; Fu, J.P.; Chen, S.N.; Nie, P. Higher antiviral response of RIG-I through enhancing RIG-I/MAVS-mediated signaling by its long insertion variant in zebrafish. Fish Shellfish Immunol. 2015, 43, 13–24. [Google Scholar] [CrossRef] [PubMed]
  74. López-Muñoz, A.; Roca, F.J.; Sepulcre, M.P.; Meseguer, J.; Mulero, V. Zebrafish larvae are unable to mount a protective antiviral response against waterborne infection by spring viremia of carp virus. Dev. Comp. Immunol. 2010, 34, 546–552. [Google Scholar] [CrossRef] [PubMed]
  75. Nie, L.; Zhang, Y.; Dong, W.; Xiang, L.; Shao, J. Involvement of zebrafish RIG-I in NF-κB and IFN signaling pathways: Insights into functional conservation of RIG-I in antiviral innate immunity. Dev. Comp. Immunol. 2015, 48, 95–101. [Google Scholar] [CrossRef] [PubMed]
  76. Fan, S.; Chen, S.; Liu, Y.; Lin, Y.; Liu, H.; Guo, L.; Lin, B.; Huang, S.; Xu, A. Zebrafish TRIF, a golgi-localized protein, participates in IFN induction and NF- B activation. J. Immunol. 2008, 180, 5373–5383. [Google Scholar] [CrossRef] [PubMed]
  77. Rakus, K.; Mojzesz, M.; Widziolek, M.; Pooranachandran, N.; Teitge, F.; Surachetpong, W.; Chadzinska, M.; Steinhagen, D.; Adamek, M. Antiviral response of adult zebrafish (Danio rerio) during tilapia lake virus (TiLV) infection. Fish Shellfish Immunol. 2020, 101, 1–8. [Google Scholar] [CrossRef] [PubMed]
  78. Chang, M.; Collet, B.; Nie, P.; Lester, K.; Campbell, S.; Secombes, C.J.; Zou, J. Expression and functional characterization of the RIG-I-Like receptors MDA5 and LGP2 in Rainbow trout (Oncorhynchus mykiss). J. Virol. 2011, 85, 8403–8412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Liu, J.; Li, J.; Xiao, J.; Chen, H.; Lu, L.; Wang, X.; Tian, Y.; Feng, H. The antiviral signaling mediated by black carp MDA5 is positively regulated by LGP2. Fish Shellfish Immunol. 2017, 66, 360–371. [Google Scholar] [CrossRef]
  80. Paria, A.; Makesh, M.; Chauhari, A.; Purushothaman, C.S.; Rajendran, K.V. Molecular characterisation, ontogeny and expression analysis of melanoma differentiation-associated factor 5 (MDA5) from Asian seabass, Lates calcarifer. Dev. Comp. Immunol. 2018, 78, 71–82. [Google Scholar] [CrossRef]
  81. Li, Y.; Jin, S.; Zhao, X.; Luo, H.; Li, R.; Li, D.; Xiao, T. Sequence and expression analysis of the cytoplasmic pattern recognition receptor melanoma differentiation-associated gene 5 from the barbel chub Squaliobarbus curriculus. Fish Shellfish Immunol. 2019, 94, 485–496. [Google Scholar] [CrossRef] [PubMed]
  82. Zhu, Y.Y.; Xing, W.X.; Shan, S.J.; Zhang, S.Q.; Li, Y.Q.; Li, T.; An, L.; Yang, G.W. Characterization and immune response expression of the Rig-I-like receptor mda5 in common carp Cyprinus carpio. J. Fish Biol. 2016, 88, 2188–2202. [Google Scholar] [CrossRef] [PubMed]
  83. Su, J.; Huang, T.; Dong, J.; Heng, J.; Zhang, R.; Peng, L. Molecular cloning and immune responsive expression of MDA5 gene, a pivotal member of the RLR gene family from grass carp Ctenopharyngodon idella. Fish Shellfish Immunol. 2010, 28, 712–718. [Google Scholar] [CrossRef] [PubMed]
  84. Bhat, A.; Paria, A.; Deepika, A.; Sreedharan, K.; Makesh, M.; Bedekar, M.K.; Purushothaman, C.S.; Rajendran, K.V. Molecular cloning, characterisation and expression analysis of melanoma differentiation associated gene 5 (MDA5) of green chromide, Etroplus suratensis. Gene 2015, 557, 172–181. [Google Scholar] [CrossRef]
  85. Ohtani, M.; Hikima, J.; Kondo, H.; Hirono, I.; Jung, T.; Aoki, T. Characterization and antiviral function of a cytosolic sensor gene, MDA5, in Japanese flounder, Paralichthys olivaceus. Dev. Comp. Immunol. 2011, 35, 554–562. [Google Scholar] [CrossRef]
  86. Hikima, J.; Yi, M.K.; Ohtani, M.; Jung, C.Y.; Kim, Y.K.; Mun, J.Y.; Kim, Y.R.; Takeyama, H.; Aoki, T.; Jung, T.S. LGP2 expression is enhanced by interferon regulatory factor 3 in Olive flounder, Paralichthys olivaceus. PLoS ONE 2012, 7, e51522. [Google Scholar] [CrossRef] [Green Version]
  87. Gu, T.; Lu, L.; An, C.; Chen, B.; Wei, W.; Wu, X.; Xu, Q.; Chen, G. MDA5 and LGP2 acts as a key regulator though activating NF-κB and IRF3 in RLRs signaling of Mandarinfish. Fish Shellfish Immunol. 2019, 86, 1114–1122. [Google Scholar] [CrossRef]
  88. Huang, Y.; Yu, Y.; Yang, Y.; Yang, M.; Zhou, L.; Huang, X.; Qin, Q. Antiviral function of grouper MDA5 against iridovirus and nodavirus. Fish Shellfish Immunol. 2016, 54, 188–196. [Google Scholar] [CrossRef]
  89. Jia, P.; Jia, K.; Chen, L.; Le, Y.; Jin, Y.; Zhang, J.; Zhu, L.; Zhang, L.; Yi, M. Identification and characterization of the melanoma differentiation-associated gene 5 in sea perch, Lateolabrax japonicus. Dev. Comp. Immunol. 2016, 61, 161–168. [Google Scholar] [CrossRef]
  90. Mu, Y.; Li, M.; Ding, F.; Ding, Y.; Ao, J.; Hu, S.; Chen, X. De novo characterization of the spleen transcriptome of the large yellow croaker (Pseudosciaena crocea) and analysis of the immune relevant genes and pathways involved in the antiviral response. PLoS ONE 2014, 9, e101069. [Google Scholar] [CrossRef] [Green Version]
  91. Shen, B.; Hu, Y.; Zhang, S.; Zheng, J.; Zeng, L.; Zhang, J.; Zhu, A.; Wu, C. Molecular characterization and expression analyses of three RIG-I-like receptor signaling pathway genes (MDA5, LGP2 and MAVS) in Larimichthys crocea. Fish Shellfish Immunol. 2016, 55, 535–549. [Google Scholar] [CrossRef] [PubMed]
  92. Zou, P.F.; Chang, M.X.; Xue, N.N.; Liu, X.Q.; Li, J.H.; Fu, J.P.; Chen, S.N.; Nie, P. Melanoma differentiation-associated gene 5 in zebrafish provoking higher interferon-promoter activity through signalling enhancing of its shorter splicing variant. Immunology 2013, 141, 192–202. [Google Scholar] [CrossRef] [PubMed]
  93. Paria, A.; Deepika, A.; Sreedharan, K.; Makesh, M.; Chaudhari, A.; Purushothaman, C.S.; Rajendran, K.V. Identification, ontogeny and expression analysis of a novel laboratory of genetics and physiology 2 (LGP2) transcript in Asian seabass, Lates calcarifer. Fish Shellfish Immunol. 2017, 62, 265–275. [Google Scholar] [CrossRef] [PubMed]
  94. Jensen, I.; Seppola, M.; Steiro, K.; Sandaker, E.; Mennen, S. Susceptibility of Atlantic cod Gadus morhua juveniles to different routes of experimental challenge with infectious pancreatic necrosis virus (IPNV). Dis. Aquat. Org. 2009, 85, 105–113. [Google Scholar] [CrossRef] [Green Version]
  95. Xiao, J.; Yan, J.; Chen, H.; Li, J.; Tian, Y.; Feng, H. LGP2 of black carp plays an important role in the innate immune response against SVCV and GCRV. Fish Shellfish Immunol. 2016, 57, 127–135. [Google Scholar] [CrossRef]
  96. Cao, X.L.; Chen, J.J.; Cao, Y.; Nie, G.X.; Wan, Q.Y.; Wang, L.F.; Su, J.G. Identification and expression of the laboratory of genetics and physiology 2 gene in common carp Cyprinus carpio. J. Fish Biol. 2015, 86, 74–91. [Google Scholar] [CrossRef]
  97. Mohanty, A.; Sadangi, S.; Paichha, M.; Samanta, M. Molecular characterization and expressional quantification of lgp2, a modulatory co-receptor of RLR-signalling pathway in the Indian major carp Labeo rohita following pathogenic challenges and PAMP stimulations. J. Fish Biol. 2020, 96, 1399–1410. [Google Scholar] [CrossRef]
  98. Han, J.; Wang, Y.; Chu, Q.; Xu, T. The evolution and functional characterization of miiuy croaker cytosolic gene LGP2 involved in immune response. Fish Shellfish Immunol. 2016, 58, 193–202. [Google Scholar] [CrossRef]
  99. Yu, Y.; Huang, Y.; Yang, Y.; Wang, S.; Yang, M.; Huang, X.; Qin, Q. Negative regulation of the antiviral response by grouper LGP2 against fish viruses. Fish Shellfish Immunol. 2016, 56, 358–366. [Google Scholar] [CrossRef]
  100. Poynter, S.J.; Herrington-Krause, S.; Dewitte-orr, S.J. Two DExD/H-box helicases, DDX3 and DHX9, identified in rainbow trout are able to bind dsRNA. Fish Shellfish Immunol. 2019, 93, 1056–1066. [Google Scholar] [CrossRef]
  101. Jia, P.; Zhang, J.; Jin, Y.; Zeng, L.; Jia, K.; Yi, M. Characterization and expression analysis of laboratory of genetics and physiology 2 gene in sea perch, Lateolabrax japonicus. Fish Shellfish Immunol. 2015, 47, 214–220. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, Q.; Zhao, X.; Li, Z.; Wu, M.; Gui, J.; Zhang, Y. Alternative splicing transcripts of zebrafish LGP2 gene differentially contribute to IFN antiviral response. J. Immunol. 2018, 200, 688–703. [Google Scholar] [CrossRef] [PubMed]
  103. Mojzesz, M.; Klak, K.; Wojtal, P.; Adamek, M.; Podlasz, P.; Chmielewska-Krzesinska, M.; Matras, M.; Reichert, M.; Chadzinska, M.; Rakus, K. Viral infection-induced changes in the expression profile of non-RLR DExD/H-box RNA helicases (DDX1, DDX3, DHX9, DDX21 and DHX36) in zebrafish and common carp. Fish Shellfish Immunol. 2020, 104, 62–73. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, J.; Huang, X.; Yu, Y.; Zhang, J.; Ni, S.; Hu, Y.; Huang, Y.; Qin, Q. Fish DDX3X exerts antiviral function against grouper nervous necrosis virus infection. Fish Shellfish Immunol. 2017, 71, 95–104. [Google Scholar] [CrossRef] [PubMed]
  105. Murakami, S.; Morimoto, N.; Kono, T.; Sakai, M.; Hikima, J. Molecular characterization and expression of the teleost cytosolic DNA sensor genes cGAS, LSm14A, DHX9, and DHX36 in Japanese medaka, Oryzias latipes. Dev. Comp. Immunol. 2019, 99, 103402. [Google Scholar] [CrossRef] [PubMed]
  106. Quynh, N.T.; Hikima, J.; Kim, Y.; Fagutao, F.F.; Kim, M.S.; Aoki, T.; Jung, T.S. The cytosolic sensor, DDX41, activates antiviral and inflammatory immunity in response to stimulation with double-stranded DNA adherent cells of the olive flounder, Paralichthys olivaceus. Fish Shellfish Immunol. 2015, 44, 576–583. [Google Scholar] [CrossRef] [PubMed]
  107. Liu, J.; Huang, Y.; Huang, X.; Li, C.; Ni, S.; Yu, Y.; Qin, Q. Grouper DDX41 exerts antiviral activity against fish iridovirus and nodavirus infection. Fish Shellfish Immunol. 2019, 91, 40–49. [Google Scholar] [CrossRef]
  108. Chang, M.X.; Zhang, J. Alternative pre-mRNA splicing in mammals and teleost fish: A effective strategy for the regulation of immune responses against pathogen infection. Int. J. Mol. Sci. 2017, 18, 1530. [Google Scholar] [CrossRef]
  109. Gong, X.; Zhang, Q.; Gui, J.; Zhang, Y. SVCV infection triggers fish IFN response through RLR signaling pathway. Fish Shellfish Immunol. 2019, 86, 1058–1063. [Google Scholar] [CrossRef]
  110. Biacchesi, S.; Leberre, M.; Lamoureux, A.; Louise, Y.; Lauret, E.; Boudinot, P.; Bremont, M. Mitochondrial antiviral signaling protein plays a major role in induction of the fish innate immune response against RNA and DNA viruses. J. Immunol. 2009, 83, 7815–7827. [Google Scholar] [CrossRef] [Green Version]
  111. Sun, F.; Zhang, Y.; Liu, T.; Shi, J.; Wang, B.; Gui, J. Fish MITA serves as a mediator for distinct fish IFN gene activation dependent on IRF3 or IRF7. J. Immunol. 2011, 187, 2531–2539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Chen, L.; Su, J.; Yang, C.; Peng, L.; Wan, Q.; Wang, L. Functional characterizations of RIG-I to GCRV and viral/bacterial PAMPs in Grass carp Ctenopharyngodon idella. PLoS ONE 2012, 7, e42182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Gabor, K.A.; Charette, J.R.; Pietraszewski, M.J.; Wingfield, D.J.; Shim, J.S.; Millard, P.J.; Kim, C.H. A DN-mda5 transgenic zebrafish model demonstrates that Mda5 plays an important role in snakehead rhabdovirus resistance. Dev. Comp. Immunol. 2015, 51, 298–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhou, Z.; Zhang, B.; Sun, L. Poly(I:C) induces antiviral immune responses in Japanese flounder (Paralichthys olivaceus) that require TLR3 and MDA5 and is negatively regulated by Myd88. PLoS ONE 2014, 9, e112918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wan, Q.; Yang, C.; Rao, Y.; Liao, Z.; Su, J. MDA5 induces a stronger interferon response than RIG-I to GCRV infection through a mechanism involving the phosphorylation and dimerization of IRF3 and IRF7 in CIK cells. Front. Immunol. 2017, 8, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Gu, T.; Rao, Y.; Su, J.; Yang, C.; Chen, X.; Chen, L.; Yan, N. Functions of MDA5 and its domains in response to GCRV or bacterial PAMPs. Fish Shellfish Immunol. 2015, 46, 693–702. [Google Scholar] [CrossRef]
  117. Gao, F.; Lu, M.; Wang, M.; Liu, Z.; Ke, X.; Zhang, D.; Cao, J. Molecular characterization and function analysis of three RIG-I-like receptor signaling pathway genes (MDA5, LGP2 and MAVS) in Oreochromis niloticus. Fish Shellfish Immunol. 2018, 82, 101–114. [Google Scholar] [CrossRef]
  118. Chen, X.; Yang, C.; Su, J.; Rao, Y.; Gu, T. LGP2 plays extensive roles in modulating innate immune responses in Ctenopharyngodon idella kidney (CIK) cells. Dev. Comp. Immunol. 2015, 49, 138–148. [Google Scholar] [CrossRef]
  119. Rao, Y.; Wan, Q.; Yang, C.; Su, J. Grass carp laboratory of genetics and physiology 2 serves as a negative regulator in retinoic acid-inducible gene I- and melanoma differentiation-associated gene 5-mediated antiviral signaling in resting state and early stage of grass carp reovirus infection. Front. Immunol. 2017, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, Z.; Kim, T.; Bao, M.; Facchinetti, V.; Jung, S.Y.; Ghaffari, A.A.; Qin, J.; Cheng, G.; Liu, Y. DDX1, DDX21, and DHX36 helicases form a complex with the adaptor molecule TRIF to sense dsRNA in dendritic cells. Immunity 2011, 34, 866–878. [Google Scholar] [CrossRef] [Green Version]
  121. Schröder, M.; Baran, M.; Bowie, A.G. Viral targeting of DEAD box protein 3 reveals its role in TBK1/IKKε-mediated IRF activation. EMBO J. 2008, 27, 2147–2157. [Google Scholar] [CrossRef] [PubMed]
  122. Soulat, D.; Bürckstümmer, T.; Westermayer, S.; Goncalves, A.; Bauch, A.; Stefanovic, A.; Hantschel, O.; Bennett, K.L.; Decker, T.; Superti-Furga, G. The DEAD-box helicase DDX3X is a critical component of the TANK-binding kinase 1-dependent innate immune response. EMBO J. 2008, 27, 2135–2146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhang, Z.; Yuan, B.; Lu, N.; Facchinetti, V.; Liu, Y. DHX9 pairs with IPS-1 to sense double-stranded RNA in myeloid dendritic cells. J. Immunol. 2011, 187, 4501–4508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mosallanejad, K.; Sekine, Y.; Ishikura-Kinoshita, S.; Kumagai, K.; Nagano, T.; Matsuzawa, A.; Takeda, K.; Naguro, I.; Ichijo, H. The DEAH-box RNA helicase DHX15 activates NF-kB and MAPK signaling downstream of MAVS during antiviral eesponses. Immunology 2014, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  125. Moy, R.H.; Cole, B.S.; Yasunaga, A.; Gold, B.; Shankarling, G.; Varble, A.; Molleston, J.M.; ten Oever, B.R.; Lynch, K.W.; Cherry, S. Stem-loop recognition by DDX17 facilitates miRNA processing and antiviral defense. Cell 2014, 158, 764–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Ruan, J.; Cao, Y.; Ling, T.; Li, P.; Wu, S.; Peng, D.; Wang, Y.; Jia, X.; Chen, S.; Xu, A.; et al. DDX23, an evolutionary conserved dsRNA sensor, participates in innate antiviral responses by pairing with TRIF or MAVS. Front. Immunol. 2019, 10, 2202. [Google Scholar] [CrossRef]
  127. Sugimoto, N.; Mitoma, H.; Kim, T.; Hanabuchi, S.; Liu, Y. Helicase proteins DHX29 and RIG-I cosense cytosolic nucleic acids in the human airway system. Proc. Natl. Acad. Sci. USA 2014, 111, 7747–7752. [Google Scholar] [CrossRef] [Green Version]
  128. Zhang, Z.; Yuan, B.; Bao, M.; Lu, N.; Kim, T.; Liu, Y. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nat. Immunol. 2011, 12, 959–965. [Google Scholar] [CrossRef] [Green Version]
  129. Miyashita, M.; Oshiumi, H.; Matsumoto, M.; Seya, T. DDX60, a DExD/H-box helicase, is a novel antiviral factor promoting RIG-I-Like receptor-mediated signaling. Mol. Cell. Biol. 2011, 31, 3802–3819. [Google Scholar] [CrossRef] [Green Version]
  130. Mitoma, H.; Hanabuchi, S.; Kim, T.; Bao, M.; Zhang, Z.; Sugimoto, N.; Liu, Y. The DHX33 RNA helicase senses cytosolic RNA and activates the NLRP3 inflammasome. Immunity 2013, 39, 123–135. [Google Scholar] [CrossRef] [Green Version]
  131. Kim, T.; Pazhoor, S.; Bao, M.; Zhang, Z.; Hanabuchi, S.; Facchinetti, V.; Bover, L.; Plumas, J.; Chaperot, L.; Qin, J.; et al. Aspartate-glutamate-alanine-histidine box motif (DEAH)/RNA helicase A helicases sense microbial DNA in human plasmacytoid dendritic cells. Proc. Natl. Acad. Sci. USA 2010, 107, 15181–15186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Oshiumi, H.; Sakai, K.; Matsumoto, M.; Seya, T. DEAD/H BOX 3 (DDX3) helicase binds the RIG-I adaptor IPS-1 to up-regulate IFN-β-inducing potential. Eur. J. Immunol. 2010, 40, 940–948. [Google Scholar] [CrossRef] [PubMed]
  133. Parvatiyar, K.; Zhang, Z.; Teles, R.M.; Ouyang, S.; Jiang, Y.; Iyer, S.S.; Zaver, S.A.; Schenk, M.; Zeng, S.; Zhong, W.; et al. The helicase DDX41 recognizes the bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat. Immunol. 2012, 13, 1155–1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Tian, C.; Tan, S.; Bao, L.; Zeng, Q.; Liu, S.; Yang, Y.; Zhong, X.; Liu, Z. DExD/H-box RNA helicase genes are differentially expressed between males and females during the critical period of male sex differentiation in channel catfish. Comp. Biochem. Physiol. Part D Genom. Proteom. 2017, 22, 109–119. [Google Scholar] [CrossRef] [Green Version]
  135. Wang, Z.; Liu, W.; Song, H.; Wang, H.; Liu, J.; Zhao, H.; Du, X.; Zhang, Q. Comparative evolution of duplicated Ddx3 genes in teleosts: Insights from Japanese flounder, Paralichthys olivaceus. G3 Genes Genomes Genet. 2015, 5, 1765–1773. [Google Scholar] [CrossRef] [Green Version]
  136. Gan, Z.; Cheng, J.; Hou, J.; Xia, H.; Chen, W.; Xia, L.; Nie, P.; Lu, Y. Molecular and functional characterization of tilapia DDX41 in IFN regulation. Fish Shellfish Immunol. 2020, 99, 386–391. [Google Scholar] [CrossRef]
  137. Lauksund, S.; Svingerud, T.; Bergan, V.; Robertsen, B. Atlantic salmon IPS-1 mediates induction of IFNa1 and activation of NF-κB and localizes to mitochondria. Dev. Comp. Immunol. 2009, 33, 1196–1204. [Google Scholar] [CrossRef]
  138. Simora, R.M.C.; Ohtani, M.; Hikima, J.; Kondo, H.; Hirono, I.; Jung, T.S.; Aoki, T. Molecular cloning and antiviral activity of IFN-β promoter stimulator-1 (IPS-1) gene in Japanese flounder, Paralichthys olivaceus. Fish Shellfish Immunol. 2010, 29, 979–986. [Google Scholar] [CrossRef]
  139. Xiang, Z.; Qi, L.; Chen, W.; Dong, C.; Liu, Z.; Liu, D.; Huang, M.; Li, W.; Yang, G.; Weng, S.; et al. Characterization of a TnMAVS protein from Tetraodon nigroviridis. Dev. Comp. Immunol. 2011, 35, 1103–1115. [Google Scholar] [CrossRef]
  140. Kasthuri, S.R.; Wan, Q.; Whang, I.; Lim, B.; Yeo, S.; Choi, C.Y.; Lee, J. Functional characterization of the evolutionarily preserved mitochondrial antiviral signaling protein (MAVS) from rock bream, Oplegnathus fasciatus. Fish Shellfish Immunol. 2014, 40, 399–406. [Google Scholar] [CrossRef]
  141. Feng, X.; Zhang, Y.; Yang, C.; Liao, L.; Wang, Y.; Su, J. Functional characterizations of IPS-1 in CIK cells: Potential roles in regulating IFN-I response dependent on IRF7 but not IRF3. Dev. Comp. Immunol. 2015, 53, 23–32. [Google Scholar] [CrossRef] [PubMed]
  142. Lu, L.F.; Li, S.; Lu, X.; Zhang, Y. Functions of the two zebrafish MAVS variants are opposite in the induction of IFN1 by targeting IRF7. Fish Shellfish Immunol. 2015, 45, 574–582. [Google Scholar] [CrossRef] [PubMed]
  143. Zhao, X.; Xiao, T.; Jin, S.; Wang, J.; Wang, J.; Luo, H.; Li, R.; Sun, T.; Zou, J.; Li, Y. Characterization and immune function of the interferon-β promoter stimulator-1 in the barbel chub, Squaliobarbus curriculus. Dev. Comp. Immunol. 2020, 104. [Google Scholar] [CrossRef] [PubMed]
  144. Krishnan, R.; Girish Babu, P.; Jeena, K.; Tripathi, G.; Pani Prasad, K. Molecular characterization, ontogeny and expression profiling of mitochondrial antiviral signaling adapter, MAVS from Asian seabass Lates calcarifer, Bloch (1790). Dev. Comp. Immunol. 2018, 79, 175–185. [Google Scholar] [CrossRef] [PubMed]
  145. Krishnan, R.; Jeena, K.; Mushtaq, Z.; Shyam, K.U.; Kurcheti, P.P. Antiviral activity of transiently expressed mitochondrial antiviral signaling adapter, MAVS orthologue from Asian seabass. Fish Shellfish Immunol. 2018, 76, 183–186. [Google Scholar] [CrossRef]
  146. Xiao, J.; Fu, Y.; Wu, H.; Chen, X.; Liu, S.; Feng, H. MAVS of triploid hybrid of red crucian carp and allotetraploid possesses the improved antiviral activity compared with the counterparts of its parents. Fish Shellfish Immunol. 2019, 89, 18–26. [Google Scholar] [CrossRef]
  147. Huang, Y.; Zhang, J.; Ouyang, Z.; Liu, J.; Zhang, Y.; Hu, Y.; Huang, X.; Qin, Q. Grouper MAVS functions as a crucial antiviral molecule against nervous necrosis virus infection. Fish Shellfish Immunol. 2018, 72, 14–22. [Google Scholar] [CrossRef]
  148. Seth, R.B.; Sun, L.; Ea, C.K.; Chen, Z.J. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-κB and IRF3. Cell 2005, 122, 669–682. [Google Scholar] [CrossRef] [Green Version]
  149. Jia, P.; Jin, Y.; Chen, L.; Zhang, J.; Jia, K.; Yi, M. Molecular characterization and expression analysis of mitochondrial antiviral signaling protein gene in sea perch, Lateolabrax japonicus. Dev. Comp. Immunol. 2016, 55, 188–193. [Google Scholar] [CrossRef]
  150. Chen, W.Q.; Hu, Y.W.; Zou, P.F.; Ren, S.S.; Nie, P.; Chang, M.X. MAVS splicing variants contribute to the induction of interferon and interferon-stimulated genes mediated by RIG-I-like receptors. Dev. Comp. Immunol. 2015, 49, 19–30. [Google Scholar] [CrossRef]
  151. Su, J.; Huang, T.; Yang, C.; Zhang, R. Molecular cloning, characterization and expression analysis of interferon-β promoter stimulator 1 (IPS-1) gene from grass carp Ctenopharyngodon idella. Fish Shellfish Immunol. 2011, 30, 317–323. [Google Scholar] [CrossRef] [PubMed]
  152. Xiao, J.; Yan, C.; Zhou, W.; Li, J.; Wu, H.; Chen, T.; Feng, H. CARD and TM of MAVS of black carp play the key role in its self-association and antiviral ability. Fish Shellfish Immunol. 2017, 63, 261–269. [Google Scholar] [CrossRef] [PubMed]
  153. Zhou, W.; Zhou, J.; Lv, Y.; Qu, Y.; Chi, M.; Li, J.; Feng, H. Identification and characterization of MAVS from black carp Mylopharyngodon piceus. Fish Shellfish Immunol. 2015, 43, 460–468. [Google Scholar] [CrossRef] [PubMed]
  154. Zou, P.F.; Chang, M.X.; Li, Y.; Xue, N.N.; Li, J.H.; Chen, S.N.; Nie, P. NOD2 in zebrafish functions in antibacterial and also antiviral responses via NF-κB, and also MDA5, RIG-I and MAVS. Fish Shellfish Immunol. 2016, 55, 173–185. [Google Scholar] [CrossRef]
  155. Zhang, J.; Zhang, Y.B.; Wu, M.; Wang, B.; Chen, C.; Gui, J.F. Fish MAVS is involved in RLR pathway-mediated IFN response. Fish Shellfish Immunol. 2014, 41, 222–230. [Google Scholar] [CrossRef]
  156. Kranzusch, P.J. cGAS and CD-NTase enzymes: Structure, mechanism, and evolution. Curr. Opin. Struct. Biol. 2019, 59, 178–187. [Google Scholar] [CrossRef]
  157. Civril, F.; Deimling, T.; de Oliveira Mann, C.C.; Ablasser, A.; Moldt, M.; Witte, G.; Hornung, V.; Hopfner, K.P. Structural mechanism of cytosolic DNA sensing by cGAS. Nature 2013, 498, 332–337. [Google Scholar] [CrossRef] [Green Version]
  158. Kranzusch, P.J.; Lee, A.S.; Berger, J.; Doudna, J. Structure of human cGAS reveals a conserved family of second-messenger enzymes in innate immunity. Cell Rep. 2013, 3, 1362–1368. [Google Scholar] [CrossRef] [Green Version]
  159. Tao, J.; Zhang, X.; Jin, J.; Du, X.; Lian, T.; Yang, J.; Zhou, X.; Jiang, Z.; Su, X. Nonspecific DNA binding of cGAS N terminus promotes cGAS activation. J. Immunol. 2017, 198, 3627–3636. [Google Scholar] [CrossRef] [Green Version]
  160. Liu, Z.; Ji, J.; Jiang, X.; Shao, T.; Fan, D.; Jiang, X.; Lin, A.; Xiang, L.; Shao, J. Characterization of cGAS homologs in innate and adaptive mucosal immunities in zebrafish gives evolutionary insights into cGAS-STING pathway. FASEB J. 2020, 34, 7786–7809. [Google Scholar] [CrossRef] [Green Version]
  161. Ablasser, A.; Chen, Z.J. cGAS in action: Expanding roles in immunity and inflammation. Science 2019, 363, eaat8657. [Google Scholar] [CrossRef] [PubMed]
  162. Li, T.; Chen, Z.J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299. [Google Scholar] [CrossRef] [PubMed]
  163. Wan, D.; Jiang, W.; Hao, J. Research Advances in How the cGAS-STING pathway controls the cellular inflammatory response. Front. Immunol. 2020, 11, 615. [Google Scholar] [CrossRef] [PubMed]
  164. Zhou, W.; Whiteley, A.T.; de Oliveira Mann, C.C.; Morehouse, B.R.; Nowak, R.P.; Fischer, E.S.; Gray, N.S.; Mekalanos, J.J.; Kranzusch, P.J. Structure of the human cGAS-DNA complex reveals enhanced control of immune surveillance. Cell 2018, 174, 300–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Luecke, S.; Holleufer, A.; Christensen, M.H.; Jønsson, K.L.; Boni, G.A.; Sørensen, L.K.; Johannsen, M.; Jakobsen, M.R.; Hartmann, R.; Paludan, S.R. cGAS is activated by DNA in a length-dependent manner. EMBO Rep. 2017, 18, 1707–1715. [Google Scholar] [CrossRef]
  166. García-Belmonte, R.; Perez-Nunez, D.; Pittau, M.; Richt, J.A.; Revilla, Y. African swine fever virus armenia/07 virulent strain controls interferon beta production through the cGAS-STING pathway. J. Virol. 2019, 93, e02298-18. [Google Scholar] [CrossRef] [Green Version]
  167. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef] [Green Version]
  168. Huang, J.; You, H.; Su, C.; Li, Y.; Chen, S.; Zheng, C. Herpes simplex virus 1 tegument protein VP22 abrogates cGAS/STING-mediated antiviral innate immunity. J. Virol. 2018, 92, e00841-18. [Google Scholar] [CrossRef] [Green Version]
  169. Verrier, E.R.; Yim, S.A.; Heydmann, L.; El-Saghire, H.; Bach, C.; Turon-Lagot, V.; Mailly, L.; Durand, S.C.; Lucifora, J.; Durantel, D.; et al. Hepatitis B virus evasion from cyclic guanosine monophosphate-adenosine monophosphate synthase sensing in human hepatocytes. Hepatology 2018, 68, 1695–1709. [Google Scholar] [CrossRef] [Green Version]
  170. Aguirre, S.; Fernandez-Sesma, A. Collateral damage during dengue virus Infection: Making sense of DNA by cGAS. J. Virol. 2017, 91, e01081-16. [Google Scholar] [CrossRef] [Green Version]
  171. Marinho, F.V.; Benmerzoug, S.; Rose, S.; Campos, P.C.; Marques, J.T.; Báfica, A.; Barber, G.; Ryffel, B.; Oliveira, S.C.; Quesniaux, V.F.J. The cGAS/STING pathway is important for dendritic cell activation but is not essential to induce protective immunity against Mycobacterium tuberculosis infection. J. Innate Immun. 2018, 10, 239–252. [Google Scholar] [CrossRef] [PubMed]
  172. Collins, A.C.; Cai, H.; Li, T.; Franco, L.H.; Li, X.D.; Nair, V.R.; Scharn, C.R.; Stamm, C.E.; Levine, B.; Chen, Z.J.; et al. Cyclic GMP-AMP synthase is an innate immune DNA sensor for Mycobacterium tuberculosis. Cell Host Microbe 2015, 17, 820–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Watson, R.O.; Bell, S.L.; MacDuff, D.A.; Kimmey, J.M.; Diner, E.J.; Olivas, J.; Vance, R.E.; Stallings, C.L.; Virgin, H.W.; Cox, J.S. The cytosolic sensor cGAS detects Mycobacterium tuberculosis DNA to induce type I interferons and activate autophagy. Cell Host Microbe 2015, 17, 811–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Nandakumar, R.; Tschismarov, R.; Meissner, F.; Prabakaran, T.; Krissanaprasit, A.; Farahani, E.; Zhang, B.C.; Assil, S.; Martin, A.; Bertrams, W.; et al. Intracellular bacteria engage a STING-TBK1-MVB12b pathway to enable paracrine cGAS-STING signaling. Nat. Microbiol. 2019, 4, 701–713. [Google Scholar] [CrossRef]
  175. Ruiz-Moreno, J.S.; Hamann, L.; Jin, L.; Sander, L.E.; Puzianowska-Kuznicka, M.; Cambier, J.; Witzenrath, M.; Schumann, R.R.; Suttorp, N.; Opitz, B. The cGAS/STING pathway detects Streptococcus pneumoniae but appears dispensable for antipneumococcal defense in mice and humans. Infect. Immun. 2017, 86, e00849-17. [Google Scholar] [CrossRef] [Green Version]
  176. Gallego-Marin, C.; Schrum, J.E.; Andrade, W.A.; Shaffer, S.A.; Giraldo, L.F.; Lasso, A.M.; Kurt-Jones, E.A.; Fitzgerald, K.A.; Golenbock, D.T. Cyclic GMP–AMP synthase is the cytosolic sensor of Plasmodium falciparum genomic DNA and activates type I IFN in malaria. J. Immunol. 2018, 200, 768–774. [Google Scholar] [CrossRef] [Green Version]
  177. Das, S.; Kumar, A.; Mandal, A.; Abhishek, K.; Verma, S.; Kumar, A.; Das, P. Nucleic acid sensing activates the innate cytosolic surveillance pathway and promotes parasite survival in visceral leishmaniasis. Sci. Rep. 2019, 9, 9825. [Google Scholar] [CrossRef] [Green Version]
  178. Ge, R.; Zhou, Y.; Peng, R.; Wang, R.; Li, M.; Zhang, Y.; Zheng, C.; Wang, C. Conservation of the STING-mediated cytosolic DNA sensing pathway in zebrafish. J. Virol. 2015, 89, 7696–7706. [Google Scholar] [CrossRef] [Green Version]
  179. Rolf, J.; Siedentop, R.; Lütz, S.; Rosenthal, K. Screening and identification of novel cGAS homologues using a combination of in vitro and in vivo protein synthesis. Int. J. Mol. Sci. 2019, 21, 105. [Google Scholar] [CrossRef] [Green Version]
  180. Zhou, Y.; Lu, L.; Lu, X.; Li, S.; Zhang, Y. Grass carp cGASL negatively regulates fish IFN response by targeting MITA. Fish Shellfish Immunol. 2019, 94, 871–879. [Google Scholar] [CrossRef]
  181. Su, C.; Zheng, C. Herpes Simplex Virus 1 abrogates the cGAS/STING-mediated cytosolic DNA sensing pathway via its virion host Shutoff Protein, UL41. J. Virol. 2017, 91, e02414-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Dai, P.; Wang, W.; Yang, N.; Serna-Tamayo, C.; Ricca, J.M.; Zamarin, D.; Shuman, S.; Merghoub, T.; Wolchok, J.D.; Deng, L. Intratumoral delivery of inactivated modified vaccinia virus ankara (iMVA) induces systemic antitumor immunity via STING and batf3-dependent dendritic cells. Sci. Immunol. 2017, 2, eaal1713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Thomsen, M.K.; Nandakumar, R.; Stadler, D.; Malo, A.; Marin-Valls, R.; Wang, F.; Reinert, L.S.; Dagnæs-Hansen, F.; Hollensen, A.K.; Mikkelsen, J.G.; et al. Lack of immunological DNA sensing in hepatocytes facilitates hepatitis B virus infection. Hepatology 2016, 64, 746–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Wong, E.B.; Montoya, B.; Ferez, M.; Stotesbury, C.; Sigal, L.J. Resistance to ectromelia virus infection requires cGAS in bone marrow-derived cells which can be bypassed with cGAMP therapy. PLoS Pathog. 2019, 15, e1008239. [Google Scholar] [CrossRef] [Green Version]
  185. Cheng, W.; He, X.; Jia, H.; Chen, G.; Jin, Q.; Long, Z.; Jing, Z. The cGAS-STING signaling pathway is required for the innate immune response against ectromelia virus. Front. Immunol. 2018, 9, 1297. [Google Scholar] [CrossRef] [Green Version]
  186. Paijo, J.; Döring, M.; Spanier, J.; Grabski, E.; Nooruzzaman, M.; Schmidt, T.; Witte, G.; Messerle, M.; Hornung, V.; Kaever, V.; et al. cGAS senses human cytomegalovirus and induces type I interferon responses in human monocyte-derived cells. PLoS Pathog. 2016, 12, e1005546. [Google Scholar] [CrossRef]
  187. Gao, D.; Wu, J.; Wu, Y.T.; Du, F.; Aroh, C.; Yan, N.; Sun, L.; Chen, Z.J. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 2013, 341, 903–906. [Google Scholar] [CrossRef] [Green Version]
  188. Lieb, B.; Carl, M.; Hock, R.; Gebauer, D.; Scheer, U. Identification of a novel mRNA-associated protein in oocytes of Pleurodeles waltl and Xenopus laevis. Exp. Cell Res. 1998, 245, 272–281. [Google Scholar] [CrossRef]
  189. Tanaka, K.J.; Ogawa, K.; Takagi, M.; Imamoto, N.; Matsumoto, K.; Tsujimoto, M. RAP55, a cytoplasmic mRNP component, represses translation in Xenopus oocytes. J. Biol. Chem. 2006, 281, 40096–40106. [Google Scholar] [CrossRef] [Green Version]
  190. Yang, W.; Yu, J.H.; Gulick, T.; Bloch, K.D.; Bloch, D.B. RNA-associated protein 55 (RAP55) localizes to mRNA processing bodies and stress granules. RNA 2006, 12, 547–554. [Google Scholar] [CrossRef] [Green Version]
  191. Li, Y.; Chen, R.; Zhou, Q.; Xu, Z.; Li, C.; Wang, S.; Mao, A.; Zhang, X.; He, W.; Shu, H.B. LSm14A is a processing body-associated sensor of viral nucleic acids that initiates cellular antiviral response in the early phase of viral infection. Proc. Natl. Acad. Sci. USA 2012, 109, 11770–11775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Wen, G.; Zhang, Y.; Zhang, X.; Hu, H.; Zhang, H.; Cheng, C.; Wang, X.; Li, X.; Fang, W. Functional characterization of porcine LSm14A in IFN-β induction. Vet. Immunol. Immunopathol. 2013, 155, 110–116. [Google Scholar] [CrossRef] [PubMed]
  193. Li, Z.; Chen, R.; Zhao, J.; Qi, Z.; Ji, L.; Zhen, Y.; Liu, B. LSM14A inhibits porcine reproductive and respiratory syndrome virus (PRRSV) replication by activating IFN-β signaling pathway in Marc-145. Mol. Cell. Biochem. 2015, 399, 247–256. [Google Scholar] [CrossRef] [PubMed]
  194. Liu, T.; Yang, Q.; Li, M.; Zhong, B.; Ran, Y.; Liu, L.; Yang, Y.; Wang, Y.; Shu, H. LSm14A plays a critical role in antiviral immune responses by regulating MITA level in a cell-specific manner. J. Immunol. 2016, 196, 5101–5111. [Google Scholar] [CrossRef] [PubMed]
  195. Hua, K.; Li, H.; Chen, H.; Foda, M.F.; Luo, R.; Jin, H. Functional characterization of duck LSm14A in IFN-β induction. Dev. Comp. Immunol. 2017, 76, 255–261. [Google Scholar] [CrossRef] [PubMed]
  196. Tian, L.; Wu, C.; Wen, G.; Li, C. Transcriptional response of LSm14A after infection of blue eggshell layers with Newcastle disease viruses. J. Vet. Med. Sci. 2019, 81, 1468–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Ko, J.H.; Asano, A.; Kon, Y.; Watanabe, T.; Agui, T. Characterization of the chicken PKR: Polymorphism of the gene and antiviral activity against vesicular stomatitis virus. Jpn. J. Vet. Res. 2004, 51, 123–133. [Google Scholar] [CrossRef]
  198. Meurs, E.; Chong, K.; Galabru, J.; Thomas, N.S.B.; Kerr, I.M.; Williams, R.G.; Hovanessian, A. G Molecular cloning and characterization of the human double-stranded RNA-activated protein kinase induced by interferon. Cell 1990, 62, 379–390. [Google Scholar] [CrossRef]
  199. Rothenburg, S.; Deigendesch, N.; Dey, M.; Dever, T.E.; Tazi, L. Double-stranded RNA-activated protein kinase PKR of fishes and amphibians: Varying the number of double-stranded RNA binding domains and lineage-specific duplications. BMC Biol. 2008, 6, 1–19. [Google Scholar] [CrossRef] [Green Version]
  200. Zhu, R.; Zhang, Y.B.; Zhang, Q.Y.; Gui, J.F. Functional domains and the antiviral effect of the double-stranded RNA-dependent protein kinase PKR from Paralichthys olivaceus. J. Virol. 2008, 82, 6889–6901. [Google Scholar] [CrossRef] [Green Version]
  201. Garcia, M.A.; Gil, J.; Ventoso, I.; Guerra, S.; Domingo, E.; Rivas, C.; Esteban, M. Impact of protein kinase PKR in cell biology: From antiviral to antiproliferative action. Microbiol. Mol. Biol. Rev. 2006, 70, 1032–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Hu, C.Y.; Zhang, Y.B.; Huang, G.P.; Zhang, Q.Y.; Gui, J.F. Molecular cloning and characterisation of a fish PKR-like gene from cultured CAB cells induced by UV inactivated virus. Fish Shellfish Immunol. 2004, 17, 353–366. [Google Scholar] [CrossRef] [PubMed]
  203. Rothenburg, S.; Deigendesch, N.; Dittmar, K.; Koch-Nolte, F.; Haag, F.; Lowenhaupt, K.; Rich, A. A PKR-like eukaryotic initiation factor 2a kinase from zebrafish contains Z-DNA binding domains instead of dsRNA binding domains. Proc. Natl. Acad. Sci. USA 2005, 102, 1602–1607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Zhang, Y.B.; Gui, J.F. Molecular regulation of interferon antiviral response in fish. Dev. Comp. Immunol. 2012, 38, 193–202. [Google Scholar] [CrossRef] [PubMed]
  205. Schwartz, T.; Rould, M.A.; Lowenhaupt, K.; Herbert, A.; Rich, A. Crystal structure of the Zα domain of the human editing enzyme ADAR1 bound to left-handed Z-DNA. Science 1999, 284, 1841–1845. [Google Scholar] [CrossRef] [PubMed]
  206. Bergan, V.; Jagus, R.; Lauksund, S.; Kileng, Ø.; Robertsen, B. The Atlantic salmon Z-DNA binding protein kinase phosphorylates translation initiation factor 2 alpha and constitutes a unique orthologue to the mammalian dsRNA-activated protein kinase R. FEBS J. 2007, 275, 184–197. [Google Scholar] [CrossRef] [PubMed]
  207. Liu, T.K.; Zhang, Y.B.; Liu, Y.; Sun, F.; Gui, J.F. Cooperative roles of fish protein kinase containing Z-DNA binding domains and double-stranded RNA-dependent protein kinase in interferon-mediated antiviral response. J. Virol. 2011, 85, 12769–12780. [Google Scholar] [CrossRef] [Green Version]
  208. Herbert, A.; Alfken, J.; Kim, Y.G.; Mian, I.S.; Nishikura, K.; Rich, A. A Z-DNA binding domain present in the human editing enzyme, double-stranded RNA adenosine deaminase. Proc. Natl. Acad. Sci. USA 1997, 94, 8421–8426. [Google Scholar] [CrossRef] [Green Version]
  209. Schwartz, T.; Behlke, J.; Lowenhaupt, K.; Heinemann, U.; Rich, A. Structure of the DLM-1-Z-DNA complex reveals a conserved family of Z-DNA-binding proteins. Nat. Struct. Biol. 2001, 8, 761–765. [Google Scholar] [CrossRef]
  210. Wu, C.; Zhang, Y.; Hu, C. PKZ, a Fish-Unique eIF2α Kinase Involved in Innate Immune Response. Front. Immunol. 2020, 11, 585. [Google Scholar] [CrossRef]
  211. Hu, Y.S.; Li, W.; Li, D.M.; Liu, Y.; Fan, L.H.; Rao, Z.C.; Lin, G.; Hu, C.Y. Cloning, expression and functional analysis of PKR from grass carp (Ctenopharyngodon idellus). Fish Shellfish Immunol. 2013, 35, 1874–1881. [Google Scholar] [CrossRef] [PubMed]
  212. Del Castillo, C.S.; Hikima, J.; Ohtani, M.; Jung, T.S.; Aoki, T. Characterization and functional analysis of two PKR genes in fugu (Takifugu rubripes). Fish Shellfish Immunol. 2012, 32, 79–88. [Google Scholar] [CrossRef] [PubMed]
  213. Zenke, K.; Nam, Y.K.; Kim, K.H. Molecular cloning and expression analysis of double-stranded RNA-dependent protein kinase (PKR) in rock bream (Oplegnathus fasciatus). Vet. Immunol. Immunopathol. 2010, 133, 290–295. [Google Scholar] [CrossRef] [PubMed]
  214. Perelygin, A.A.; Lear, T.L.; Zharkikh, A.A.; Brinton, M.A. Comparative analysis of vertebrate EIF2AK2 (PKR) genes and assignment of the equine gene to ECA15q24-q25 and the bovine gene to BTA11q12-q15. Genet. Sel. Evol. 2006, 38, 551–563. [Google Scholar] [CrossRef]
  215. Su, J.; Zhu, Z.; Wang, Y. Molecular cloning, characterization and expression analysis of the PKZ gene in rare minnow Gobiocypris rarus. Fish Shellfish Immunol. 2008, 25, 106–113. [Google Scholar] [CrossRef]
  216. Yang, P.J.; Wu, C.X.; Li, W.; Fan, L.H.; Lin, G.; Hu, C.Y. Cloning and functional analysis of PKZ (PKR-like) from grass carp (Ctenopharyngodon idellus). Fish Shellfish Immunol. 2011, 31, 1173–1178. [Google Scholar] [CrossRef]
Figure 1. RLR (A), cGAS-STING (B) and non-RLR DExD/H-box RNA helicases (A,B) pathways detect cytoplasmic nucleic acids and activate type I IFN and pro-inflammatory cytokines. Dotted lines represents pathways confirmed in fish. (A) RIG-I and MDA5 upon binding of viral RNA induce signal transduction in MAVS-dependent manner leading to the induction of the expression of type I IFNs and pro-inflammatory cytokines. LGP2 does not possess signaling CARD domains and its role in the mediating of antiviral response is unclear. LGP2 receptor has been shown to function as a positive or negative regulator of RLR-mediated signaling. Viral RNA in cytoplasm is also recognized by non-RLR DExD/H-box RNA helicases: DDX3 has been proposed to sensitize RLR to sense viral RNA. It acts as a signaling intermediate downstream of TBK1 and IKKε and as a transcriptional regulator of the ifnb promoter. DHX9 was shown to sense dsRNA and to signal in a MAVS-dependent manner. A complex of DDX1-DDX21-DHX36 has been suggested to detect dsRNA and to signal in a TRIF-dependent manner. This leads to induction of the expression of type I IFNs and pro-inflammatory cytokines. (B) Upon recognition of virus or bacterial-derived DNA released into the cytoplasm, viral DNA synthesized by reverse transcription, and upon infection-induced mitochondrial dsDNA damaged and leaked cGAS synthesizes 2′3′-cGAMP. 2′3′-cGAMP binds to STING in the ER and causes its translocation to the Golgi system. STING activates TBK1-IRF3 and NF-κB and induces the production of type I IFNs and pro-inflammatory cytokines. DNA in cytoplasm is also recognized by non-RLR DExD/H-box RNA helicases: DDX41 upon binding of cytoplasmic DNA signals in a STING-dependent manner while DHX9 and DHX36 upon binding of cytoplasmic DNA signal in a MyD88-depending manner. This leads to induction of the expression of type I IFNs and pro-inflammatory cytokines.
Figure 1. RLR (A), cGAS-STING (B) and non-RLR DExD/H-box RNA helicases (A,B) pathways detect cytoplasmic nucleic acids and activate type I IFN and pro-inflammatory cytokines. Dotted lines represents pathways confirmed in fish. (A) RIG-I and MDA5 upon binding of viral RNA induce signal transduction in MAVS-dependent manner leading to the induction of the expression of type I IFNs and pro-inflammatory cytokines. LGP2 does not possess signaling CARD domains and its role in the mediating of antiviral response is unclear. LGP2 receptor has been shown to function as a positive or negative regulator of RLR-mediated signaling. Viral RNA in cytoplasm is also recognized by non-RLR DExD/H-box RNA helicases: DDX3 has been proposed to sensitize RLR to sense viral RNA. It acts as a signaling intermediate downstream of TBK1 and IKKε and as a transcriptional regulator of the ifnb promoter. DHX9 was shown to sense dsRNA and to signal in a MAVS-dependent manner. A complex of DDX1-DDX21-DHX36 has been suggested to detect dsRNA and to signal in a TRIF-dependent manner. This leads to induction of the expression of type I IFNs and pro-inflammatory cytokines. (B) Upon recognition of virus or bacterial-derived DNA released into the cytoplasm, viral DNA synthesized by reverse transcription, and upon infection-induced mitochondrial dsDNA damaged and leaked cGAS synthesizes 2′3′-cGAMP. 2′3′-cGAMP binds to STING in the ER and causes its translocation to the Golgi system. STING activates TBK1-IRF3 and NF-κB and induces the production of type I IFNs and pro-inflammatory cytokines. DNA in cytoplasm is also recognized by non-RLR DExD/H-box RNA helicases: DDX41 upon binding of cytoplasmic DNA signals in a STING-dependent manner while DHX9 and DHX36 upon binding of cytoplasmic DNA signal in a MyD88-depending manner. This leads to induction of the expression of type I IFNs and pro-inflammatory cytokines.
Ijms 21 07289 g001
Table 1. The upregulation of the expression of RLR and non-RLR DExD/H-box RNA helicases induced by viral/bacterial infection or PAMPs stimulation in fish.
Table 1. The upregulation of the expression of RLR and non-RLR DExD/H-box RNA helicases induced by viral/bacterial infection or PAMPs stimulation in fish.
SpeciesCell Line/TissueInfection/TreatmentReferences
rig-I
Atlantic salmon
(Salmo salar)
In vitro: TO cell linesalmon alphavirus subtype 3 (SAV-3), infectious pancreatic necrosis virus (IPNV), infectious salmon anaemia virus (ISAV)[59,60]
Ex vivo: erythrocytes piscine orthoreovirus (PRV)[61]
In vivo: head kidneyinfectious pancreatic necrosis virus (IPNV)[62]
Channel catfish
(Ictalurus punctatus)
In vitro: CCO cells linechannel catfish virus (CCV)[63]
In vivo: liverEdwardsiella ictaluri[63]
Common carp
(Cyprinus carpio)
In vivo: spleen, head kidney, intestinespring viremia of carp virus (SVCV)[64]
Gibel carp
(Carassius gibelio)
In vivo: head kidney, spleen, liver Carassius auratus herpesvirus (CaHV)[65]
Grass carp
(Ctenopharyngodon idella)
In vitro: CIK cell linegrass carp reovirus (GCRV), poly(I:C)[66]
In vitro: primary trunk kidney cellsgrass carp reovirus (GCRV), poly(I:C), LPS, PGN[67]
In vivo: spleen, liver, trunk kidney, hepatopancreasgrass carp reovirus (GCRV)[66,67,68]
Japanese eel
(Anguilla japonica)
In vivo: liver, spleen, kidneypoly(I:C), LPS, Aeromonas hydrophila[69]
In vivo: liver, spleen, head kidney, skin, intestine, and gillspoly(I:C)[70]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linenervous necrosis virus (NNV), snakehead fish vesiculovirus (SHVV), spring viremia of carp virus (SVCV), Edwardsiella tarda[71,72,73]
In vivo: larvaespring viremia of carp virus (SVCV), poly(I:C)[74,75]
In vivo: visceral tissuespoly(I:C)[76]
In vivo: spleen, kidneytilapia lake virus (TiLV) [77]
mda5
Atlantic salmon
(Salmo salar)
In vitro: TO cell line salmon alphavirus subtype 3 (SAV-3), infectious pancreatic necrosis virus (IPNV), infectious salmon anaemia virus (ISAV), salmonid alphavirus [59,60,78]
In vivo: head kidneyinfectious pancreatic necrosis virus (IPNV)[62]
Black carp
(Mylopharyngodon piceus)
In vitro: MPF cell linespring viremia of carp virus (SVCV), grass carp Reovirus (GCRV), poly(I:C)[79]
In vivo: heart, liver, spleen, kidney, intestine, skin, gillgrass carp reovirus (GCRV), spring viremia of carp virus SVCV[79]
Asian seabass
(Lates calcarifer)
In vitro: SISK cell linepoli (I:C), LPS, PGN[80]
In vivo: spleen, kidney, gills, heart, liver, intestinepoli (I:C)
In vivo: gills, heart, kidney, liver, intestineVibrio alginolyticus
In vivo: gills, heart, kidney, liver, intestineStaphylococcus aureus
Barbel chub
(Squaliobarbus curriculus)
In vivo: livergrass carp reovirus (GCRV)[81]
Channel catfish
(Ictalurus punctatus)
In vitro: CCO cell line channel catfish virus (CCV)[63]
In vivo: liverEdwardsiella ictaluri.[63]
Common carp
(Cyprinus carpio)
In vitro: peripheral blood leukocytespoly(I:C)[82]
In vivo: liver, spleen, head kidney, foregut, hindgut, gills, skinpoly(I:C), Aeromonas hydrophila[82]
Grass carp
(Ctenopharyngodon idella)
In vitro: primarly trunk kidney cellsgrass carp reovirus (GCRV), poly(I:C), LPS, PGN[67]
In vivo: trunk kidney, spleen, liver, hepatopancreasgrass carp reovirus (GCRV)
[67,68,83]
Green chromide
(Etroplus suratensis)
In vivo: spleen, kidney, liver, intestine, gills, heartpoly(I:C)[84]
Japanese flounder (Olive flounder)
(Paralichthys olivaceus)
In vitro: peripheral blood leukocytes, kidney leukocytespoly(I:C), LPS
[85]
In vivo: kidneyviral hemorrhagic septicemia virus (VHSV)[85]
In vitro: HINEA cell linepoly(I:C)[86]
Mandrinfish
(Siniperca chuatsi)
In vivo: spleen, gills, head kidneypoly(I:C), LPS[87]
Orange-spotted groupers
(Epinephelus coioides)
In vivo: spleenSingapore grouper iridovirus (SGIV), poly(I:C)[88]
Rainbow trout
(Oncorhynchus mykiss)
In vitro: RTG-2 cell line, RTS-11 cell lines poly(I:C), recombinant trout IFN2 protein[78]
In vivo: head kidneyviral hemorrhagic septicemia virus (VHSV)[78]
Sea perch
(Lateolabrax japonicas)
In vitro: LJB cell line, LJF cell linePoly(I:C), nervous necrosis virus (NNV)[89]
In vivo: Spleen, kidney, eye, thymus, brain, intestine, muscle, gill, liver, heartnervous necrosis virus (NNV)
Large yellow croaker
(Pseudosciaena crocea)
In vivo: peripheral blood, liver, spleen and head kidneypoly(I:C)[90,91]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linespring viremia of carp virus (SVCV), nervous necrosis virus (NNV), snakehead fish vesiculovirus (SHVV), Edwardsiella tarda[71,72,92]
lgp2
Asian seabass
(Lates calcarifer)
In vitro: SISK cell linepoly(I:C), LPS[93]
In vivo: liver, spleen, kidney, gill, heart, intestinepoly(I:C), Vibrio alginolyticus, Staphylococcus aureus[93]
Atlantic salmon
(Salmo salar)
In vitro: TO cell lineinfectious pancreatic necrosis virus (IPNV), infectious salmon anaemia virus (ISAV), salmonid alphavirus (SAV)[60,78]
Atlantic cod
(Gadus morhua)
In vivo: head kidneyinfectious pancreatic necrosis virus (IPNV)[94]
Black carp
(Mylopharyngodon piceus)
In vitro: MPF cell linespring viremia of carp virus (SVCV), grass carp reovirus (GCRV), poly(I:C) [95]
In vivo: liver, spleen, kidney, intestine, heart, muscle, skinspring viremia of carp virus (SVCV), grass carp reovirus (GCRV) [95]
Common carp
(Cyprinus carpio)
In vivo: muscle, spleen, gill, brain, skin, heart, intestine, liver, head kidneykoi herpes virus (KHV)[96]
Channel catfish
(Ictalurus punctatus)
In vitro: CCO cell linechannel catfish virus (CCV)[63]
In vivo: liverEdwardsiella ictaluri[63]
Fathead minnow (Pimephales promelas)In vitro: EPC cell linekoi herpes virus, (KHV), poly(I:C)[96]
Gibel carp
(Carassius gibelio)
In vivo: head kidney, spleen, liverCarassius auratus herpesvirus (CaHV)[65]
Grass carp
(Ctenopharyngodon idella)
In vitro: primary trunk kidney cellsgrass carp reovirus (GCRV), poly(I:C), LPS, PGN[67]
In vivo: trunk kidney, spleen, liver, hepatopancreasgrass carp reovirus (GCRV)[58,67,68]
Indian major carp
(Labeo rohita)
In vitro: LRG cell linepoly(I:C), iE-DAP, MDP[97]
In vivo: liver, spleen, kidney, blood, gillpoly (I:C), Aeromonas hydrophila, Bacillus subtilis[97]
Japanese flounder (Olive flounder)
(Paralichthys olivaceus)
In vitro: leukocytes isolated from kidneypoly(I:C), LPS[57]
In vivo: kidneyviral hemorrhagic septicemia virus (VHSV)[57]
In vitro: HINEA cell linepoly(I:C)[86]
Large yellow croaker
(Pseudosciaena crocea)
In vivo: peripheral blood, liver, spleen, head kidneypoly(I:C)[91]
Mandrinfish
(Siniperca chuatsi)
In vivo: spleen, gills, head kidneypoly(I:C), LPS[87]
Miiuy croaker
(Miichthys miiuy)
In vitro: macrophages
In vivo: liver, spleen, kidney
poly(I:C)[98]
Orange-spotted groupers
(Epinephelus coioides)
In vivo: spleenSingapore grouper iridovirus (SGIV), poly(I:C)[99]
Rainbow trout
(Oncorhynchus mykiss)
In vitro: RTG-2 cell line, RTS-11 cell lines poly(I:C), recombinant trout IFN2 protein[78,100]
In vivo: head kidneyviral hemorrhagic septicemia virus (VHSV)[78]
Sea perch
(Lateolabrax japonicas)
In vitro: LJF cell line, LJH cell line, LJB cell linepoly(I:C), nervous necrosis virus NNV[101]
In vivo: liver, heart, intestines, gill, spleen, muscle, brain, kidney, thymus, eyenervous necrosis virus (NNV)[101]
Zebrafish
(Danio rerio)
In vitro: ZF4 and ZFL cell linespoly(I:C), nervous necrosis virus (NNV), snakehead fish vesiculovirus (SHVV)[71,72,102]
In vivo: gill, liver, spleen, head kidney, and body kidneyspring viremia of carp virus (SVCV)[102]
ddx1
Common carp
(Cyprinus carpio)
In vivo: head kidneyspring viremia of carp virus (SVCV)[103]
Zebrafish
(Danio rerio)
In vivo: kidney, spleenspring viremia of carp virus (SVCV), chum salmon reovirus (CSV)[103]
ddx3
Orange-spotted grouper (Epinephelus coioides)In vitro: GS cell line poly(I:C), red-spotted grouper nervous necrosis virus RGNNV)[104]
Rainbow trout
(Oncorhynchus mykiss)
In vitro: RTG-2 cell linepoly(I:C)[100]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linechum salmon reovirus (CSV)[103]
dhx9
Common carp
(Cyprinus carpio)
In vivo: head kidneyspring viremia of carp virus (SVCV)[103]
Japanese medeka
(Oryzias latipes)
In vivo: spleen, kidney, intestineCpG mixture, FKC of Edwardsiella tarda[105]
Rainbow trout
(Oncorhynchus mykiss)
In vitro: RTG-2 cell linepoly(I:C)[100]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linechum salmon reovirus (CSV)[103]
In vivo: spleenchum salmon reovirus (CSV)[103]
ddx21
Common carp
(Cyprinus carpio)
In vivo: head kidneyspring viremia of carp virus (SVCV)[103]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linechum salmon reovirus (CSV)[103]
dhx36
Japanese medeka
(Oryzias latipes)
In vivo: spleen, kidney, intestineCpG-mixture, FKC of Edwardsiella tarda[105]
Zebrafish
(Danio rerio)
In vitro: ZF4 cell linechum salmon reovirus (CSV)[103]
In vivo: kidneyspring viremia of carp virus (SVCV)
ddx41
Japanese flounder (Olive flounder)
(Paralichthys olivaceus)
In vitro: adherent (monocyte-like) and non-adherent (lymphocyte- enriched) cellsranavirus[106]
In vivo: spleen, kidney, liver, heart, gilllymphocystis disease virus (LCDV)[106]
Orange-spotted grouper
(Epinephelus coioides)
In vitro: GS cell lineSingapore grouper iridovirus (SGIV), red-spotted grouper nervous necrosis virus (RGNNV)[107]
poly (I:C), polyriboinosinic:polyribocytidylic acid; LPS, lipopolysaccharide; PGN, peptidoglycan; iE-DEP, γ-D-glutamyl-meso-diaminopimelic acid; MDP, muramyl dipeptide; CpG mixture, unmethylated CpG DNA motif; FKC of Edwardsiella tarda, formalin-killed cells of Edwardsiella tarda.
Table 2. Function of cGAS against pathogenic virus, bacteria and parasite in fish and mammals.
Table 2. Function of cGAS against pathogenic virus, bacteria and parasite in fish and mammals.
PathogensSpeciesFunction/MechanismReferences
DNA viruses
African swine fever virus (ASFV)Pig (Wild boar)
(Sus scrofa domesticus)
ASFV occurs in attenuated and virulent forms. When avian alveolar macrophages are infected with attenuated ASFV, cGAS senses viral DNA and induces IFNβ. On the other hand, the virulent strain of ASFV strongly inhibits IFNβ via the cGAS-STING pathway by suppressing the downstream cascade of cGAS.[166]
Herpes simplex virus 1 (HSV-1)Mouse
(Mus musculus)
Knockdown of cGAS by shRNA in murine fibrosarcoma cell line L929 strongly inhibits IRF3 dimerization induced by HSV-1 infection.[167]
Human
(Homo sapiens)
HSV-1 tegument protein UL41 prevents cGAS DNA sensing by degrading cGAS mRNA.[181]
HSV-1 segment protein VP22 binds to cGAS protein and directly acts on the cGAS protein to degrade it.[168]
Zebrafish
(Danio rerio)
The knockdown of cGAS did not cause an obvious effect on the induction of IFN-φ1, ISG15, and viperin in zebrafish infected with HSV-1.[178]
Vaccinia virus Ankara (MVA)Human,
Mouse
Infection of human and mouse dendritic cells (DCs) with heat- or UV-inactivated MVA has been shown to induce higher levels of IFNs through the cGAS-STING pathway other than wild type-MVA.[182]
MVA DNAs in the cytosol are sensed by cGAS, what leads to activation of STING and downstream transcription factors, IRF3 and IRF7, resulting in the activation of type I IFN gene expression.[182]
Chronic hepatitis B virus (HBV)Human,
Mouse
HBV rcDNA (rcDNA; a precursor of ccc) is sensed by cGAS in the cytoplasm of hepatocytes, but may form a viral capsid that covers the DNA and escapes from cGAS sensing. HBV cccDNA was increased in cGAS knockout cells and decreased in cGAS overexpressing cells.[169]
Constitutive low expression of cGAS-STING in the liver may explain liver-specific HBV infection and a weak capacity of hepatocyte cells to clear HBV infection.[183]
Ectromelia virus (ECTV)MouseCells originated from bone marrow are the main type I IFN producers, required for ISG expression.[184]
In addition to TLR9, type I IFN production stimulated by the cGAS-STING pathway is also important for survival of mice after ECTV infection.[185]
Human cytomegalovirus (HCMV)HumancGAS recognizes HCMV DNA and induces type I IFN in human monocyte-derived plasmacytoid dendritic cells and macrophages.[186]
RNA viruses (ssRNA)
Human immunodeficiency virus (HIV)Human,
Mouse
cGAS senses reverse transcribed HIV DNA in the cytoplasm and induces cGAMP-STING-dependent IFNβ production. Knockout or knockdown of cGAS in mouse or human cell lines blocks cytokine induction by HIV, MLV and SIV.[187]
Murine leukemia virus (MLV)
Simian immunodeficiency virus (SIV)
Dengue virus (DENV)HumanDENV infects cells and localizes to endoplasmic reticulum (ER) and mitochondrial membrane. Mitochondria are disrupted by infection stress, and cGAS senses mtDNA leaked into the cytoplasm.[170]
Intracellular bacteria
Mycobacterium tuberculosisHuman,
mouse
The cGAS-STING pathway appears to activate dendritic cells by sensing Mycobacterium tuberculosis (Mtb) DNA in the cytoplasm, but does not contribute to host protection in vivo (lung cells).[171]
cGAS induces production of type I IFN and promotes early regulation of intracellular replication by inducing autophagy.[172]
Sensing Mtb DNA via the cGAS-STING pathway induces type I IFN and autophagy.[173]
Lysteria monocytogenesHumanCell infected with L. monocytogenes DNA is carried to the extracellular vesicles (EV) of infected cells and delivered to bystander cells to stimulate the cGAS-STING pathway.[174]
Edwardsiella tarda (E. piscicida)Japanese medaka
(Oryzias latipes)
E. tarda infection in medaka in vivo upregulated cgas gene expression.[105]
Zebrafish
(Danio rerio)
DrcGASa contributed to IgZ/IgZ2 induction in response to E tarda infect/ion by upregulating ifnφ1 expression in zebrafish gill γδ T cells.[160]
Extracellular bacteria
Streptococcus pneumoniaeHuman,
Mouse
The type I IFN response induced by mouse S. pneumoniae is highly dependent on the cGAS-STING pathway.[175]
Parasites
Malaria, Plasmodium falciparumHumancGAS is the cytosolic sensor of P. falciparum DNA and is required for the induction of IFNβ by malaria hemozoin (Hz) as carrier of P. falciparum gDNA.[176]
Leishmania donovaniHumancGAS dependent targeting of L. donovani DNA induces IFN-β over-production that contributes to antimony resistance in L. donovani infection.[177]
ASFV, African swine fever virus; cGAS, cyclic GMP-AMP synthase; IFN, interferon; STING, stimulator of interferon genes; shRNA, short hairpin RNA; HSV-1, herpes simplex virus 1; ISG15, interferon-stimulated gene 15; DCs, dendritic cells; rcDNA, relaxed circular DNA; cccDNA, covalently closed circular DNA; MVA, modified vaccinia virus Ankara; HBV, hepatitis B virus; TLR9, toll-like receptor 9; ECTV, ectromelia virus; HCMV, human cytomegalovirus; HIV, human immunodeficiency virus; MLV, murine leukemia virus; SIV, simian immunodeficiency virus; DENV, dengue virus; Mtb, Mycobacterium tuberculosis; EV, extracellular vesicles; IgZ, immunoglobulin Z.
Table 3. Features and functions of PKR and PKZ in fish.
Table 3. Features and functions of PKR and PKZ in fish.
Fish SpeciesSuperorder/OrderFeatures/FunctionReferences
PKR
Goldfish
(Carassius auratus)
Ostariophysi/CypriniformesTandem arrangement of PKR and PKZ genes
Induction of PKR mRNA by IFN-stimulation
Phosphorylation of eIF2α
Inhibition of translation
Interaction with poly(I:C)
[207]
Grass carp
(Ctenopharyngodon idella)
Ostariophysi/CypriniformesInduction of PKR mRNA by GCHV injection
Inhibition of translation
[211]
Japanese flounder (Olive flounder)
(Paralichthys olivaceus)
Acanthopterygii/PleuronectiformesInduction of PKR mRNA by SMRV injection
Inhibition of translation
Phosphorylation of eIF2α in response to SMRV infection
[200]
Japanese pufferfish (torafugu)
(Takifugu rubripes)
Acanthopterygii/TetraodontiformesTandem duplication of two PKR genes; Induction of PKR1 mRNA by poly(I:C) stimulation
Inhibition of translation and NF-κB activation by PKR1 and PKR2
[199,212]
Rock bream (Barred knifejaw)
(Oplegnathus fasciatus)
Acanthopterygii/PerciformesInduction of PKR mRNA by poly(I:C)[213]
Spotted green pufferfish
(Tetraodon nigroviridis)
Acanthopterygii/TetraodontiformesTandem duplication of three PKR genes[199]
Zebrafish
(Danio rerio)
Ostariophysi/CypriniformesTandem arrangement of PKR and PKZ genes
Phosphorylation of eIF2α
[199,207,214]
PKZ
Atlantic salmon
(Salmo salar)
Protacanthopterygii/SalmoniformesInduction of PKZ mRNA by IFN-stimulation
Phosphorylation of eIF2α by PKZ-stimulated with Z-DNA in vitro
[206]
Chinese rare minnow
(Gobiocypris rarus)
Ostariophysi/CypriniformesInduction of PKZ mRNA by GCRV and Aeromonas hydrophila infection[215]
Goldfish
(Carassius auratus)
OstariophysiCypriniformesInduction of PKZ mRNA by IFN-stimulation; [202,207]
Grass carp
(Ctenopharyngodon idella)
Ostariophysi/CypriniformesPhosphorylation of eIF2α by PKZ in vitro
Inhibition of translation
[216]
Zebrafish
(Danio rerio)
Ostariophysi/CypriniformesSpecific binding of Zα domains to Z-DNA
Inhibition of translation
Phosphorylation of eIF2α by both PKR and PKZ
[199,203]
eIF2α, eukaryotic initiation factor 2 alpha; GCHV, grass carp hemorrhage virus; SMRV, Scophthalmus maximus Rhabdovirus; GCRV, grass carp reovirus.

Share and Cite

MDPI and ACS Style

Mojzesz, M.; Rakus, K.; Chadzinska, M.; Nakagami, K.; Biswas, G.; Sakai, M.; Hikima, J.-i. Cytosolic Sensors for Pathogenic Viral and Bacterial Nucleic Acids in Fish. Int. J. Mol. Sci. 2020, 21, 7289. https://doi.org/10.3390/ijms21197289

AMA Style

Mojzesz M, Rakus K, Chadzinska M, Nakagami K, Biswas G, Sakai M, Hikima J-i. Cytosolic Sensors for Pathogenic Viral and Bacterial Nucleic Acids in Fish. International Journal of Molecular Sciences. 2020; 21(19):7289. https://doi.org/10.3390/ijms21197289

Chicago/Turabian Style

Mojzesz, Miriam, Krzysztof Rakus, Magdalena Chadzinska, Kentaro Nakagami, Gouranga Biswas, Masahiro Sakai, and Jun-ichi Hikima. 2020. "Cytosolic Sensors for Pathogenic Viral and Bacterial Nucleic Acids in Fish" International Journal of Molecular Sciences 21, no. 19: 7289. https://doi.org/10.3390/ijms21197289

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