Next Article in Journal
Molecular and Serological Characterization of the SARS-CoV-2 Delta Variant in Bangladesh in 2021
Next Article in Special Issue
INMI1 Zika Virus NS4B Antagonizes the Interferon Signaling by Suppressing STAT1 Phosphorylation
Previous Article in Journal
Huntingtin-Interacting Protein 1 Promotes Vpr-Induced G2 Arrest and HIV-1 Infection in Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How Influenza A Virus NS1 Deals with the Ubiquitin System to Evade Innate Immunity

by
Laurie-Anne Lamotte
and
Lionel Tafforeau
*
Cell Biology Laboratory, Research Institute for Biosciences, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium
*
Author to whom correspondence should be addressed.
Viruses 2021, 13(11), 2309; https://doi.org/10.3390/v13112309
Submission received: 27 October 2021 / Revised: 14 November 2021 / Accepted: 18 November 2021 / Published: 19 November 2021
(This article belongs to the Special Issue Viral Evasion of Innate Immunity and Drug Development)

Abstract

:
Ubiquitination is a post-translational modification regulating critical cellular processes such as protein degradation, trafficking and signaling pathways, including activation of the innate immune response. Therefore, viruses, and particularly influenza A virus (IAV), have evolved different mechanisms to counteract this system to perform proper infection. Among IAV proteins, the non-structural protein NS1 is shown to be one of the main virulence factors involved in these viral hijackings. NS1 is notably able to inhibit the host’s antiviral response through the perturbation of ubiquitination in different ways, as discussed in this review.

1. The Ubiquitin and Ubiquitin-like Systems

1.1. The Ubiquitin-Proteasome System

Ubiquitin is a small 8.5 kDa protein consisting of 76 amino acids, expressed in almost every cell type and largely conserved through eukaryotes. The binding of the C-terminal ubiquitin end to the ε-amino group of lysine (K) residue on a protein substrate is called ubiquitination. Ubiquitination is one of the best described post-translational modifications and it regulates, among other things, targeted protein localization, function, cellular trafficking, and degradation by the 26S proteasome [1,2]. Specific substrate ubiquitination requires a three-step enzymatic cascade involving E1, E2 and E3 enzymes. First, E1 enzyme activates the ubiquitin in an ATP-dependent way and transfers it to the ubiquitin-conjugating enzyme, E2. Finally, the ubiquitin ligase E3 catalyzes an isopeptide bond between the ubiquitin molecule and a lysine on the protein substrate [3,4,5]. After this mono-ubiquitination, the ubiquitin moiety itself can be ubiquitinated on eight amino groups: on the ε-amino groups of its seven K residues (K6, K11, K27, K29, K33, K48 and K63) and on the α-amino group of its N-terminal methionine. Various poly-ubiquitin chains can thereby be formed and are decisive for the targeted protein’s fate. The best known poly-ubiquitinations are K48-linked chains, that trigger protein degradation by the 26S proteasome, and K63-linked chains, that control endocytosis, cellular trafficking and protein activity (Figure 1) [2,6,7,8]. The 26S proteasome is a large protein complex with a barrel shape that consists of a catalytic central core (20S) and two 19S regulatory caps at both ends of the core. The 20S core is made of two inner rings of seven β subunits and two outer rings with seven α subunits. The two 19S bind to the 20S and activate it in an ATP-dependent manner, thus allowing the targeted protein to enter the cavity to be proteolyzed into small peptides [9].
Ubiquitinated proteins are subsequently recognized by other proteins or enzymes containing ubiquitin-binding domains, among which are de-ubiquitinase enzymes (DUBs). DUBs are able to hydrolyze isopeptide bonds formed between ubiquitin and the protein substrate or between ubiquitin moieties, with substrate and linkage specificity [10,11]. This allows the recycling of ubiquitin molecules in the cell and other crucial cellular processes that rely on free ubiquitins, such as innate immune response and autophagy (Figure 1) [11,12,13,14].
The number of each enzyme implied in this system reflects their specificity: to date, 9 E1s, 43 E2s, 919 E3s and 126 DUBs have been described in humans [15]. Usually, E2 enzymes define the type of linkage, while E3 ligases determine the substrate specificity. Three main families can be found within E3 enzymes, according to the way E3s trigger ubiquitin transfer from E2 enzyme to the substrate: the really interesting new genes (RING) family, the homologous to E6-associated protein C terminus (HECT) domain family and the RING-between-RING (RBR) family [6,7,16,17,18,19,20,21,22,23]. RING ligases are the most abundant E3 enzymes and act as mediators for direct ubiquitin transfer. They contain a zinc-binding RING domain or a U-box domain, comprising the same fold as RING but lacking zinc-chelating residues. RING ligases can be found in monomeric, homodimeric or heterodimeric states, or can be composed by multiple subunits, as with the cullin-RING ligases (CRLs) [24]. HECT and RBR ligases catalyze ubiquitin transfer through an internal thioester bond before transferring it to the substrate [6,12]. These two differ by their characteristic domain [24]. The whole ubiquitination system, including ubiquitin, enzymes and proteasome, is commonly referred to as the ubiquitin-proteasome system (UPS). Crucially, UPS is a key player in signal transduction pathways, including in innate immune responses. Increasing evidence depicts UPS’s role during viral infections, such as influenza A virus (IAV) infections, notably through tripartite motif assembly domain (TRIM) proteins from the RING family [25,26]. Besides the UPS, there are other systems implying ubiquitin-like and E1, E2 and E3 enzyme-like proteins, such as SUMOylation, ISGylation or NEDDylation [27].

1.2. Ubiquitin-like Systems

1.2.1. SUMOylation

As ubiquitination, SUMOylation is a post-translational modification of K residues on a protein substrate by a small ubiquitin-like modifier (SUMO) protein conjugation that can thus compete with K residues ubiquitination. This modification is, for instance, involved in transcription, mRNA synthesis and DNA replication or damage responses [28]. SUMOylation mainly occurs under cellular stress to resolve issue such as hypoxia, heat shock or genotoxic stresses [29]. As the UPS, this system also plays a role in the viral-dependent innate immunity pathway [30], notably by regulating functions of some pattern recognition receptors (PRRs) [31,32] and the stimulation of interferons (IFNs) [33,34].

1.2.2. ISGylation

ISGylation of cellular or viral proteins is also a post-translational modification, induced by type I IFNs stimulation in infected cells [35]. The interferon stimulated gene 15 protein (ISG15) is the first ubiquitin-like protein described in the literature [36,37]. This protein contains two ubiquitin-like domains separated by a linker region [38]. Some studies indicate that ISG15 can be secreted similarly to a soluble cytokine from epithelial cells and lymphocytes to stimulate inflammatory response and IFNγ (type II IFNs) release [39,40,41]. As an ISG protein, ISG15 expression is induced by innate immune responses through type I IFN stimulation [42], but also by other factors [43,44,45]. Similarly, ISG15 E1 (Ube1L/Uba7) [46], E2 (Ube2L6) [47,48] and E3 (Herc5) enzymes are induced by type I IFN expression [49,50,51], as well as its main DUB (USP18) [52]. Herc5 is associated with ribosomes, leading to random and large ISGylations during protein translation. Therefore, several substrates can be ISGylated, even though oligomeric viral structure proteins are mainly targeted by ISG15 to be inactivated [53].

1.2.3. NEDDylation

Conjugation of neural precursor cell expressed developmentally down-regulated protein 8 (NEDD8) to K residues of a protein substrate involves NEDD8-activating enzyme E1 (NAE), NEDD8-conjugating enzyme E2 (Ube2M and Ube2F) and different substrate-specific E3 enzymes (reviewed in [54]). The best characterized substrates of NEDD8 are cullin proteins, components of CRLs. Upon cullin NEDDylation, CRL undergoes conformational change which facilitates ubiquitin transfer from E2 enzyme to the substrate [55]. NEDDylation can be reversed by de-NEDDylase enzymes such as COP9 signalosome (CSN), which thus inactivates CRLs [56,57]. More recently, non-cullin targets of NEDD8 were also highlighted, including the itchy-homolog (ITCH) [58], Parkin [59] and MDM2 [60,61] E3 ligases. NEDDylation of these other substrates seems to enhance their stability [62].

2. Influenza A Virus and Ubiquitin

2.1. Influenza A Virus

Influenza A virus is a member of the Orthomyxoviridae family and consists of eight single-stranded negative sense RNA (ssRNA(−)) that encode 10 main proteins [63]. Hemagglutinin (HA) and neuraminidase (NA) are surface glycoproteins that facilitate viral entry and release, respectively. The M2 protein is an ion channel found in the virus envelope, itself surrounding a matrix formed by M1 protein oligomers. Each RNA segment is encapsidated by many nucleoprotein monomers (NP) and associated with an RNA-dependent RNA polymerase (PB1, PB2 and PA subunits), forming a viral ribonucleoprotein (vRNP) complex. The genome also encodes a nuclear export protein (NEP or NS2) and a non-structural protein (NS1) [63]. More recently, other non-structural proteins produced from alternative splicing have been identified in infected cells (PB1-F2, PA-X, PA-N40) [64]. The IAV polymerase acts in two ways during the course of infection, as it first triggers viral RNA transcription for proteins expression, and then RNA replication for viral particles production through viral positive-sense complementary RNA (cRNA) production [65,66]. Previous studies suggested that these two successive functions could be regulated by post-translational modifications of viral polymerase proteins such as PB1, PB2, PA and NP phosphorylation [67] and ubiquitination [68] or PB1 and NP SUMOylation [69,70].

2.2. Importance of the UPS in the IAV Life Cycle

As mentioned above, UPS has a crucial impact on the IAV life cycle. On the one hand, viral proteins ubiquitination mainly down-regulates their stability and function, but on the other hand, the lack of these modifications is clearly detrimental for proper infection [71,72,73,74,75,76,77,78]. For instance, it was shown that 26S proteasome inhibition prevents IAV entry in the cell and negatively impacts viral polymerase activity [68,74,79], while K48-linked poly-ubiquitination of viral proteins lead to their degradation [72,75]. IAV is no exception, and a lot of viruses therefore evolved to counteract and exploit the UPS. For example, some viral proteins can target cellular proteins for K48-linked degradation or use cellular DUBs to protect themselves, as well as change cellular E3s’ specificity or encode their own ubiquitin ligase enzymes [80,81,82]. Moreover, the UPS is particularly up-regulated in IAV-infected cells [83].
The IAV life cycle follows different steps in which the UPS is involved, beginning with viral entry in host cell [26]. First, the viral particle binds to sialic acids on the host cell surface by its HA protein [84]. This binding leads to the virus’ internalization in endosomes [63], and the viral particle is acidified by M2 ion channel opening [85,86]. Upon low pH, interactions between viral proteins weaken and HA undergoes a conformational change that triggers the fusion of the viral envelope with the endosome membrane [87], leading to the release of the viral genome into the cytosol by M1 destabilization. At these steps, ITCH E3 ligase mediates ubiquitination and degradation of M1, facilitating vRNPs’ release from endosomes [77], and the cullin 3 (Cul3) E3 promotes endosome maturation and thus uncoating process [88,89].
Following this uncoating, vRNPs are then imported into the nucleus through importin α/β and are processed by the viral polymerase and host factors for transcription of viral mRNA and replication [90,91,92]. In the nucleus, the interaction between NP and RNA seems to be regulated by K184 residue mono-ubiquitination of NP. This modification is crucial for viral replication and is reversed by the cellular DUB USP11 [71,73]. NP can also be poly-ubiquitinated on other K residues by other E3 ligases without targeting it for degradation. In fact, all viral polymerase proteins (PB1, PB2, PA and NP) are ubiquitinated and these modifications lead to enhanced polymerase activity and accumulation of viral RNA, cRNA and mRNA in the infected cell [68]. Besides ubiquitination, NP can also be SUMOylated at K7. This modification plays a crucial role in NP intracellular trafficking and therefore ensures its early nuclear retention for mature vRNPs’ assembly. Notably, this SUMOylation is counteracted at the late stage of infection by host caspase-dependent cleavage [70].
Meanwhile, viral proteins are produced in the cytoplasm and on the membrane of the endoplasmic reticulum, and are afterwards assembled into new virus particles with the newly synthetized vRNPs and the host plasma membrane-derived envelope, leading to the budding of new virions from the infected cell [63]. Aminoacyl-tRNA synthase complex-interacting multifunctional protein 2 (AIMP2) is a cellular protein that promotes vRNPs’ nuclear export and replication at the late steps of infection. NEP protects this protein from K48-linked poly-ubiquitination by E3 ligases such as Parkin [93,94]. In turn, AIMP2 inhibits K242 ubiquitination of M1 by ITCH and its subsequent degradation by the 26S proteasome [95]. At the late stages of infection, this K242 residue can therefore be SUMOylated, a crucial modification for vRNPs’ nuclear export [96].

3. IFNs and Cytokines Activation: The Innate Immune Response during IAV Infection

3.1. IFNs Activation

In an infected cell, foreign and potentially pathogenic materials, namely pathogen-associated molecular patterns (PAMPs), are recognized by pattern recognition receptors (PRRs) localized on the cell and/or endosome membranes or in the cytoplasm. This recognition leads to type I (IFNα et IFNβ) and type III (IFNλ) IFNs as well as pro-inflammatory cytokines production through the activation of signaling pathways [97]. Subsequent paracrine and autocrine signals then establish an antiviral state in the infected cell and in surrounding cells through ISGs expression. Post-translational modifications such as ubiquitination and phosphorylation of several cellular factors regulate this signalization pathway [80,98]. This regulation allows a duration and intensity balance in innate immune response [99]. In fact, excessive type I IFNs production leads to tissue injury and apoptosis induction [100,101].
Among innate immune response regulators, TRIM E3 ligases are key factors, leading to IRF3 and NF-κB activation [102]. A lot of TRIM proteins are known to inhibit IAV through different mechanisms [71,72,103,104,105], and the best example described so far is TRIM25, as an IAV antagonist and innate immunity pathway activator (Figure 2) [106,107]. To ensure an immunity balance, TRIM25 itself can either be K48-linked poly-ubiquitinated to be discarded or de-ubiquitinated by ubiquitin specific protease 15 (USP15) to be stabilized [108]. TRIM proteins possess a conserved architecture consisting of a N-terminal RING domain, followed by one or two B-box domain(s), and a coiled-coil domain (CCD) involved in TRIM dimerization [109,110,111] and in interactions with several proteins [112,113,114,115,116,117]. TRIMs’ C-terminal region is characterized by non-catalytic domains responsible for substrate or other proteins recognition and for subcellular localization [116,118]. Notably, this region possesses a PRY-SPRY domain in half of the TRIM proteins [118].
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are cytoplasmic PRRs consisting of three described members: RIG-I, melanoma differentiation-associated protein 5 (MDA5) and LGP2. The first and best characterized RLR is RIG-I, which is particularly active during IAV infection and therefore the key sensor in these infected cells [119,120,121]. RIG-I possesses two N-terminally located caspase activation and recruitment domains (CARDs), followed by a central DExD/H-box helicase-like domain with ATPase and translocase activities, a linker (Br) and a C-terminal regulatory/repressor domain (CTR/RD) in its C-terminal region (Figure 3). RIG-I principally detects short 5′-triphosphorylated double-stranded (ds)RNAs (5′-ppp dsRNAs) such as those produced in 5′ and 3′ UTR ends of IAV genome that are hybridized in a panhandle structure [97,121,122,123,124,125]. Therefore, RIG-I senses IAV genomic RNAs associated with NP when they are imported into the nucleus or when they are incorporated into new virion particles at late stages of infection [125,126,127]. The Toll-like receptor 3 (TLR3), a membrane PRR, seems to be also involved in this IAV sensing [128].
In the cytosol of non-infected cells, RIG-I is in an inactive state where its helicase domain binds its CARDs, hiding them from interactions and modifications [125,129,130]. Upon the sensing of a 5′-ppp dsRNA by CTR/RD, RIG-I undergoes conformational changes mediated by ATP hydrolysis [131,132,133]. CARDs become exposed and RIG-I is K63-linked poly-ubiquitinated at K172 residue by TRIM25 [106] and at K849 and K851 residues by the E3 Riplet [134,135,136,137,138], respectively, even though recent studies suggest that RIG-I poly-ubiquitination mainly relies on Riplet activity (Figure 3) [138]. Cadena et al. showed that Riplet plays both ubiquitin-dependent and independent roles in RIG-I activation, depending on dsRNAs’ length. It mediates RIG-I poly-ubiquitination upon the recognition of short dsRNAs and induces RIG-I aggregate formation on longer dsRNAs, therefore amplifying RIG-I signaling [138].
TRIM25 catalytic activity requires its own RING domain dimerization [139], thus allowing TRIM25 PRY-SPRY domain to recognize the first RIG-I CARD and mediate the poly-ubiquitination at K172 on the second one (Figure 3) [106,140]. With less efficiency, RIG-I could also be activated by free K63 chains of ubiquitins synthetized by TRIM25 [14,141], and some studies suggest that these chains coil around CARDs and are then used as a scaffold for the next step [14,141,142].
RIG-I poly-ubiquitination leads to its tetramerization and TRIM25 release [14,133,143,144]. RIG-I then accumulates around mitochondria, where it binds to the mitochondrial antiviral signaling protein (MAVS) by its CARDs, leading to MAVS multimerization in filamentous structures [76,145,146,147,148]. MAVS mitochondrial localization is a determinant for innate immune response [149], and the MAVS C-terminal transmembrane domain allows its anchor into the mitochondrial membrane, where it aggregates upon RIG-I activation [146,150]. Through signaling cascades notably involving TAK1 and TRAFs proteins, MAVS multimerization leads to TBK1 and IKK kinases’ recruitment. In turn, TBK1 phosphorylates and activates IRF3 and IRF7 transcription factors that direct type I and type III IFNs expression, while IKK kinases mediate phosphorylation of NF-κB inhibitor IκB [14,97,108,145,146,151,152,153,154,155,156]. Once phosphorylated, IκB is targeted by the SCFβ-TrCP E3 ligase complex for proteasomal degradation, therefore activating NF-κB that translocates into the nucleus to direct pro-inflammatory cytokines as well as IFNβ expression [157,158,159]. Notably, it was shown that TBK1 and IKK kinases’ recruitment relies on K63 poly-ubiquitination mechanisms (Figure 2) [160,161].

3.2. ISGs Activation

Produced IFNs are secreted in an autocrine and a paracrine way from the infected cell and bind to IFNAR1/2 (type I IFNs) or IFNLR1 and IL10RB (type III IFNs) membrane receptors. Type I and type III IFNs are induced by the same PRRs and trigger similar downstream signaling, but recent studies suggest that type III IFNs’ response is the primary defense in epithelial cells, while type I IFNs’ machinery is more systemic and forms the second line of defense in case of broader infection [162]. Moreover, type III IFNs seem to trigger less inflammation than type I IFNs, thereby protecting epithelial tissue from immunopathology [163]. The binding of IFNs to their receptors activates signaling cascades that establish an antiviral state through phosphorylation of signal transducer and activator of transcription (STAT) 1 and STAT2 transcription factors by Janus kinase 1 (JAK1) and tyrosine kinase 2 (TYK2) [164,165]. These activated factors then associate with IRF9 to form an ISG factor 3 (ISGF3) complex that translocates into the nucleus where it binds to IFN-stimulated response element (ISRE) sequences in ISG promoters to induce their transcription (Figure 2) [164,166].
Many ISGs particularly inhibit IAV replication, including myxovirus resistance (MxA), IFN-induced transmembrane (IFITM), 2′–5′ oligoadenylate synthetase (OAS) and ribonuclease L (RNAseL) from OAS-RNase L pathway, protein kinase R (PKR), and ISG20 [97,167,168,169]. Human cytosolic protein MxA is a guanylate-binding protein (GBP) that inhibits IAV by targeting viral transcription and by binding to the NP protein [170,171,172]. The IFITM protein family contains three members (IFITM1, IFITM2 and IFITM3) that have antiviral activities by blocking the fusion of the viral particle with cellular host membrane at the uncoating step [169,173,174,175]. IFITM3 is notably down-regulated by its ubiquitination triggered by NEDD4 E3 ligase. In the absence of this E3 enzyme, cells are more resistant to IAV infection [76,77]. dsRNAs activate OAS and lead to the production of poly(A) chains that in turn bind to and activate RNAseL [176]. This ribonuclease then cleaves viral RNA and therefore inhibits viral replication in infected cells [177]. Moreover, products from viral RNA degradation can activate RIG-I, therefore amplifying IFN response [124,178]. The presence of dsRNAs in infected cells also activates the multifunctional PKR protein, which, for instance, phosphorylates the α subunit of the eukaryotic initiation factor 2 (eIF2α), subsequently inhibiting cellular but also viral proteins translation [179]. PKR also phosphorylates the NF-κB inhibitor IκB, and activates IRF3, leading to these transcription factors’ activation [180,181]. In addition, PKR stabilizes type I IFNs mRNA, thus enhancing IFNs production [182]. Finally, ISG20 is a 3′—5′ exonuclease known to inhibit several ssRNA viruses. During IAV infection, ISG20 binds to NP to block virus replication and transcription [168].

3.3. Inflammasome Complexes

In response to PAMPs, RIG-I also activates inflammasomes, multiproteic complexes consisting of nucleotide oligomerization domain (NOD)-like receptor family member LRR- and pyrin domain containing-3 (NLRP3), apoptosis-associated speck-like containing a caspase-recruitment domain (ASC) and pro-caspase-1 proteins [183,184]. Inflammasomes are involved in the defense against several viruses of IAV [183,185,186,187]. Indeed, NLRP3 is notably expressed in human bronchial epithelial cells [188]. Inflammasomes’ activation also relies on PAMP detection by NLRP3 and on PKR activity [189], as well as on protein flux through the M2 ion channel in the trans-Golgi network [190] and on IAV PB1-F2’s presence in the cell [191]. Inflammasomes stimulate caspase-1 activation, which in turn cleaves inactive pro-interleukin (IL)-1β and pro-IL-18 into mature IL-1β and IL-18 forms, respectively, which are then secreted to stimulate inflammatory response (Figure 2) [97,183,192].

3.4. Autophagy and Apoptosis

Autophagy, a self-eating mechanism, is involved in several cellular processes such as cell death or protein and organelle elimination through lysosomes, but it also plays a crucial part in innate immune response through TLRs activation, in inflammatory disorders, and in tumor suppression [193,194,195]. Depending on the host and on the virus strain, autophagy can further be used as a replication and pathogenesis enhancer [72,194,195,196,197,198] or is subverted by viral proteins such as IAV M2 to enhance virion stability [199,200]. Another cellular defense pathway triggered in response to intracellular pathogens is apoptosis, which is also involved in cell growth control [201]. This programmed cell death is notably hijacked by IAV to facilitate its replication [202]. Autophagy and apoptosis are both regulated by PKR signaling pathway [203,204].

4. The Innate Immune Battle between UPS and IAV

4.1. Antiviral Roles of UPS Factors

In addition to TRIM25 and Riplet, other UPS and UPS-like factors are involved in the antiviral pathway during IAV infection, either by directly and indirectly targeting viral proteins or by amplifying immune response.
TRIM and DUB proteins are notable key players in antiviral responses against IAV infection. Among TRIMs, TRIM5, TRIM14, TRIM22, TRIM32 and TRIM41 directly target viral proteins to restrain replication [105,116]. TRIM14 and TRIM41 bind to NP, leading to its ubiquitination and degradation, thus inhibiting vRNPs’ formation and viral RNA replication [205,206]. TRIM22 also interacts with NP to promote its K48-linked poly-ubiquitination and subsequent proteasomal degradation [71]. Nonetheless, it seems that some H1N1 strains evolved to evade TRIM22-mediated antiviral activity [207]. PB1 is targeted by TRIM32, which leads to a consecutive viral polymerase activity decrease [72]. Moreover, TRIM32 ligase activity stimulates the unc-51-like autophagy activating kinase 1 (ULK1), which controls autophagy induction through the formation of Beclin 1 complexes [208]. Several other TRIMs are implied in autophagy-associated antiviral mechanisms as well as in IAV-induced autophagy (reviewed in [105]). Another TRIM, TRIM44, stabilizes MAVS by inhibiting its proteasomal degradation, therefore stabilizing and enhancing the IFN response [209], while the TRIM28 protein is involved in pro-inflammatory cytokines production [210].
Among DUBs regulating the RIG-I dependent innate immune response, USP21 is able to remove RIG-I K63-linked poly-ubiquitin chains [211], USP4 hydrolyses RIG-I K48-linked chains [212], and OTU domain-containing protein 1 (OTUD1) stabilizes a not yet identified E3 ligase, thus facilitating MAVS degradation [213]. Interestingly, Otubain-1 DUB (OTUB1) was recently described as an antiviral key regulator in IAV-infected cells. Upon infection, OTUB1 is induced by type I IFNs and is associated with RIG-I at the mitochondrial membranes, where it activates RIG-I by a double mechanism [214]. First, OTUB1 specifically hydrolyses RIG-I K48-linked chains, and then it inhibits these RIG-I K48-linked poly-ubiquitinations through the formation of an E2-repressive complex [214,215,216].
Inflammasome complexes are also regulated by E3 and DUB enzymes. Indeed, the linear ubiquitin chain assembly complex (LUBAC) and TRIM33 E3 ligases promote inflammasomes’ formation [217,218], while breast cancer 1 (BRCA1)/breast cancer 2 (BRCA2)-containing complex subunit 3 (BRCC3) de-ubiquitinates NLRP3 [219].
Other mechanisms, such as SUMOylation, ISGylation and NEDDylation, also regulate several viruses, such as IAV [220,221,222]. SUMOylated TRIM28 acts with other proteins to inhibit endogenous retroviral (ERV) elements’ expression [223,224,225,226,227,228]. Cellular ERVs can be recognized by RIG-I and/or MDA5, therefore leading to aberrant IFN stimulation [229,230,231,232,233,234]. In IAV-infected cells, Schmidt et al. observed a diminution of SUMOylated TRIM28 levels, thus preparing cells for antiviral state establishment [220]. It was also shown that ISG15 conjugation, even at a low level, has a negative impact on the ability of the virus to replicate [235,236]. In addition, MDM2 E3 ligase seems to enhance M1 and PB2 NEDDylation, leading to the destabilization of these viral proteins and the inhibition of IAV replication in vitro and in vivo [222,237]. On the contrary, Sun et al. observed that IAV infection leads to the activation of NEDDylation pathway, which promotes virus growth but also pathogenicity. Indeed, cullin 1 NEDDylation can activate the NF-κB pathway and lead to the over-expression of pro-inflammatory cytokines that enhance viral pathogenicity [238]. NEDDylation modifications therefore seem to play a balancing role in the regulation of IAV replication.
Finally, the UPS is also indirectly involved in defense against IAV, through enhancement of proteasomal-mediated degradation of viral proteins. For example, Cyclophilin A accelerates the degradation of M1 [239], while the long isoform of zinc finger antiviral protein (ZAP-L) associates with PA and PB2 to promote their presumably K48-linked poly-ubiquitination, although ZAP-L activity is counteracted by PB1 protein [75].

4.2. IAV Hijacks UPS to Evade Innate Immune Response

As a response to antiviral state triggered by infected cells, many viruses evolved to counteract RIG-I-mediated innate immune response [240,241]. For instance, IAV PB1-F2 and PB2 proteins interact with MAVS, NP prevents PKR-mediated IRF3 phosphorylation by binding to HSP40, and HA expression leads to IFNAR1 ubiquitination and degradation, thus inhibiting type I IFN production and response pathways [242,243,244,245]. Accordingly, IAV can use the UPS not only to facilitate its replication, but also to evade the host’s immune system.
Hence, different examples of UPS hijacking by IAV are described in the literature. Among others, the ubiquitination of M2 on its K78 residue regulates IAV replication by coordinating apoptosis and autophagy, two mechanisms used by the virus to spread and to control infection-mediated cellular death, even though the involved E3 ligase is still unknown [246]. Regardless of these significant examples, the main IAV virulence protein remains NS1, acting by direct interactions with proteins such as TRIM25 or indirect interactions by regulating host gene expression [107,247,248,249,250,251,252,253,254,255].

5. Mechanisms Developed by NS1 to Inhibit the Innate Immune Response

5.1. IAV NS1 Protein

The RNA segment 8 of the IAV genome encodes for a mRNA, from which is synthetized the non-structural protein NS1. An alternative splicing of this mRNA results in the production of the NEP protein, which represents 10–15% of the segment 8 transcripts [256,257]. A third transcript with a punctual mutation and a subsequently alternative splicing is also found in some viral strains, therefore expressing a NS3 protein with the same terminal ends as NS1 [258]. NS1 is generally a 230 amino acid (AAs) protein, even though its length may vary due to mutations depleting the stop codon at position 231 or creating a premature stop codon. For example, human IAV NS1 displayed 237 AAs from the late 1940s to the middle of the 1980s, while NS1 from the 2009 pandemic H1N1 strain only harbors 219 AAs [252,259,260].
NS1 possesses an RNA-binding domain (RBD) in N-terminus (AAs 1–73), separated by a flexible linker region from an effector domain (ED) (AAs 85–207), which is followed by a C-terminal tail. The RBD forms three α-helices that are crucial for NS1 dimerization and for interaction with dsRNA [261,262,263]. Amino acids at position 38 and 41 are important for this binding function [263,264], while dimerization allows NS1 homodimer and then oligomer formation in infected cells [265,266,267,268]. The RBD also displays a nuclear localization signal (NLS) from AA 35 to 41 [269,270]. The ED contains a β-sheet structure and a long central helix as well as a nuclear export signal (NES) (AAs 137–146). Together with NLS, this signal allows NS1 to shuttle from the nucleus to the cytoplasm during infection [271]. Interestingly, several cellular proteins have been shown to interact with NS1 ED [272]. Similarly to the linker region (10 to 15 AAs long), the C-terminal tail varies in length (11 to 33 AAs long), and it may contain, depending on the viral strain, a PDZ-binding motif involved in viral pathogenesis [273,274]. Notably, H1N1 1918 NS1 contains a SUMOylation consensus site embedded in the PDZ-binding domain [275]. Most of the IAV strains also display a NLS in the tail domain (AAs 216–221) (Figure 3) [269].

5.2. NS1 against Host Antiviral Response

NS1 is known to inhibit the antiviral response in the host cell by means of a wide range of mechanisms, including through protein interactions, host shutoff and ubiquitination perturbations (reviewed in [252,272,276]), turning NS1 into a key protein for viral replication cycle. Some studies therefore suggest that IFNβ-competent cells infected with IAV lacking NS1 considerably impair viral replication [277,278], as well as cells infected with IAV expressing a truncated form, low level or functional mutations of NS1 [279,280,281,282]. Non-canonical antiviral pathways seem to be also targeted by NS1. Indeed, Schmidt et al. showed that NS1 can bind to ERV dsRNA and then inhibits the subsequent innate immune response [220].

5.2.1. IFNs, Cytokines and ISGs Inhibition by NS1

During the battle between IAV infection and innate immune response, NS1 inhibits IFN activation through the interaction with proteins such as Riplet [283] and IRF3 [247]. Moreover, it was shown that the NS1 ED of some IAV strains suppresses TRAF3 K63-linked poly-ubiquitination, probably through DUBs recruitment. This leads to the disruption of the MAVS and TRAF3 complex and to the impairment of subsequent IRF3 phosphorylation [284]. NS1 also inhibits NF-κB activation by blocking the β subunit of IKK [285]. Its RBD seems to play a crucial role in cytokine production and in cytokine sensitivity [286].
In addition, NS1 was shown to regulate the expression of some JAK/STAT signaling inhibitors by binding to the DNA methyltransferase 3B (DNMT3B). Upon IAV infection, NS1 dissociates DNMT3B from gene promoters and changes its localization to the cytosol, where it is K48-linked poly-ubiquitinated and degraded, therefore leading to an enhanced expression of JAK/STAT signaling inhibitors [287]. NS1 also inhibits some specific ISGs, such as IFITM3, the expression of which is attenuated by NS1-mediated eIF4B degradation [272]. By binding viral RNA, NS1 competes with cellular RNA, binding proteins such as OAS, RNAse L, PKR and RIG-I, thus protecting them from degradation and inhibiting antiviral response (Figure 2) [251,288,289,290].
NS1 antagonizes inflammasome by a direct interaction with NLRP3, which prevents ASC ubiquitination and inflammasome activation (Figure 2) [185,291,292].
Finally, NS1 up-regulates the phosphoinositide-3-kinase (PI3K) pathway through direct interaction with the regulating subunit p85β of PI3K, causing downstream phosphorylation of the protein kinase B (Akt) [293,294,295,296]. Notably, p85β possesses a CCD to which NS1 ED binds [297,298]. This leads to an increase in the viral internalization rate, apoptosis inhibition [299], and an enhancement of IRF3 activity [300,301]. While NS1 seems to inhibit apoptosis, recent studies suggest that it also has a pro-apoptotic effect [302]. NS1 dual function in apoptosis regulation therefore may vary depending on the stage of viral infection. Indeed, induction of apoptosis seems to be essential for vRNPs’ nuclear trafficking at the beginning of the infection, while the limitation of apoptosis at later stages could prevent premature viral particles release and death of infected cells [303].

5.2.2. Host Shutoff

Another mechanism triggered by NS1 to impair the antiviral state in infected cells is host shutoff, i.e., inhibition of host genes expression. NS1 mainly inhibits cellular mRNAs processing by binding to CPSF30, a key component of pre-mRNA 3′-end formation machinery [260,279,304,305,306,307]. This interaction prevents the recognition by CPSF30 of poly(A) signals at 3′-end of mRNAs during their transcription, thereby blocking immature mRNAs’ cleavage and poly(A) tail addition [307,308]. Poly(A) tail allows mRNAs’ stabilization, nuclear export, and translation [308]. In its absence, immature mRNAs thus accumulate in the nucleus of infected cells, leading to a general host shutoff, affecting, among other things, the expression of IFNs, ISGs, and pro-inflammatory proteins [272,306,309]. In this way, NS1 also binds and inhibits PABPII, the nuclear poly(A)-binding protein II, which stimulates the synthesis of long poly(A) tails [309]. It was shown that NS1 RBD is able to bind host dsDNAs, therefore inhibiting the loading of transcriptional machinery to IAV antiviral genes [310]. Nuclear RNA export factor 1 (NFX1), ribonucleic acid export 1 (RAE1) and p15 are examples of mRNA export machinery proteins also targeted by NS1 from the H1N1 WSN strain [311]. Additionally, NS1 enhances viral mRNAs translation to the detriment of cellular mRNAs by recruiting eIF4GI to the viral mRNA 5′-UTR region [312].

5.3. Ubiquitination Perturbations by NS1

5.3.1. TRIM25

One of the main antiviral targets of NS1 is TRIM25 [107,283], therefore interfering with its E3 ligase activity and with IFN signaling pathway activation (Figure 2) [107]. The interaction between PRY-SPRY domain of TRIM25 and its own CCD is essential to mediate RIG-I ubiquitination [118], and NS1 directly competes with this binding [107,118,144]. Indeed, NS1 presumably binds to TRIM25 CCD via its functional ED amino acids 96 and 97 (Figure 3) [107]. Interestingly, this interface is the same as the one in NS1 and p85β interaction, suggesting that NS1 uses the same ED surface to recognize similar structures in various targets [118,297,298]. NS1 and TRIM25 complex formation therefore inhibits the correct juxtaposition of TRIM25 PRY-SPRY and RING domains, and thus TRIM25 multimerization and activity [107,118]. However, this binding does not impair free K63-linked poly-ubiquitin chains formation by TRIM25, neither the formation of TRIM25 and RIG-I complex in infected-cells cytoplasm [118]. On the contrary, NS1 also interacts with RIG-I and inhibits its activation by sequestrating its RNA helicase and its ligand [255,313,314]. Moreover, a recent study suggested that NS1 RBD from 1918 H1N1 strain directly binds to the RIG-I second CARD and inhibits its ubiquitination (Figure 3) [315], and that the R21Q natural mutation of some NS1 proteins intriguingly impairs this inhibition, leading to a stronger immune response [315,316]. This mutation could therefore be a specific adaptation of some strains to host species [283,315]. Indeed, it has been shown that H3N2 and H2N2 IAV strains infections lead to a high IRF3 and IFNβ activation, despite a proper interaction between NS1 and TRIM25 [317,318]. Meyerson et al. thus suggested that NS1 inhibits another antiviral function of TRIM25, independent of its ligase activity and of RIG-I. In fact, TRIM25 can bind to vRNP RNA and proteins in infected-cell nucleus, preventing viral RNA elongation [196]. Finally, NS1 is also able to suppress TRIM25 expression at a transcriptional level through up-regulation of Dot1L methyltransferase during infection [319].

5.3.2. DUBs

NS1 inhibits the RIG-I pathway by up-regulating the expression of A20 DUB (other name TNFAIP3), a negative regulator of the innate immune response [99,320,321]. A20 suppresses the antiviral state established during infection [322], and its up-regulation by NS1 seems to be proportional to strain virulence [99]. A20 is a dual-ubiquitin editing enzyme that contains a N-terminal DUB activity and seven Zn finger domains with E3 ligase activity in its C-terminal region, which regulate innate immune response [323,324].
OTUB1 is also targeted by NS1, even though the mechanism is still undefined. NS1 could trigger OTUB1 proteasomal degradation at the later stages of infection, leading to the inhibition of IRF3 and NF-κB activation, and thus preventing the antiviral response (Figure 2) [214].

5.3.3. p53 and MDM2

p53 is a well-known transcription factor and tumor suppressor that accumulates in the nucleus upon cellular stress [325,326]. It regulates several biological processes, including apoptosis and antiviral mechanisms by regulating infected-cell fate [327,328,329,330,331,332]. p53 activates, among other things, the transcription of its main negative regulator, MDM2, a RING E3 ligase that in turn binds to p53 and mediates its poly-ubiquitination and proteasomal degradation. MDM2 can also mediate p53 NEDDylation [60]. p53 stability is therefore dependent on its interaction with MDM2, as well as on the MDM2 level or cellular localization [295].
IAV was shown to regulate p53 transcriptional activity, through its stabilization by NS1 [330,333,334]. This p53 stabilization in infected cells is associated with a defect of MDM2-mediated ubiquitination of p53 [335]. Pizzorno et al. showed that NS1 is responsible for proteasomal-mediated degradation of MDM2 at early stages of infection, and that it also modifies MDM2 subcellular localization during IAV infection [336]. As with p53, MDM2 could also plays an antiviral role, independently of its ligase activity and p53 [336]. According to the ambivalent role of apoptosis during IAV infection, MDM2 could be used by NS1 at early stages of infection to promote IAV propagation [303]. However, MDM2 is involved in many other pathways linked to IAV infection, such as cell cycle control or NF-κB signalization, perhaps ensuring antiviral effect by other mechanisms [337,338].

5.3.4. SUMOylation System

Similarly to NP discussed previously, NS1 also interacts with the cellular SUMOylation system during IAV infection. Indeed, some studies suggest that most of IAV strains possess a SUMOylated NS1 protein, and that different lysine residues can be targeted, even though K131 seems to be the main NS1 SUMOylation site. These studies also highlight the crucial, but not essential, role of NS1 SUMOylation in virus replication [339,340,341]. Santos et al. notably showed that SUMOylation at K70 and K219 has no impact on NS1 stability and localization but can affect NS1 oligomerization and therefore NS1 functions. Furthermore, they showed that a modification of NS1 SUMOylation level negatively impacts the ability of NS1 to counteract IFN response [340].
Moreover, SUMOylation of NS1 was recently shown to take part in host shutoff by enhancing NS1 association with nuclear ribonucleoprotein complexes that control the activity of the RNA polymerase II [275].

5.3.5. ISG15

Interestingly, NS1 from influenza B (IBV) virus also antagonizes the RIG-I-mediated antiviral response [342]. Notably, IBV NS1 binds to the N-terminal ubiquitin-like domain and to the linker region of ISG15 [46,343,344], inhibiting its activity and its secretion [39]. Moreover, IBV NS1 binds and sequesters ISGylated proteins, such as NP, to prevent its undesirable and premature inclusion into vRNPs [235,343].

6. Conclusions

The ubiquitin-proteasome system, as well as ubiquitin-like machineries, are key cellular processes involved in various pathways, including notably innate immune response. Upon influenza A virus infection, ubiquitination-mediated RIG-I activation leads to an antiviral state establishment in infected and surrounding cells, thus inhibiting IAV replication through IFNs, ISGs and pro-inflammatory cytokines expression. Several UPS factors are described to enhance and stabilize this innate immune response and to directly target IAV proteins for their proteasomal-mediated degradation. In turn, IAV evolved to hijack the UPS and use it to enter the cell and spread, disrupting UPS activity from antiviral to proviral roles. Furthermore, the viral non-structural protein NS1, a well-known virulence factor, developed many mechanisms to antagonize innate immunity, from direct interactions with cellular proteins to the general inhibition of host gene expression. Interestingly, NS1 specifically targets UPS factors such as TRIM25, A20, OTUB1 and MDM2, SUMOylation system, as well as ISG15 in the case of influenza B virus. NS1 activity seems to vary depending on the infection timing, and further investigations are therefore needed to decipher the role of NS1 in ubiquitin perturbations during the course of IAV infection. We are currently investigating the identification of UPS factors interacting with IAV NS1 and the functional characterization of some of them.

Author Contributions

Writing—original draft preparation, L.-A.L.; writing—review and editing, L.-A.L. and L.T.; supervision, project administration and funding acquisition, L.T. All authors have read and agreed to the published version of the manuscript.

Funding

L.-A.L. is supported by a FRIA PhD fellowship. This work was supported by the Fonds de la Recherche Scientifique-FNRS under CDR grant J.0198.20 and by the Fonds pour la Recherche Médicale dans le Hainaut-FRMH.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dikic, I.; Dötsch, V. Ubiquitin Linkages Make a Difference. Nat. Struct. Mol. Biol. 2009, 16, 1209–1210. [Google Scholar] [CrossRef]
  2. Chen, Z.J.; Sun, L.J. Nonproteolytic Functions of Ubiquitin in Cell Signaling. Mol. Cell 2009, 33, 275–286. [Google Scholar] [CrossRef] [PubMed]
  3. Chau, V.; Tobias, J.W.; Bachmair, A.; Marriott, D.; Ecker, D.J.; Gonda, D.K.; Varshavsky, A. A Multiubiquitin Chain Is Confined to Specific Lysine in a Targeted Short-Lived Protein. Science 1989, 243, 1576–1583. [Google Scholar] [CrossRef] [PubMed]
  4. Ciechanover, A. The Unravelling of the Ubiquitin System. Nat. Rev. Mol. Cell Biol. 2015, 16, 322–324. [Google Scholar] [CrossRef] [PubMed]
  5. Hershko, A.; Heller, H.; Elias, S.; Ciechanover, A. Components of Ubiquitin-Protein Ligase System. Resolution, Affinity Purification, and Role in Protein Breakdown. J. Biol. Chem. 1983, 258, 8206–8214. [Google Scholar] [CrossRef]
  6. Komander, D.; Rape, M. The Ubiquitin Code. Annu. Rev. Biochem. 2012, 81, 203–229. [Google Scholar] [CrossRef] [Green Version]
  7. Kulathu, Y.; Komander, D. Atypical Ubiquitylation-the Unexplored World of Polyubiquitin beyond Lys48 and Lys63 Linkages. Nat. Rev. Mol. Cell Biol. 2012, 13, 508–523. [Google Scholar] [CrossRef] [PubMed]
  8. Tanno, H.; Komada, M. The Ubiquitin Code and Its Decoding Machinery in the Endocytic Pathway. J. Biochem. 2013, 153, 497–504. [Google Scholar] [CrossRef] [Green Version]
  9. Tanaka, K. The Proteasome: Overview of Structure and Functions. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 12–36. [Google Scholar] [CrossRef] [Green Version]
  10. Komander, D.; Clague, M.J.; Urbé, S. Breaking the Chains: Structure and Function of the Deubiquitinases. Nat. Rev. Mol. Cell Biol. 2009, 10, 550–563. [Google Scholar] [CrossRef]
  11. Clague, M.J.; Barsukov, I.; Coulson, J.M.; Liu, H.; Rigden, D.J.; Urbé, S. Deubiquitylases from Genes to Organism. Physiol. Rev. 2013, 93, 1289–1315. [Google Scholar] [CrossRef]
  12. Swatek, K.N.; Komander, D. Ubiquitin Modifications. Cell Res. 2016, 26, 399–422. [Google Scholar] [CrossRef] [Green Version]
  13. Hao, R.; Nanduri, P.; Rao, Y.; Panichelli, R.; Ito, A.; Yoshida, M.; Yao, T. Proteasomes Activate Aggresome Disassembly and Clearance by Producing Unanchored Ubiquitin Chains. Mol. Cell 2013, 51, 819–828. [Google Scholar] [CrossRef] [Green Version]
  14. Zeng, W.; Sun, L.; Jiang, X.; Chen, X.; Hou, F.; Adhikari, A.; Xu, M.; Chen, Z.J. Reconstitution of the RIG-I Pathway Reveals a Signaling Role of Unanchored Polyubiquitin Chains in Innate Immunity. Cell 2010, 141, 315–330. [Google Scholar] [CrossRef] [Green Version]
  15. Zhou, J.; Xu, Y.; Lin, S.; Guo, Y.; Deng, W.; Zhang, Y.; Guo, A.; Xue, Y. IUUCD 2.0: An Update with Rich Annotations for Ubiquitin and Ubiquitin-like Conjugations. Nucleic Acids Res. 2018, 46, D447–D453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Berndsen, C.E.; Wolberger, C. New insights into ubiquitin E3 ligase mechanism. Nat. Struct. Mol. Biol. 2014, 21, 301–307. [Google Scholar] [CrossRef] [PubMed]
  17. Zheng, N.; Shabek, N. Ubiquitin ligases: Structure, function, and regulation. Annu. Rev. Biochem. 2017, 86, 129–157. [Google Scholar] [CrossRef] [PubMed]
  18. Metzger, M.B.; Pruneda, J.N.; Klevit, R.E.; Weissman, A.M. RING-type E3 ligases: Master manipulators of E2 ubiquitin-conjugating enzymes and ubiquitination. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 47–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Sluimer, J.; Distel, B. Regulating the Human HECT E3 Ligases. Cell Mol. Life Sci. 2018, 75, 3121–3141. [Google Scholar] [CrossRef] [Green Version]
  20. Metzger, M.B.; Hristova, V.A.; Weissman, A.M. HECT and RING finger families of E3 ubiquitin ligases at a glance. J. Cell Sci. 2012, 125, 531–537. [Google Scholar] [CrossRef] [Green Version]
  21. Bernassola, F.; Karin, M.; Ciechanover, A.; Melino, G. The HECT Family of E3 Ubiquitin Ligases: Multiple Players in Cancer Development. Cancer Cell 2008, 14, 10–21. [Google Scholar] [CrossRef]
  22. Bosu, D.R.; Kipreos, E.T. Cullin-RING Ubiquitin Ligases: Global Regulation and Activation Cycles. Cell Div. 2008, 3, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hatakeyama, S. TRIM family proteins: Roles in autophagy, immunity, and carcinogenesis. Trends Biochem. Sci. 2017, 42, 297–311. [Google Scholar] [CrossRef] [PubMed]
  24. Morreale, F.E.; Walden, H. Types of Ubiquitin Ligases. Cell 2016, 165, 248.e1. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, M.; Wang, L.; Li, S. Influenza A virus-host protein interactions control viral pathogenesis. Int. J. Mol. Sci. 2017, 18, 1673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Rudnicka, A.; Yamauchi, Y. Ubiquitin in influenza virus entry and innate immunity. Viruses 2016, 8, 293. [Google Scholar] [CrossRef] [Green Version]
  27. Bedford, L.; Lowe, J.; Dick, L.R.; Mayer, R.J.; Brownell, J.E. Ubiquitin-like Protein Conjugation and the Ubiquitin-Proteasome System as Drug Targets. Nat. Rev. Drug Discov. 2011, 10, 29–46. [Google Scholar] [CrossRef] [PubMed]
  28. Zhao, X. SUMO-Mediated Regulation of Nuclear Functions and Signaling Processes. Mol. Cell 2018, 71, 409–418. [Google Scholar] [CrossRef] [Green Version]
  29. Enserink, J.M. Sumo and the Cellular Stress Response. Cell Div. 2015, 10, 4. [Google Scholar] [CrossRef] [Green Version]
  30. Everett, R.D.; Boutell, C.; Hale, B.G. Interplay between Viruses and Host Sumoylation Pathways. Nat. Rev. Microbiol. 2013, 11, 400–411. [Google Scholar] [CrossRef]
  31. Hu, M.-M.; Liao, C.-Y.; Yang, Q.; Xie, X.-Q.; Shu, H.-B. Innate Immunity to RNA Virus Is Regulated by Temporal and Reversible Sumoylation of RIG-I and MDA5. J. Exp. Med. 2017, 214, 973–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Hu, M.M.; Yang, Q.; Xie, X.Q.; Liao, C.Y.; Lin, H.; Liu, T.T.; Yin, L.; Shu, H. Sumoylation Promotes the Stability of the DNA Sensor CGAS and the Adaptor STING to Regulate the Kinetics of Response to DNA Virus. Immunity 2016, 45, 555–569. [Google Scholar] [CrossRef] [Green Version]
  33. Crowl, J.T.; Stetson, D.B. SUMO2 and SUMO3 Redundantly Prevent a Noncanonical Type I Interferon Response. Proc. Natl. Acad. Sci. USA 2018, 115, 6798–6803. [Google Scholar] [CrossRef] [Green Version]
  34. Decque, A.; Joffre, O.; Magalhaes, J.G.; Cossec, J.-C.; Blecher-Gonen, R.; Lapaquette, P.; Silvin, A.; Manel, N.; Joubert, P.-E.; Seeler, J.-S.; et al. Sumoylation Coordinates the Repression of Inflammatory and Anti-Viral Gene-Expression Programs during Innate Sensing. Nat. Immunol. 2016, 17, 140–149. [Google Scholar] [CrossRef]
  35. Zhao, C.; Denison, C.; Huibregtse, J.M.; Gygi, S.; Krug, R.M. Human ISG15 Conjugation Targets Both IFN-Induced and Constitutively Expressed Proteins Functioning in Diverse Cellular Pathways. Proc. Natl. Acad. Sci. USA 2005, 102, 10200–10205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Haas, A.L.; Ahrens, P.; Bright, P.M.; Ankel, H. Interferon Induces a 15-Kilodalton Protein Exhibiting Marked Homology to Ubiquitin. J. Biol. Chem. 1987, 262, 11315–11323. [Google Scholar] [CrossRef]
  37. Loeb, K.R.; Haas, A.L. The Interferon-Inducible 15-KDa Ubiquitin Homolog Conjugates to Intracellular Proteins. J. Biol. Chem. 1992, 267, 7806–7813. [Google Scholar] [CrossRef]
  38. Narasimhan, J.; Wang, M.; Fu, Z.; Klein, J.M.; Haas, A.L.; Kim, J.-J.P. Crystal Structure of the Interferon-Induced Ubiquitin-like Protein ISG15. J. Biol. Chem. 2005, 280, 27356–27365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Swaim, C.D.; Canadeo, L.A.; Monte, K.J.; Khanna, S.; Lenschow, D.J.; Huibregtse, J.M. Modulation of Extracellular ISG15 Signaling by Pathogens and Viral Effector Proteins. Cell Rep. 2020, 31, 107772. [Google Scholar] [CrossRef]
  40. D’Cunha, J.; Ramanujam, S.; Wagner, R.J.; Witt, P.L.; Knight, E.; Borden, E.C. In Vitro and in Vivo Secretion of Human ISG15, an IFN-Induced Immunomodulatory Cytokine. J. Immunol. 1996, 157, 4100–4108. [Google Scholar]
  41. Recht, M.; Borden, E.C.; Knight, E. A Human 15-KDa IFN-Induced Protein Induces the Secretion of IFN-Gamma. J. Immunol. 1991, 147, 2617–2623. [Google Scholar]
  42. Farrell, P.J.; Broeze, R.J.; Lengyel, P. Accumulation of an MRNA and Protein in Interferon-Treated Ehrlich Ascites Tumour Cells. Nature 1979, 279, 523–525. [Google Scholar] [CrossRef]
  43. Ashley, C.L.; Abendroth, A.; McSharry, B.P.; Slobedman, B. Interferon-Independent Upregulation of Interferon-Stimulated Genes during Human Cytomegalovirus Infection Is Dependent on IRF3 Expression. Viruses 2019, 11, 246. [Google Scholar] [CrossRef] [Green Version]
  44. Lertsooksawat, W.; Wongnoppavich, A.; Chairatvit, K. Up-Regulation of Interferon-Stimulated Gene 15 and Its Conjugation Machinery, UbE1L and UbcH8 Expression by Tumor Necrosis Factor-α through P38 MAPK and JNK Signaling Pathways in Human Lung Carcinoma. Mol. Cell Biochem. 2019, 462, 51–59. [Google Scholar] [CrossRef]
  45. Radoshevich, L.; Impens, F.; Ribet, D.; Quereda, J.J.; Nam Tham, T.; Nahori, M.-A.; Bierne, H.; Dussurget, O.; Pizarro-Cerdá, J.; Knobeloch, K.-P.; et al. ISG15 Counteracts Listeria Monocytogenes Infection. eLife 2015, 4, e06848. [Google Scholar] [CrossRef]
  46. Yuan, W.; Krug, R.M. Influenza B Virus NS1 Protein Inhibits Conjugation of the Interferon (IFN)-Induced Ubiquitin-like ISG15 Protein. EMBO J. 2001, 20, 362–371. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, K.I.; Giannakopoulos, N.V.; Virgin, H.W.; Zhang, D.-E. Interferon-Inducible Ubiquitin E2, Ubc8, Is a Conjugating Enzyme for Protein ISGylation. Mol. Cell Biol. 2004, 24, 9592–9600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Zhao, C.; Beaudenon, S.L.; Kelley, M.L.; Waddell, M.B.; Yuan, W.; Schulman, B.A.; Huibregtse, J.M.; Krug, R.M. The UbcH8 Ubiquitin E2 Enzyme Is Also the E2 Enzyme for ISG15, an IFN-α/β-Induced Ubiquitin-like Protein. Proc. Natl. Acad. Sci. USA 2004, 101, 7578–7582. [Google Scholar] [CrossRef] [Green Version]
  49. Dastur, A.; Beaudenon, S.; Kelley, M.; Krug, R.M.; Huibregtse, J.M. Herc5, an Interferon-Induced HECT E3 Enzyme, Is Required for Conjugation of ISG15 in Human Cells. J. Biol. Chem. 2006, 281, 4334–4338. [Google Scholar] [CrossRef] [Green Version]
  50. Oudshoorn, D.; van Boheemen, S.; Sánchez-Aparicio, M.T.; Rajsbaum, R.; García-Sastre, A.; Versteeg, G.A. HERC6 Is the Main E3 Ligase for Global ISG15 Conjugation in Mouse Cells. PLoS ONE 2012, 7, e29870. [Google Scholar] [CrossRef] [Green Version]
  51. Ketscher, L.; Basters, A.; Prinz, M.; Knobeloch, K.-P. MHERC6 Is the Essential ISG15 E3 Ligase in the Murine System. Biochem. Biophys. Res. Commun. 2012, 417, 135–140. [Google Scholar] [CrossRef] [PubMed]
  52. Malakhov, M.P.; Malakhova, O.A.; Kim, K.I.; Ritchie, K.J.; Zhang, D.-E. UBP43 (USP18) Specifically Removes ISG15 from Conjugated Proteins. J. Biol. Chem. 2002, 277, 9976–9981. [Google Scholar] [CrossRef] [Green Version]
  53. Durfee, L.A.; Lyon, N.; Seo, K.; Huibregtse, J.M. The ISG15 Conjugation System Broadly Targets Newly Synthesized Proteins: Implications for the Antiviral Function of ISG15. Mol. Cell 2010, 38, 722–732. [Google Scholar] [CrossRef]
  54. Enchev, R.I.; Schulman, B.A.; Peter, M. Protein Neddylation: Beyond Cullin–RING Ligases. Nat. Rev. Mol. Cell Biol. 2015, 16, 30–44. [Google Scholar] [CrossRef] [PubMed]
  55. Duda, D.M.; Borg, L.A.; Scott, D.C.; Hunt, H.W.; Hammel, M.; Schulman, B.A. Structural Insights into NEDD8 Activation of Cullin-RING Ligases: Conformational Control of Conjugation. Cell 2008, 134, 995. [Google Scholar] [CrossRef] [Green Version]
  56. Dubiel, W.; Chaithongyot, S.; Dubiel, D.; Naumann, M. The COP9 Signalosome: A Multi-DUB Complex. Biomolecules 2020, 10, 1082. [Google Scholar] [CrossRef] [PubMed]
  57. Flick, K.; Kaiser, P. Set Them Free: F-Box Protein Exchange by Cand1. Cell Res. 2013, 23, 870–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Li, H.; Zhu, H.; Liu, Y.; He, F.; Xie, P.; Zhang, L. Itch Promotes the Neddylation of JunB and Regulates JunB-Dependent Transcription. Cell. Signal. 2016, 28, 1186–1195. [Google Scholar] [CrossRef] [Green Version]
  59. Um, J.W.; Han, K.A.; Im, E.; Oh, Y.; Lee, K.; Chung, K.C. Neddylation Positively Regulates the Ubiquitin E3 Ligase Activity of Parkin. J. Neurosci. Res. 2012, 90, 1030–1042. [Google Scholar] [CrossRef]
  60. Xirodimas, D.P.; Saville, M.K.; Bourdon, J.-C.; Hay, R.T.; Lane, D.P. Mdm2-Mediated NEDD8 Conjugation of P53 Inhibits Its Transcriptional Activity. Cell 2004, 118, 83–97. [Google Scholar] [CrossRef] [Green Version]
  61. Batuello, C.N.; Hauck, P.M.; Gendron, J.M.; Lehman, J.A.; Mayo, L.D. Src Phosphorylation Converts Mdm2 from a Ubiquitinating to a Neddylating E3 Ligase. Proc. Natl. Acad. Sci. USA 2015, 112, 1749–1754. [Google Scholar] [CrossRef] [Green Version]
  62. Zou, T.; Zhang, J. Diverse and Pivotal Roles of Neddylation in Metabolism and Immunity. FEBS J. 2021, 288, 3884–3912. [Google Scholar] [CrossRef] [PubMed]
  63. Krammer, F.; Smith, G.J.D.; Fouchier, R.A.M.; Peiris, M.; Kedzierska, K.; Doherty, P.C.; Palese, P.; Shaw, M.L.; Treanor, J.; Webster, R.G.; et al. Influenza. Nat. Rev. Dis. Primers 2018, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  64. Vasin, A.V.; Temkina, O.A.; Egorov, V.V.; Klotchenko, S.A.; Plotnikova, M.A.; Kiselev, O.I. Molecular Mechanisms Enhancing the Proteome of Influenza A Viruses: An Overview of Recently Discovered Proteins. Virus. Res. 2014, 185, 53–63. [Google Scholar] [CrossRef]
  65. Subbarao, E.K.; London, W.; Murphy, B.R. A single amino acid in the PB2 gene of influenza A virus is a determinant of host range. J. Virol. 1993, 67, 1761–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Robb, N.C.; Smith, M.; Vreede, F.T.; Fodor, E. NS2/NEP Protein Regulates Transcription and Replication of the Influenza Virus RNA Genome. J. Gen. Virol. 2009, 90, 1398–1407. [Google Scholar] [CrossRef] [PubMed]
  67. Hutchinson, E.C.; Denham, E.M.; Thomas, B.; Trudgian, D.C.; Hester, S.S.; Ridlova, G.; York, A.; Turrell, L.; Fodor, E. Mapping the Phosphoproteome of Influenza A and B Viruses by Mass Spectrometry. PLoS Pathog. 2012, 8, e1002993. [Google Scholar] [CrossRef] [Green Version]
  68. Kirui, J.; Mondal, A.; Mehle, A. Ubiquitination upregulates influenza virus polymerase function. J. Virol. 2016, 90, 10906–10914. [Google Scholar] [CrossRef] [Green Version]
  69. Pal, S.; Santos, A.; Rosas, J.M.; Ortiz-Guzman, J.; Rosas-Acosta, G. Influenza A Virus Interacts Extensively with the Cellular SUMOylation System during Infection. Virus. Res. 2011, 158, 12–27. [Google Scholar] [CrossRef]
  70. Han, Q.; Chang, C.; Li, L.; Klenk, C.; Cheng, J.; Chen, Y.; Xia, N.; Shu, Y.; Chen, Z.; Gabriel, G.; et al. Sumoylation of Influenza A Virus Nucleoprotein Is Essential for Intracellular Trafficking and Virus Growth. J. Virol. 2014, 88, 9379–9390. [Google Scholar] [CrossRef] [Green Version]
  71. Di Pietro, A.; Kajaste-Rudnitski, A.; Oteiza, A.; Nicora, L.; Towers, G.J.; Mechti, N.; Vicenzi, E. TRIM22 Inhibits Influenza A Virus Infection by Targeting the Viral Nucleoprotein for Degradation. J. Virol. 2013, 87, 4523–4533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Fu, B.; Wang, L.; Ding, H.; Schwamborn, J.C.; Li, S.; Dorf, M.E. TRIM32 senses and restricts influenza A virus by ubiquitination of PB1 polymerase. PLoS Pathog. 2015, 11, e1004960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Liao, T.L.; Wu, C.Y.; Su, W.C.; Jeng, K.S.; Lai, M.M.C. Ubiquitination and deubiquitination of NP protein regulates influenza A virus RNA replication. EMBO J. 2010, 29, 3879–3890. [Google Scholar] [CrossRef] [PubMed]
  74. Widjaja, I.; De Vries, E.; Tscherne, D.M.; García-Sastre, A.; Rottier, P.J.M.; De Haan, C.A.M. Inhibition of the Ubiquitin-Proteasome System Affects Influenza A Virus Infection at a Postfusion Step. J. Virol. 2010, 84, 9625–9631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Liu, C.-; Zhou, L.; Chen, G.; Krug, R.M. Battle between Influenza A Virus and a Newly Identified Antiviral Activity of the PARP-Containing ZAPL Protein. Proc. Natl. Acad. Sci. USA 2015, 112, 14048–14053. [Google Scholar] [CrossRef] [Green Version]
  76. Chesarino, N.M.; McMichael, T.M.; Yount, J.S. E3 Ubiquitin Ligase NEDD4 Promotes Influenza Virus Infection by Decreasing Levels of the Antiviral Protein IFITM3. PLoS Pathog. 2015, 11, e1005095. [Google Scholar] [CrossRef] [Green Version]
  77. Su, W.C.; Chen, Y.C.; Tseng, C.H.; Hsu, P.W.C.; Tung, K.F.; Jeng, K.S.; Lai, M.M.C. Pooled RNAi Screen Identifies Ubiquitin Ligase Itch as Crucial for Influenza A Virus Release from the Endosome during Virus Entry. Proc. Natl. Acad. Sci. USA 2013, 110, 17516–17521. [Google Scholar] [CrossRef] [Green Version]
  78. Watanabe, T.; Watanabe, S.; Kawaoka, Y. Cellular networks involved in the influenza virus life cycle. Cell Host Microbe 2010, 7, 427–439. [Google Scholar] [CrossRef] [Green Version]
  79. Khor, R.; McElroy, L.J.; Whittaker, G.R. The Ubiquitin-Vacuolar Protein Sorting System Is Selectively Required during Entry of Influenza Virus into Host Cells. Traffic 2003, 4, 857–868. [Google Scholar] [CrossRef] [Green Version]
  80. Calistri, A.; Munegato, D.; Carli, I.; Parolin, C.; Palu, G. The Ubiquitin-Conjugating System: Multiple Roles in Viral Replication and Infection. Cells 2014, 3, 386–417. [Google Scholar] [CrossRef] [Green Version]
  81. Wimmer, P.; Schreiner, S. Viral mimicry to usurp ubiquitin and sumo host pathways. Viruses 2015, 7, 4854–4877. [Google Scholar] [CrossRef] [Green Version]
  82. Smith, M.C.; Boutell, C.; Davido, D.J. HSV-1 ICP0: Paving the Way for Viral Replication. Future Virol. 2011, 6, 421–429. [Google Scholar] [CrossRef] [Green Version]
  83. Kroeker, A.L.; Ezzati, P.; Coombs, K.M.; Halayko, A.J. Influenza A Infection of Primary Human Airway Epithelial Cells Up-Regulates Proteins Related to Purine Metabolism and Ubiquitin-Related Signaling. J. Proteome Res. 2013, 12, 3139–3151. [Google Scholar] [CrossRef] [PubMed]
  84. Suzuki, Y.; Ito, T.; Suzuki, T.; Holland, R.E., Jr.; Chambers, T.M.; Kiso, M.; Ishida, H.; Kawaoka, Y. Sialic acid species as a determinant of the host range of influenza A viruses. J. Virol. 2000, 74, 11825–11831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Pinto, L.H.; Holsinger, L.J.; Lamb, R.A. Influenza virus M2 protein has ion channel activity. Cell 1992, 69, 517–528. [Google Scholar] [CrossRef]
  86. Stauffer, S.; Feng, Y.; Nebioglu, F.; Heilig, R.; Picotti, P.; Helenius, A. Stepwise priming by acidic pH and a high K+ concentration is required for efficient uncoating of influenza a virus cores after penetration. J. Virol. 2014, 88, 13029–13046. [Google Scholar] [CrossRef] [Green Version]
  87. White, J.; Kartenbeck, J.; Helenius, A. Membrane fusion activity of influenza virus. EMBO J. 1982, 1, 217–222. [Google Scholar] [CrossRef]
  88. Huotari, J.; Meyer-Schaller, N.; Hubner, M.; Stauffer, S.; Katheder, N.; Horvath, P.; Mancini, R.; Helenius, A.; Peter, M. Cullin-3 Regulates Late Endosome Maturation. Proc. Natl. Acad. Sci. USA 2012, 109, 823–828. [Google Scholar] [CrossRef] [Green Version]
  89. Hubner, M.; Peter, M. Cullin-3 and the endocytic system: New functions of ubiquitination for endosome maturation. Cell Logist 2012, 2, 166–168. [Google Scholar] [CrossRef] [Green Version]
  90. Cros, J.F.; García-Sastre, A.; Palese, P. An unconventional NLS is critical for the nuclear import of the influenza A virus nucleoprotein and ribonucleoprotein. Traffic 2005, 6, 205–213. [Google Scholar] [CrossRef]
  91. Eisfeld, A.J.; Neumann, G.; Kawaoka, Y. At the centre: Influenza A virus ribonucleoproteins. Nat. Rev. Microbiol. 2015, 13, 28–41. [Google Scholar] [CrossRef] [Green Version]
  92. Wu, W.W.H.; Sun, Y.-B.; Panté, N. Nuclear Import of Influenza A Viral Ribonucleoprotein Complexes Is Mediated by Two Nuclear Localization Sequences on Viral Nucleoprotein. Virol. J. 2007, 4, 49. [Google Scholar] [CrossRef] [Green Version]
  93. Ko, H.S.; Lee, Y.; Shin, J.-H.; Karuppagounder, S.S.; Gadad, B.S.; Koleske, A.J.; Pletnikova, O.; Troncoso, J.C.; Dawson, V.L.; Dawson, T.M. Phosphorylation by the C-Abl Protein Tyrosine Kinase Inhibits Parkin’s Ubiquitination and Protective Function. Proc. Natl. Acad. Sci. USA 2010, 107, 16691–16696. [Google Scholar] [CrossRef] [Green Version]
  94. Lee, Y.; Karuppagounder, S.S.; Shin, J.-H.; Lee, Y.-I.; Ko, H.S.; Swing, D.; Jiang, H.; Kang, S.-U.; Lee, B.D.; Kang, H.C.; et al. Parthanatos Mediates AIMP2 Activated Age Dependent Dopaminergic Neuronal Loss. Nat. Neurosci. 2013, 16, 1392–1400. [Google Scholar] [CrossRef] [PubMed]
  95. Gao, S.; Wu, J.; Liu, R.; Li, J.; Song, L.; Teng, Y.; Sheng, C.; Liu, D.; Yao, C.; Chen, H.; et al. Interaction of NS2 with AIMP2 Facilitates the Switch from Ubiquitination to SUMOylation of M1 in Influenza a Virus-Infected Cells. J. Virol. 2015, 89, 300–311. [Google Scholar] [CrossRef] [Green Version]
  96. Wu, C.-; Jeng, K.; Lai, M.M. The SUMOylation of Matrix Protein M1 Modulates the Assembly and Morphogenesis of Influenza A Virus. J. Virol. 2011, 85, 6618–6628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Iwasaki, A.; Pillai, P.S. Innate Immunity to Influenza Virus Infection. Nat. Rev. Immunol. 2014, 14, 315–328. [Google Scholar] [CrossRef] [PubMed]
  98. Davis, M.E.; Gack, M.U. Ubiquitination in the Antiviral Immune Response. Virology 2015, 479, 52–65. [Google Scholar] [CrossRef] [Green Version]
  99. Feng, W.; Sun, X.; Shi, N.; Zhang, M.; Guan, Z.; Duan, M. Influenza a Virus NS1 Protein Induced A20 Contributes to Viral Replication by Suppressing Interferon-Induced Antiviral Response. Biochem. Biophys. Res. Commun. 2017, 482, 1107–1113. [Google Scholar] [CrossRef]
  100. Davidson, S.; Crotta, S.; McCabe, T.M.; Wack, A. Pathogenic Potential of Interferon Aβ in Acute Influenza Infection. Nat. Commun. 2014, 5, 3864. [Google Scholar] [CrossRef] [Green Version]
  101. Davidson, S.; McCabe, T.M.; Crotta, S.; Gad, H.H.; Hessel, E.M.; Beinke, S.; Hartmann, R.; Wack, A. IFNλ Is a Potent Anti-influenza Therapeutic without the Inflammatory Side Effects of IFNα Treatment. EMBO Mol. Med. 2016, 8, 1099–1112. [Google Scholar] [CrossRef] [PubMed]
  102. Han, K.; Lou, D.I.; Sawyer, S.L. Identification of a Genomic Reservoir for New TRIM Genes in Primate Genomes. PLoS Genet. 2011, 7, e1002388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Liu, B.; Li, N.L.; Wang, J.; Shi, P.-Y.; Wang, T.; Miller, M.A.; Li, K. Overlapping and Distinct Molecular Determinants Dictating the Antiviral Activities of TRIM56 against Flaviviruses and Coronavirus. J. Virol. 2014, 88, 13821–13835. [Google Scholar] [CrossRef] [Green Version]
  104. Rajsbaum, R.; García-Sastre, A.; Versteeg, G.A. TRIMmunity: The Roles of the TRIM E3-Ubiquitin Ligase Family in Innate Antiviral Immunity. J. Mol. Biol. 2014, 426, 1265–1284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Lee, H.-R.; Lee, M.K.; Kim, C.W.; Kim, M. TRIM Proteins and Their Roles in the Influenza Virus Life Cycle. Microorganisms 2020, 8, 1424. [Google Scholar] [CrossRef]
  106. Gack, M.U.; Shin, Y.C.; Joo, C.-; Urano, T.; Liang, C.; Sun, L.; Takeuchi, O.; Akira, S.; Chen, Z.; Inoue, S.; et al. TRIM25 RING-Finger E3 Ubiquitin Ligase Is Essential for RIG-I-Mediated Antiviral Activity. Nature 2007, 446, 916–920. [Google Scholar] [CrossRef]
  107. Gack, M.U.; Albrecht, R.A.; Urano, T.; Inn, K.-; Huang, I.-; Carnero, E.; Farzan, M.; Inoue, S.; Jung, J.U.; García-Sastre, A. Influenza A Virus NS1 Targets the Ubiquitin Ligase TRIM25 to Evade Recognition by the Host Viral RNA Sensor RIG-I. Cell Host Microbe 2009, 5, 439–449. [Google Scholar] [CrossRef] [Green Version]
  108. Pauli, E.-; Chan, Y.K.; Davis, M.E.; Gableske, S.; Wang, M.K.; Feister, K.F.; Gack, M.U. The Ubiquitin-Specific Protease USP15 Promotes RIG-I-Mediated Antiviral Signaling by Deubiquitylating TRIM25. Sci. Signal. 2014, 7, ra3. [Google Scholar] [CrossRef] [Green Version]
  109. Marín, I. Origin and diversification of TRIM ubiquitin ligases. PLoS ONE 2012, 7, e50030. [Google Scholar] [CrossRef] [Green Version]
  110. Esposito, D.; Koliopoulos, M.G.; Rittinger, K. Structural Determinants of TRIM Protein Function. Biochem. Soc. Trans. 2017, 45, 183–191. [Google Scholar] [CrossRef]
  111. Reymond, A.; Meroni, G.; Fantozzi, A.; Merla, G.; Cairo, S.; Luzi, L.; Riganelli, D.; Zanaria, E.; Messali, S.; Cainarca, S.; et al. The Tripartite Motif Family Identifies Cell Compartments. EMBO J. 2001, 20, 2140–2151. [Google Scholar] [CrossRef] [Green Version]
  112. Scherer, M.; Klingl, S.; Sevvana, M.; Otto, V.; Schilling, E.-; Stump, J.D.; Müller, R.; Reuter, N.; Sticht, H.; Muller, Y.A.; et al. Crystal Structure of Cytomegalovirus IE1 Protein Reveals Targeting of TRIM Family Member PML via Coiled-Coil Interactions. PLoS Pathog. 2014, 10, e1004512. [Google Scholar] [CrossRef] [Green Version]
  113. Schilling, E.-M.; Scherer, M.; Reuter, N.; Schweininger, J.; Muller, Y.A.; Stamminger, T. The Human Cytomegalovirus IE1 Protein Antagonizes PML Nuclear Body-Mediated Intrinsic Immunity via the Inhibition of PML De Novo SUMOylation. J. Virol. 2017, 91, e02049-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Doyle, J.M.; Gao, J.; Wang, J.; Yang, M.; Potts, P.R. MAGE-RING Protein Complexes Comprise a Family of E3 Ubiquitin Ligases. Mol. Cell 2010, 39, 963–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Pineda, C.T.; Ramanathan, S.; Fon Tacer, K.; Weon, J.L.; Potts, M.B.; Ou, Y.-; White, M.A.; Potts, P.R. Degradation of AMPK by a Cancer-Specific Ubiquitin Ligase. Cell 2015, 160, 715–728. [Google Scholar] [CrossRef] [Green Version]
  116. Van Gent, M.; Sparrer, K.M.J.; Gack, M.U. TRIM Proteins and Their Roles in Antiviral Host Defenses. Annu. Rev. Virol. 2018, 5, 385–405. [Google Scholar] [CrossRef]
  117. Cambiaghi, V.; Giuliani, V.; Lombardi, S.; Marinelli, C.; Toffalorio, F.; Pelicci, P.G. TRIM Proteins in Cancer. Adv. Exp. Med. Biol. 2012, 770, 77–91. [Google Scholar] [CrossRef] [PubMed]
  118. Koliopoulos, M.G.; Lethier, M.; Van Der Veen, A.G.; Haubrich, K.; Hennig, J.; Kowalinski, E.; Stevens, R.V.; Martin, S.R.; Reis e Sousa, C.; Cusack, S.; et al. Molecular mechanism of influenza A NS1-mediated TRIM25 recognition and inhibition. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef]
  119. Yoneyama, M.; Fujita, T. Structural Mechanism of RNA Recognition by the RIG-I-like Receptors. Immunity 2008, 29, 178–181. [Google Scholar] [CrossRef] [Green Version]
  120. Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA Helicase RIG-I Has an Essential Function in Double-Stranded RNA-Induced Innate Antiviral Responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef] [PubMed]
  121. Baum, A.; Sachidanandam, R.; García-Sastre, A. Preference of RIG-I for Short Viral RNA Molecules in Infected Cells Revealed by next-Generation Sequencing. Proc. Natl. Acad. Sci. USA 2010, 107, 16303–16308. [Google Scholar] [CrossRef] [Green Version]
  122. Versteeg, G.A.; García-Sastre, A. Viral Tricks to Grid-Lock the Type I Interferon System. Curr. Opin. Microbiol. 2010, 13, 508–516. [Google Scholar] [CrossRef]
  123. Hornung, V.; Ellegast, J.; Kim, S.; Brzózka, K.; Jung, A.; Kato, H.; Poeck, H.; Akira, S.; Conzelmann, K.; Schlee, M.; et al. 5′Triphosphate RNA Is the Ligand for RIG-I. Science 2006, 314, 994–997. [Google Scholar] [CrossRef] [Green Version]
  124. Weber-Gerlach, M.; Weber, F. Standing on Three Legs: Antiviral Activities of RIG-I against Influenza Viruses. Curr. Opin. Immunol. 2016, 42, 71–75. [Google Scholar] [CrossRef] [PubMed]
  125. Pichlmair, A.; Schulz, O.; Tan, C.P.; Näslund, T.I.; Liljeström, P.; Weber, F.; Sousa, C.R.E. RIG-I-Mediated Antiviral Responses to Single-Stranded RNA Bearing 5′-Phosphates. Science 2006, 314, 997–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Rehwinkel, J.; Tan, C.P.; Goubau, D.; Schulz, O.; Pichlmair, A.; Bier, K.; Robb, N.; Vreede, F.; Barclay, W.; Fodor, E.; et al. RIG-I Detects Viral Genomic RNA during Negative-Strand RNA Virus Infection. Cell 2010, 140, 397–408. [Google Scholar] [CrossRef] [Green Version]
  127. Weber, M.; Sediri, H.; Felgenhauer, U.; Binzen, I.; Bänfer, S.; Jacob, R.; Brunotte, L.; García-Sastre, A.; Schmid-Burgk, J.L.; Schmidt, T.; et al. Influenza Virus Adaptation PB2-627K Modulates Nucleocapsid Inhibition by the Pathogen Sensor RIG-I. Cell Host Microbe 2015, 17, 309–319. [Google Scholar] [CrossRef] [Green Version]
  128. Wu, W.; Zhang, W.; Duggan, E.S.; Booth, J.L.; Zou, M.-H.; Metcalf, J.P. RIG-I and TLR3 Are Both Required for Maximum Interferon Induction by Influenza Virus in Human Lung Alveolar Epithelial Cells. Virology 2015, 482, 181–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Kolakofsky, D.; Kowalinski, E.; Cusack, S. A Structure-Based Model of RIG-I Activation. RNA 2012, 18, 2118–2127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Reikine, S.; Nguyen, J.B.; Modis, Y. Pattern Recognition and Signaling Mechanisms of RIG-I and MDA5. Front. Immunol. 2014, 5, 342. [Google Scholar] [CrossRef] [Green Version]
  131. Patel, J.R.; Jain, A.; Chou, Y.; Baum, A.; Ha, T.; García-Sastre, A. ATPase-Driven Oligomerization of RIG-I on RNA Allows Optimal Activation of Type-I Interferon. EMBO Rep. 2013, 14, 780–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Ferrage, F.; Dutta, K.; Nistal-Villán, E.; Patel, J.R.; Sánchez-Aparicio, M.T.; De Ioannes, P.; Buku, A.; Aseguinolaza, G.G.; García-Sastre, A.; Aggarwal, A.K. Structure and Dynamics of the Second CARD of Human RIG-I Provide Mechanistic Insights into Regulation of RIG-I Activation. Structure 2012, 20, 2048–2061. [Google Scholar] [CrossRef] [Green Version]
  133. Kowalinski, E.; Lunardi, T.; McCarthy, A.A.; Louber, J.; Brunel, J.; Grigorov, B.; Gerlier, D.; Cusack, S. Structural Basis for the Activation of Innate Immune Pattern-Recognition Receptor RIG-I by Viral RNA. Cell 2011, 147, 423–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Oshiumi, H.; Miyashita, M.; Inoue, N.; Okabe, M.; Matsumoto, M.; Seya, T. The ubiquitin ligase riplet is essential for RIG-I-dependent innate immune responses to RNA virus infection. Cell Host Microbe 2010, 8, 496–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Gao, D.; Yang, Y.-K.; Wang, R.-P.; Zhou, X.; Diao, F.-C.; Li, M.-D.; Zhai, Z.-H.; Jiang, Z.-F.; Chen, D.-Y. REUL Is a Novel E3 Ubiquitin Ligase and Stimulator of Retinoic-Acid-Inducible Gene-I. PLoS ONE 2009, 4, e5760. [Google Scholar] [CrossRef] [Green Version]
  136. Oshiumi, H.; Matsumoto, M.; Hatakeyama, S.; Seya, T. Riplet/RNF135, a RING Finger Protein, Ubiquitinates RIG-I to Promote Interferon-Beta Induction during the Early Phase of Viral Infection. J. Biol. Chem. 2009, 284, 807–817. [Google Scholar] [CrossRef] [Green Version]
  137. Sun, X.; Xian, H.; Tian, S.; Sun, T.; Qin, Y.; Zhang, S.; Cui, J. A hierarchical mechanism of RIG-I ubiquitination provides sensitivity, robustness and synergy in antiviral immune responses. Sci. Rep. 2016, 6, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Cadena, C.; Ahmad, S.; Xavier, A.; Willemsen, J.; Park, S.; Park, J.W.; Oh, S.-W.; Fujita, T.; Hou, F.; Binder, M.; et al. Ubiquitin-Dependent and -Independent Roles of E3 Ligase RIPLET in Innate Immunity. Cell 2019, 177, 1187–1200.e16. [Google Scholar] [CrossRef]
  139. Sanchez, J.G.; Chiang, J.J.; Sparrer, K.M.J.; Alam, S.L.; Chi, M.; Roganowicz, M.D.; Sankaran, B.; Gack, M.U.; Pornillos, O. Mechanism of TRIM25 Catalytic Activation in the Antiviral RIG-I Pathway. Cell Rep. 2016, 16, 1315–1325. [Google Scholar] [CrossRef] [Green Version]
  140. D’Cruz, A.A.; Kershaw, N.J.; Chiang, J.J.; Wang, M.K.; Nicola, N.A.; Babon, J.J.; Gack, M.U.; Nicholson, S.E. Crystal Structure of the TRIM25 B30.2 (PRYSPRY) Domain: A Key Component of Antiviral Signalling. Biochem. J. 2013, 456, 231–240. [Google Scholar] [CrossRef] [Green Version]
  141. Peisley, A.; Wu, B.; Xu, H.; Chen, Z.J.; Hur, S. Structural Basis for Ubiquitin-Mediated Antiviral Signal Activation by RIG-I. Nature 2014, 508, 110–114. [Google Scholar] [CrossRef]
  142. Okamoto, M.; Kouwaki, T.; Fukushima, Y.; Oshiumi, H. Regulation of RIG-I activation by K63-linked polyubiquitination. Front. Immunol. 2018, 8, 1942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jiang, Q.; Chen, Z.J. Structural Insights into the Activation of RIG-I, a Nanosensor for Viral RNAs. EMBO Rep. 2012, 13, 7–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Sánchez-Aparicio, M.T.; Ayllón, J.; Leo-Macias, A.; Wolff, T.; García-Sastre, A. Subcellular Localizations of RIG-I, TRIM25, and MAVS Complexes. J. Virol. 2017, 91, e01155-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. 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]
  146. Kawai, T.; Takahashi, K.; Sato, S.; Coban, C.; Kumar, H.; Kato, H.; Ishii, K.J.; Takeuchi, O.; Akira, S. IPS-1, an Adaptor Triggering RIG-I- and Mda5-Mediated Type I Interferon Induction. Nat. Immunol. 2005, 6, 981–988. [Google Scholar] [CrossRef]
  147. Wu, B.; Peisley, A.; Tetrault, D.; Li, Z.; Egelman, E.H.; Magor, K.E.; Walz, T.; Penczek, P.A.; Hur, S. Molecular Imprinting as a Signal-Activation Mechanism of the Viral RNA Sensor RIG-I. Mol. Cell 2014, 55, 511–523. [Google Scholar] [CrossRef] [Green Version]
  148. Xu, H.; He, X.; Zheng, H.; Huang, L.J.; Hou, F.; Yu, Z.; de la Cruz, M.J.; Borkowski, B.; Zhang, X.; Chen, Z.J.; et al. Correction: Structural Basis for the Prion-like MAVS Filaments in Antiviral Innate Immunity. eLife 2015, 4, e07546. [Google Scholar] [CrossRef]
  149. Li, X.-D.; Sun, L.; Seth, R.B.; Pineda, G.; Chen, Z.J. Hepatitis C Virus Protease NS3/4A Cleaves Mitochondrial Antiviral Signaling Protein off the Mitochondria to Evade Innate Immunity. Proc. Natl. Acad. Sci. USA 2005, 102, 17717–17722. [Google Scholar] [CrossRef] [Green Version]
  150. Hou, F.; Sun, L.; Zheng, H.; Skaug, B.; Jiang, Q.-X.; Chen, Z.J. MAVS Forms Functional Prion-like Aggregates to Activate and Propagate Antiviral Innate Immune Response. Cell 2011, 146, 448–461. [Google Scholar] [CrossRef] [Green Version]
  151. Xu, L.-G.; Wang, Y.-Y.; Han, K.-J.; Li, L.-Y.; Zhai, Z.; Shu, H.-B. VISA Is an Adapter Protein Required for Virus-Triggered IFN-Beta Signaling. Mol. Cell 2005, 19, 727–740. [Google Scholar] [CrossRef] [PubMed]
  152. Gack, M.U. Mechanisms of RIG-I-Like Receptor Activation and Manipulation by Viral Pathogens. J. Virol. 2014, 88, 5213–5216. [Google Scholar] [CrossRef] [Green Version]
  153. 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]
  154. Takeuchi, O.; Akira, S. Innate Immunity to Virus Infection. Immunol. Rev. 2009, 227, 75–86. [Google Scholar] [CrossRef]
  155. Wolff, T.; Ludwig, S. Influenza Viruses Control the Vertebrate Type I Interferon System: Factors, Mechanisms, and Consequences. J. Interferon Cytokine Res. 2009, 29, 549–557. [Google Scholar] [CrossRef] [PubMed]
  156. Hermant, P.; Michiels, T. Interferon-λ in the Context of Viral Infections: Production, Response and Therapeutic Implications. J. Innate. Immun. 2014, 6, 563–574. [Google Scholar] [CrossRef]
  157. Orian, A.; Gonen, H.; Bercovich, B.; Fajerman, I.; Eytan, E.; Israël, A.; Mercurio, F.; Iwai, K.; Schwartz, A.L.; Ciechanover, A. SCFβ-TrCP Ubiquitin Ligase-Mediated Processing of NF-ΚB P105 Requires Phosphorylation of Its C-Terminus by IκB Kinase. EMBO J. 2000, 19, 2580–2591. [Google Scholar] [CrossRef] [Green Version]
  158. Mogensen, T.H.; Paludan, S.R. Molecular Pathways in Virus-Induced Cytokine Production. Microbiol. Mol. Biol. Rev. 2001, 65, 131–150. [Google Scholar] [CrossRef] [Green Version]
  159. Balachandran, S.; Beg, A.A. Defining Emerging Roles for NF-ΚB in Antivirus Responses: Revisiting the Interferon-β Enhanceosome Paradigm. PLoS Pathog. 2011, 7, e1002165. [Google Scholar] [CrossRef] [Green Version]
  160. Adhikari, A.; Xu, M.; Chen, Z.J. Ubiquitin-Mediated Activation of TAK1 and IKK. Oncogene 2007, 26, 3214–3226. [Google Scholar] [CrossRef] [Green Version]
  161. Fang, R.; Jiang, Q.; Zhou, X.; Wang, C.; Guan, Y.; Tao, J.; Xi, J.; Feng, J.-M.; Jiang, Z. MAVS Activates TBK1 and IKKε through TRAFs in NEMO Dependent and Independent Manner. PLoS Pathog. 2017, 13, e1006720. [Google Scholar] [CrossRef] [PubMed]
  162. Andreakos, E.; Salagianni, M.; Galani, I.E.; Koltsida, O. Interferon-Λs: Front-Line Guardians of Immunity and Homeostasis in the Respiratory Tract. Front. Immunol. 2017, 8, 1232. [Google Scholar] [CrossRef] [Green Version]
  163. Galani, I.E.; Triantafyllia, V.; Eleminiadou, E.-E.; Koltsida, O.; Stavropoulos, A.; Manioudaki, M.; Thanos, D.; Doyle, S.E.; Kotenko, S.V.; Thanopoulou, K.; et al. Interferon-λ Mediates Non-Redundant Front-Line Antiviral Protection against Influenza Virus Infection without Compromising Host Fitness. Immunity 2017, 46, 875–890.e6. [Google Scholar] [CrossRef] [PubMed]
  164. Levy, D.E.; Marié, I.J.; Durbin, J.E. Induction and Function of Type I and III Interferon in Response to Viral Infection. Curr. Opin. Virol. 2011, 1, 476–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Schindler, C.; Levy, D.E.; Decker, T. JAK-STAT Signaling: From Interferons to Cytokines. J. Biol. Chem. 2007, 282, 20059–20063. [Google Scholar] [CrossRef] [Green Version]
  166. Hoffmann, H.-H.; Schneider, W.M.; Rice, C.M. Interferons and Viruses: An Evolutionary Arms Race of Molecular Interactions. Trends Immunol. 2015, 36, 124–138. [Google Scholar] [CrossRef] [Green Version]
  167. Villalón-Letelier, F.; Brooks, A.G.; Saunders, P.M.; Londrigan, S.L.; Reading, P.C. Host Cell Restriction Factors That Limit Influenza A Infection. Viruses 2017, 9, 376. [Google Scholar] [CrossRef] [Green Version]
  168. Qu, H.; Li, J.; Yang, L.; Sun, L.; Liu, W.; He, H. Influenza A Virus-Induced Expression of ISG20 Inhibits Viral Replication by Interacting with Nucleoprotein. Virus Genes 2016, 52, 759–767. [Google Scholar] [CrossRef]
  169. Desai, T.M.; Marin, M.; Chin, C.R.; Savidis, G.; Brass, A.L.; Melikyan, G.B. IFITM3 Restricts Influenza A Virus Entry by Blocking the Formation of Fusion Pores Following Virus-Endosome Hemifusion. PLoS Pathog. 2014, 10, e1004048. [Google Scholar] [CrossRef] [Green Version]
  170. Turan, K.; Mibayashi, M.; Sugiyama, K.; Saito, S.; Numajiri, A.; Nagata, K. Nuclear MxA Proteins Form a Complex with Influenza Virus NP and Inhibit the Transcription of the Engineered Influenza Virus Genome. Nucleic Acids Res. 2004, 32, 643–652. [Google Scholar] [CrossRef]
  171. Pavlovic, J.; Arzet, H.A.; Hefti, H.P.; Frese, M.; Rost, D.; Ernst, B.; Kolb, E.; Staeheli, P.; Haller, O. Enhanced Virus Resistance of Transgenic Mice Expressing the Human MxA Protein. J. Virol. 1995, 69, 4506–4510. [Google Scholar] [CrossRef] [Green Version]
  172. Haller, O.; Staeheli, P.; Schwemmle, M.; Kochs, G. Mx GTPases: Dynamin-like Antiviral Machines of Innate Immunity. Trends Microbiol. 2015, 23, 154–163. [Google Scholar] [CrossRef]
  173. Feeley, E.M.; Sims, J.S.; John, S.P.; Chin, C.R.; Pertel, T.; Chen, L.M.; Gaiha, G.D.; Ryan, B.J.; Donis, R.O.; Elledge, S.J.; et al. IFITM3 Inhibits Influenza a Virus Infection by Preventing Cytosolic Entry. PLoS Pathog. 2011, 7, e1002337. [Google Scholar] [CrossRef] [Green Version]
  174. Brass, A.L.; Huang, I.-C.; Benita, Y.; John, S.P.; Krishnan, M.N.; Feeley, E.M.; Ryan, B.J.; Weyer, J.L.; van der Weyden, L.; Fikrig, E.; et al. The IFITM Proteins Mediate Cellular Resistance to Influenza A H1N1 Virus, West Nile Virus, and Dengue Virus. Cell 2009, 139, 1243–1254. [Google Scholar] [CrossRef] [Green Version]
  175. Bailey, C.C.; Zhong, G.; Huang, I.-C.; Farzan, M. IFITM-Family Proteins: The Cell’s First Line of Antiviral Defense. Annu. Rev. Virol. 2014, 1, 261–283. [Google Scholar] [CrossRef] [Green Version]
  176. Clemens, M.J.; Williams, B.R.G. Inhibition of Cell-Free Protein Synthesis by PppA2′ P5′ A2′ P5′ A: A Novel Oligonucleotide Synthesized by Interferon-Treated L Cell Extracts. Cell 1978, 13, 565–572. [Google Scholar] [CrossRef]
  177. Silverman, R.H.; Weiss, S.R. Viral Phosphodiesterases That Antagonize Double-Stranded RNA Signaling to RNase L by Degrading 2–5A. J. Interferon Cytokine Res. 2014, 34, 455–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Malathi, K.; Dong, B.; Gale, M.; Silverman, R.H. Small Self-RNA Generated by RNase L Amplifies Antiviral Innate Immunity. Nature 2007, 448, 816–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Patel, R.C.; Sen, G.C. PACT, a Protein Activator of the Interferon-Induced Protein Kinase, PKR. EMBO J. 1998, 17, 4379–4390. [Google Scholar] [CrossRef]
  180. Kumar, A.; Haque, J.; Lacoste, J.; Hiscott, J.; Williams, B.R. Double-Stranded RNA-Dependent Protein Kinase Activates Transcription Factor NF-Kappa B by Phosphorylating I Kappa, B. Proc. Natl. Acad. Sci. USA 1994, 91, 6288–6292. [Google Scholar] [CrossRef] [Green Version]
  181. Zhang, P.; Samuel, C.E. Induction of Protein Kinase PKR-Dependent Activation of Interferon Regulatory Factor 3 by Vaccinia Virus Occurs through Adapter IPS-1 Signaling. J. Biol. Chem. 2008, 283, 34580–34587. [Google Scholar] [CrossRef] [Green Version]
  182. Schulz, O.; Pichlmair, A.; Rehwinkel, J.; Rogers, N.C.; Scheuner, D.; Kato, H.; Takeuchi, O.; Akira, S.; Kaufman, R.J.; Reis e Sousa, C. Protein Kinase R Contributes to Immunity against Specific Viruses by Regulating Interferon MRNA Integrity. Cell Host Microbe 2010, 7, 354–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Chen, I.-Y.; Ichinohe, T. Response of Host Inflammasomes to Viral Infection. Trends Microbiol. 2015, 23, 55–63. [Google Scholar] [CrossRef]
  184. Groslambert, M.; Py, B.F. Spotlight on the NLRP3 Inflammasome Pathway. J. Inflamm. Res. 2018, 11, 359–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Moriyama, M.; Chen, I.-Y.; Kawaguchi, A.; Koshiba, T.; Nagata, K.; Takeyama, H.; Hasegawa, H.; Ichinohe, T. The RNA- and TRIM25-Binding Domains of Influenza Virus NS1 Protein Are Essential for Suppression of NLRP3 Inflammasome-Mediated Interleukin-1β Secretion. J. Virol. 2016, 90, 4105–4114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Wang, X.; Jiang, W.; Yan, Y.; Gong, T.; Han, J.; Tian, Z.; Zhou, R. RNA Viruses Promote Activation of the NLRP3 Inflammasome through a RIP1-RIP3-DRP1 Signaling Pathway. Nat. Immunol. 2014, 15, 1126–1133. [Google Scholar] [CrossRef]
  187. Chen, W.; Xu, Y.; Li, H.; Tao, W.; Xiang, Y.; Huang, B.; Niu, J.; Zhong, J.; Meng, G. HCV Genomic RNA Activates the NLRP3 Inflammasome in Human Myeloid Cells. PLoS ONE 2014, 9, e84953. [Google Scholar] [CrossRef] [Green Version]
  188. Pothlichet, J.; Meunier, I.; Davis, B.K.; Ting, J.P.-Y.; Skamene, E.; von Messling, V.; Vidal, S.M. Type I IFN Triggers RIG-I/TLR3/NLRP3-Dependent Inflammasome Activation in Influenza A Virus Infected Cells. PLoS Pathog. 2013, 9, e1003256. [Google Scholar] [CrossRef] [Green Version]
  189. Lu, B.; Nakamura, T.; Inouye, K.; Li, J.; Tang, Y.; Lundbäck, P.; Valdes-Ferrer, S.I.; Olofsson, P.S.; Kalb, T.; Roth, J.; et al. Novel Role of PKR in Inflammasome Activation and HMGB1 Release. Nature 2012, 488, 670–674. [Google Scholar] [CrossRef] [Green Version]
  190. Ichinohe, T.; Pang, I.K.; Iwasaki, A. Influenza Virus Activates Inflammasomes via Its Intracellular M2 Ion Channel. Nat. Immunol. 2010, 11, 404–410. [Google Scholar] [CrossRef]
  191. McAuley, J.L.; Tate, M.D.; MacKenzie-Kludas, C.J.; Pinar, A.; Zeng, W.; Stutz, A.; Latz, E.; Brown, L.E.; Mansell, A. Activation of the NLRP3 Inflammasome by IAV Virulence Protein PB1-F2 Contributes to Severe Pathophysiology and Disease. PLoS Pathog. 2013, 9, e1003392. [Google Scholar] [CrossRef] [Green Version]
  192. Schroder, K.; Tschopp, J. The Inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [Green Version]
  193. García-Sastre, A. Induction and Evasion of Type I Interferon Responses by Influenza Viruses. Virus Res. 2011, 162, 12–18. [Google Scholar] [CrossRef] [Green Version]
  194. Mizushima, N.; Komatsu, M. Autophagy: Renovation of Cells and Tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [Green Version]
  195. Levine, B.; Kroemer, G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 2019, 176, 11–42. [Google Scholar] [CrossRef] [Green Version]
  196. Meyerson, N.R.; Zhou, L.; Guo, Y.R.; Zhao, C.; Tao, Y.J.; Krug, R.M.; Sawyer, S.L. Nuclear TRIM25 Specifically Targets Influenza Virus Ribonucleoproteins to Block the Onset of RNA Chain Elongation. Cell Host Microbe 2017, 22, 627–638. [Google Scholar] [CrossRef] [Green Version]
  197. Liu, B.; Li, N.L.; Shen, Y.; Bao, X.; Fabrizio, T.; Elbahesh, H.; Webby, R.J.; Li, K. The C-Terminal Tail of TRIM56 Dictates Antiviral Restriction of Influenza A and B Viruses by Impeding Viral RNA Synthesis. J. Virol. 2016, 90, 4369–4382. [Google Scholar] [CrossRef] [Green Version]
  198. Ma, J.; Sun, Q.; Mi, R.; Zhang, H. Avian Influenza A Virus H5N1 Causes Autophagy-Mediated Cell Death through Suppression of MTOR Signaling. J. Genet. Genom. 2011, 38, 533–537. [Google Scholar] [CrossRef] [PubMed]
  199. Beale, R.; Wise, H.; Stuart, A.; Ravenhill, B.J.; Digard, P.; Randow, F. A LC3-Interacting Motif in the Influenza A Virus M2 Protein Is Required to Subvert Autophagy and Maintain Virion Stability. Cell Host Microbe 2014, 15, 239–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Gannagé, M.; Dormann, D.; Albrecht, R.; Dengjel, J.; Torossi, T.; Rämer, P.C.; Lee, M.; Strowig, T.; Arrey, F.; Conenello, G.; et al. Matrix Protein 2 of Influenza A Virus Blocks Autophagosome Fusion with Lysosomes. Cell Host Microbe 2009, 6, 367–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Elmore, S. Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
  202. Galluzzi, L.; Brenner, C.; Morselli, E.; Touat, Z.; Kroemer, G. Viral Control of Mitochondrial Apoptosis. PLoS Pathog. 2008, 4, e1000018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Tallóczy, Z.; Jiang, W.; Virgin, H.W.; Leib, D.A.; Scheuner, D.; Kaufman, R.J.; Eskelinen, E.-L.; Levine, B. Regulation of Starvation- and Virus-Induced Autophagy by the EIF2alpha Kinase Signaling Pathway. Proc. Natl. Acad. Sci. USA 2002, 99, 190–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Liu, Y.; Wang, M.; Cheng, A.; Yang, Q.; Wu, Y.; Jia, R.; Liu, M.; Zhu, D.; Chen, S.; Zhang, S.; et al. The Role of Host EIF2α in Viral Infection. Virol. J. 2020, 17, 112. [Google Scholar] [CrossRef]
  205. Wu, X.; Wang, J.; Wang, S.; Wu, F.; Chen, Z.; Li, C.; Cheng, G.; Qin, F.X. Inhibition of Influenza A Virus Replication by TRIM14 via Its Multifaceted Protein-Protein Interaction with NP. Front. Microbiol. 2019, 10, 344. [Google Scholar] [CrossRef] [Green Version]
  206. Patil, G.; Zhao, M.; Song, K.; Hao, W.; Bouchereau, D.; Wang, L.; Li, S. TRIM41- Mediated Ubiquitination of Nucleoprotein Limits Influenza A Virus Infection. J. Virol. 2018, 92, e00905-18. [Google Scholar] [CrossRef] [Green Version]
  207. Pagani, I.; Di Pietro, A.; Oteiz, A.; Ghitti, M.; Mechti, N.; Naffakh, N.; Vicenzi, E. Mutations Conferring Increased Sensitivity to Tripartite Motif 22 Restriction Accumulated Progressively in the Nucleoprotein of Seasonal Influenza A (H1N1) Viruses between 1918 and 2009. MSphere 2018, 3, e00110-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Di Rienzo, M.; Romagnoli, A.; Antonioli, M.; Piacentini, M.; Fimia, G.M. TRIM Proteins in Autophagy: Selective Sensors in Cell Damage and Innate Immune Responses. Cell Death Differ. 2020, 27, 887–902. [Google Scholar] [CrossRef]
  209. Yang, B.; Wang, J.; Wang, Y.; Zhou, H.; Wu, X.; Tian, Z.; Sun, B. Novel Function of Trim44 Promotes an Antiviral Response by Stabilizing VISA. J. Immunol. 2013, 190, 3613–3619. [Google Scholar] [CrossRef] [Green Version]
  210. Krischuns, T.; Günl, F.; Henschel, L.; Binder, M.; Willemsen, J.; Schloer, S.; Rescher, U.; Gerlt, V.; Zimmer, G.; Nordhoff, C.; et al. Phosphorylation of TRIM28 Enhances the Expression of IFN-β and Proinflammatory Cytokines During HPAIV Infection of Human Lung Epithelial Cells. Front. Immunol. 2018, 9, 2229. [Google Scholar] [CrossRef] [Green Version]
  211. Fan, Y.; Mao, R.; Yu, Y.; Liu, S.; Shi, Z.; Cheng, J.; Zhang, H.; An, L.; Zhao, Y.; Xu, X.; et al. USP21 Negatively Regulates Antiviral Response by Acting as a RIG-I Deubiquitinase. J. Exp. Med. 2014, 211, 313–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Wang, L.; Zhao, W.; Zhang, M.; Wang, P.; Zhao, K.; Zhao, X.; Yang, S.; Gao, C. USP4 Positively Regulates RIG-I-Mediated Antiviral Response through Deubiquitination and Stabilization of RIG-I. J. Virol. 2013, 87, 4507–4515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Zhang, L.; Liu, J.; Qian, L.; Feng, Q.; Wang, X.; Yuan, Y.; Zuo, Y.; Cheng, Q.; Miao, Y.; Guo, T.; et al. Induction of OTUD1 by RNA Viruses Potently Inhibits Innate Immune Responses by Promoting Degradation of the MAVS/TRAF3/TRAF6 Signalosome. PLoS Pathog. 2018, 14, e1007067. [Google Scholar] [CrossRef] [Green Version]
  214. Jahan, A.S.; Biquand, E.; Muñoz-Moreno, R.; Le Quang, A.; Mok, C.K.-P.; Wong, H.H.; Teo, Q.W.; Valkenburg, S.A.; Chin, A.W.H.; Man Poon, L.L.; et al. OTUB1 Is a Key Regulator of RIG-I-Dependent Immune Signaling and Is Targeted for Proteasomal Degradation by Influenza A NS1. Cell Rep. 2020, 30, 1570–1584.e6. [Google Scholar] [CrossRef] [PubMed]
  215. Herhaus, L.; Al-Salihi, M.; Macartney, T.; Weidlich, S.; Sapkota, G.P. OTUB1 Enhances TGFβ Signalling by Inhibiting the Ubiquitylation and Degradation of Active SMAD2/3. Nat. Commun. 2013, 4, 2519. [Google Scholar] [CrossRef] [Green Version]
  216. Sun, X.-X.; Challagundla, K.B.; Dai, M.-S. Positive Regulation of P53 Stability and Activity by the Deubiquitinating Enzyme Otubain 1. EMBO J. 2012, 31, 576–592. [Google Scholar] [CrossRef] [PubMed]
  217. Rodgers, M.A.; Bowman, J.W.; Fujita, H.; Orazio, N.; Shi, M.; Liang, Q.; Amatya, R.; Kelly, T.J.; Iwai, K.; Ting, J.; et al. The Linear Ubiquitin Assembly Complex (LUBAC) Is Essential for NLRP3 Inflammasome Activation. J. Exp. Med. 2014, 211, 1333–1347. [Google Scholar] [CrossRef] [Green Version]
  218. Weng, L.; Mitoma, H.; Trichot, C.; Tricot, C.; Bao, M.; Liu, Y.; Zhang, Z.; Liu, Y.-J. The E3 Ubiquitin Ligase Tripartite Motif 33 Is Essential for Cytosolic RNA-Induced NLRP3 Inflammasome Activation. J. Immunol. 2014, 193, 3676–3682. [Google Scholar] [CrossRef] [Green Version]
  219. Py, B.F.; Kim, M.-S.; Vakifahmetoglu-Norberg, H.; Yuan, J. Deubiquitination of NLRP3 by BRCC3 Critically Regulates Inflammasome Activity. Mol. Cell 2013, 49, 331–338. [Google Scholar] [CrossRef] [Green Version]
  220. Schmidt, N.; Domingues, P.; Golebiowski, F.; Patzina, C.; Tatham, M.H.; Hay, R.T.; Hale, B.G. An Influenza Virus-Triggered SUMO Switch Orchestrates Co-Opted Endogenous Retroviruses to Stimulate Host Antiviral Immunity. Proc. Natl. Acad. Sci. USA 2019, 116, 17399–17408. [Google Scholar] [CrossRef] [Green Version]
  221. Morales, D.J.; Lenschow, D.J. The Antiviral Activities of ISG15. J. Mol. Biol. 2013, 425, 4995–5008. [Google Scholar] [CrossRef] [PubMed]
  222. Li, Y.; Chai, W.; Min, J.; Ye, Z.; Tong, X.; Qi, D.; Liu, W.; Luo, E.; Li, J.; Ye, X. Neddylation of M1 Negatively Regulates the Replication of Influenza A Virus. J. Gen. Virol. 2020, 101, 1242–1250. [Google Scholar] [CrossRef]
  223. Rowe, H.M.; Jakobsson, J.; Mesnard, D.; Rougemont, J.; Reynard, S.; Aktas, T.; Maillard, P.V.; Layard-Liesching, H.; Verp, S.; Marquis, J.; et al. KAP1 Controls Endogenous Retroviruses in Embryonic Stem Cells. Nature 2010, 463, 237–240. [Google Scholar] [CrossRef]
  224. Turelli, P.; Castro-Diaz, N.; Marzetta, F.; Kapopoulou, A.; Raclot, C.; Duc, J.; Tieng, V.; Quenneville, S.; Trono, D. Interplay of TRIM28 and DNA Methylation in Controlling Human Endogenous Retroelements. Genome Res. 2014, 24, 1260–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Matsui, T.; Leung, D.; Miyashita, H.; Maksakova, I.A.; Miyachi, H.; Kimura, H.; Tachibana, M.; Lorincz, M.C.; Shinkai, Y. Proviral Silencing in Embryonic Stem Cells Requires the Histone Methyltransferase ESET. Nature 2010, 464, 927–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Schultz, D.C.; Friedman, J.R.; Rauscher, F.J. Targeting Histone Deacetylase Complexes via KRAB-Zinc Finger Proteins: The PHD and Bromodomains of KAP-1 Form a Cooperative Unit That Recruits a Novel Isoform of the Mi-2α Subunit of NuRD. Genes Dev. 2001, 15, 428–443. [Google Scholar] [CrossRef] [Green Version]
  227. Ivanov, A.V.; Peng, H.; Yurchenko, V.; Yap, K.L.; Negorev, D.G.; Schultz, D.C.; Psulkowski, E.; Fredericks, W.J.; White, D.E.; Maul, G.G.; et al. PHD Domain-Mediated E3 Ligase Activity Directs Intramolecular Sumoylation of an Adjacent Bromodomain Which Is Required for Gene Silencing. Mol. Cell 2007, 28, 823–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Mascle, X.H.; Germain-Desprez, D.; Huynh, P.; Estephan, P.; Aubry, M. Sumoylation of the Transcriptional Intermediary Factor 1beta (TIF1beta), the Co-Repressor of the KRAB Multifinger Proteins, Is Required for Its Transcriptional Activity and Is Modulated by the KRAB Domain. J. Biol. Chem. 2007, 282, 10190–10202. [Google Scholar] [CrossRef] [Green Version]
  229. Roulois, D.; Yau, H.L.; Singhania, R.; Wang, Y.; Danesh, A.; Shen, S.Y.; Han, H.; Liang, G.; Pugh, T.J.; Jones, P.A.; et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell 2015, 162, 961–973. [Google Scholar] [CrossRef] [Green Version]
  230. Cuellar, T.L.; Herzner, A.-M.; Zhang, X.; Goyal, Y.; Watanabe, C.; Friedman, B.A.; Janakiraman, V.; Durinck, S.; Stinson, J.; Arnott, D.; et al. Silencing of Retrotransposons by SETDB1 Inhibits the Interferon Response in Acute Myeloid Leukemia. J. Cell Biol. 2017, 216, 3535–3549. [Google Scholar] [CrossRef] [Green Version]
  231. Chiappinelli, K.B.; Strissel, P.L.; Desrichard, A.; Li, H.; Henke, C.; Akman, B.; Hein, A.; Rote, N.S.; Cope, L.M.; Snyder, A.; et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via DsRNA Including Endogenous Retroviruses. Cell 2015, 162, 974–986. [Google Scholar] [CrossRef] [Green Version]
  232. Rajagopalan, D.; Tirado-Magallanes, R.; Bhatia, S.S.; Teo, W.S.; Sian, S.; Hora, S.; Lee, K.K.; Zhang, Y.; Jadhav, S.P.; Wu, Y.; et al. TIP60 Represses Activation of Endogenous Retroviral Elements. Nucleic. Acids. Res. 2018, 46, 9456–9470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Tie, C.H.; Fernandes, L.; Conde, L.; Robbez-Masson, L.; Sumner, R.P.; Peacock, T.; Rodriguez-Plata, M.T.; Mickute, G.; Gifford, R.; Towers, G.J.; et al. KAP1 Regulates Endogenous Retroviruses in Adult Human Cells and Contributes to Innate Immune Control. EMBO Rep. 2018, 19, e45000. [Google Scholar] [CrossRef]
  234. Liu, M.; Ohtani, H.; Zhou, W.; Ørskov, A.D.; Charlet, J.; Zhang, Y.W.; Shen, H.; Baylin, S.B.; Liang, G.; Grønbæk, K.; et al. Vitamin C Increases Viral Mimicry Induced by 5-Aza-2′-Deoxycytidine. Proc. Natl. Acad. Sci. USA 2016, 113, 10238–10244. [Google Scholar] [CrossRef] [Green Version]
  235. Zhao, C.; Sridharan, H.; Chen, R.; Baker, D.P.; Wang, S.; Krug, R.M. Influenza B Virus Non-Structural Protein 1 Counteracts ISG15 Antiviral Activity by Sequestering ISGylated Viral Proteins. Nat. Commun. 2016, 7, 12754. [Google Scholar] [CrossRef] [Green Version]
  236. Hsiang, T.-Y.; Zhao, C.; Krug, R.M. Interferon-Induced ISG15 Conjugation Inhibits Influenza A Virus Gene Expression and Replication in Human Cells. J. Virol. 2009, 83, 5971–5977. [Google Scholar] [CrossRef] [Green Version]
  237. Zhang, T.; Ye, Z.; Yang, X.; Qin, Y.; Hu, Y.; Tong, X.; Lai, W.; Ye, X. NEDDylation of PB2 Reduces Its Stability and Blocks the Replication of Influenza A Virus. Sci. Rep. 2017, 7, 43691. [Google Scholar] [CrossRef]
  238. Sun, H.; Yao, W.; Wang, K.; Qian, Y.; Chen, H.; Jung, Y.-S. Inhibition of Neddylation Pathway Represses Influenza Virus Replication and Pro-Inflammatory Responses. Virology 2018, 514, 230–239. [Google Scholar] [CrossRef] [PubMed]
  239. Liu, X.; Zhao, Z.; Xu, C.; Sun, L.; Chen, J.; Zhang, L.; Liu, W. Cyclophilin a Restricts Influenza a Virus Replication through Degradation of the M1 Protein. PLoS ONE 2012, 7, e31063. [Google Scholar] [CrossRef] [PubMed]
  240. Zinzula, L.; Tramontano, E. Strategies of Highly Pathogenic RNA Viruses to Block DsRNA Detection by RIG-I-like Receptors: Hide, Mask, Hit. Antiviral Res. 2013, 100, 615–635. [Google Scholar] [CrossRef]
  241. Liu, Y.; Olagnier, D.; Lin, R. Host and Viral Modulation of RIG-I-Mediated Antiviral Immunity. Front. Immunol. 2017, 7, 662. [Google Scholar] [CrossRef] [Green Version]
  242. Varga, Z.T.; Grant, A.; Manicassamy, B.; Palese, P. Influenza Virus Protein PB1-F2 Inhibits the Induction of Type I Interferon by Binding to MAVS and Decreasing Mitochondrial Membrane Potential. J. Virol. 2012, 86, 8359–8366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Graef, K.M.; Vreede, F.T.; Lau, Y.-F.; McCall, A.W.; Carr, S.M.; Subbarao, K.; Fodor, E. The PB2 Subunit of the Influenza Virus RNA Polymerase Affects Virulence by Interacting with the Mitochondrial Antiviral Signaling Protein and Inhibiting Expression of Beta Interferon. J. Virol. 2010, 84, 8433–8445. [Google Scholar] [CrossRef] [Green Version]
  244. Sharma, K.; Tripathi, S.; Ranjan, P.; Kumar, P.; Garten, R.; Deyde, V.; Katz, J.M.; Cox, N.J.; Lal, R.B.; Sambhara, S.; et al. Influenza A Virus Nucleoprotein Exploits Hsp40 to Inhibit PKR Activation. PLoS ONE 2011, 6, e20215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Xia, C.; Vijayan, M.; Pritzl, C.J.; Fuchs, S.Y.; McDermott, A.B.; Hahm, B. Hemagglutinin of influenza A virus antagonizes type I interferon (IFN) responses by inducing degradation of type I IFN receptor 1. J. Virol. 2016, 90, 2403–2417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Su, W.-C.; Yu, W.-Y.; Huang, S.-H.; Lai, M.M.C. Ubiquitination of the Cytoplasmic Domain of Influenza A Virus M2 Protein Is Crucial for Production of Infectious Virus Particles. J. Virol. 2018, 92, e01972-17. [Google Scholar] [CrossRef] [Green Version]
  247. Talon, J.; Horvath, C.M.; Polley, R.; Basler, C.F.; Muster, T.; Palese, P.; Garcia-Sastre, A. Activation of Interferon Regulatory Factor 3 Is Inhibited by the Influenza a Virus NS1 Protein. J. Virol. 2000, 74, 7989–7996. [Google Scholar] [CrossRef] [Green Version]
  248. Rückle, A.; Haasbach, E.; Julkunen, I.; Planz, O.; Ehrhardt, C.; Ludwig, S. The NS1 Protein of Influenza A Virus Blocks RIG-I-Mediated Activation of the Noncanonical NF-ΚB Pathway and P52/RelB-Dependent Gene Expression in Lung Epithelial Cells. J. Virol. 2012, 86, 10211–10217. [Google Scholar] [CrossRef] [Green Version]
  249. Wang, X.; Li, M.; Zheng, H.; Muster, T.; Palese, P.; Beg, A.A.; García-Sastre, A. Influenza A Virus NS1 Protein Prevents Activation of NF-ΚB and Induction of Alpha/Beta Interferon. J. Virol. 2000, 74, 11566. [Google Scholar] [CrossRef] [Green Version]
  250. Min, J.-Y.; Li, S.; Sen, G.C.; Krug, R.M. A Site on the Influenza A Virus NS1 Protein Mediates Both Inhibition of PKR Activation and Temporal Regulation of Viral RNA Synthesis. Virology 2007, 363, 236–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Min, J.-Y.; Krug, R.M. The Primary Function of RNA Binding by the Influenza A Virus NS1 Protein in Infected Cells: Inhibiting the 2’-5’ Oligo (A) Synthetase/RNase L Pathway. Proc. Natl. Acad. Sci. USA 2006, 103, 7100–7105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Marc, D. Influenza virus non-structural protein NS1: Interferon antagonism and beyond. J. Gen. Virol. 2014, 95, 2594–2611. [Google Scholar] [CrossRef]
  253. Carrillo, B.; Choi, J.-; Bornholdt, Z.A.; Sankaran, B.; Rice, A.P.; Venkataram Prasad, B.V. The influenza A virus protein NS1 displays structural polymorphism. J. Virol. 2014, 88, 4113–4122. [Google Scholar] [CrossRef] [Green Version]
  254. Ludwig, S.; Wang, X.; Ehrhardt, C.; Zheng, H.; Donelan, N.; Planz, O.; Pleschka, S.; García-Sastre, A.; Heins, G.; Wolff, T. The Influenza A Virus NS1 Protein Inhibits Activation of Jun N-Terminal Kinase and AP-1 Transcription Factors. J. Virol. 2002, 76, 11166–11171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Mibayashi, M.; Martínez-Sobrido, L.; Loo, Y.-M.; Cárdenas, W.B.; Gale, M.; García-Sastre, A. Inhibition of Retinoic Acid-Inducible Gene I-Mediated Induction of Beta Interferon by the NS1 Protein of Influenza A Virus. J. Virol. 2007, 81, 514–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Lamb, R.A.; Lai, C.-J. Sequence of Interrupted and Uninterrupted MRNAs and Cloned DNA Coding for the Two Overlapping Nonstructural Proteins of Influenza Virus. Cell 1980, 21, 475–485. [Google Scholar] [CrossRef]
  257. Robb, N.C.; Jackson, D.; Vreede, F.T.; Fodor, E. Splicing of Influenza A Virus NS1 MRNA Is Independent of the Viral NS1 Protein. J. Gen. Virol. 2010, 91, 2331–2340. [Google Scholar] [CrossRef] [PubMed]
  258. Dubois, J.; Terrier, O.; Rosa-Calatrava, M. Influenza Viruses and MRNA Splicing: Doing More with Less. MBio 2014, 5, e00070-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  259. Lohrmann, F.; Dijkman, R.; Stertz, S.; Thiel, V.; Haller, O.; Staeheli, P.; Kochs, G. Emergence of a C-Terminal Seven-Amino-Acid Elongation of NS1 in Around 1950 Conferred a Minor Growth Advantage to Former Seasonal Influenza A Viruses. J. Virol. 2013, 87, 11300–11303. [Google Scholar] [CrossRef] [Green Version]
  260. Chauché, C.; Nogales, A.; Zhu, H.; Goldfarb, D.; Ahmad Shanizza, A.I.; Gu, Q.; Parrish, C.R.; Martínez-Sobrido, L.; Marshall, J.F.; Murcia, P.R. Mammalian Adaptation of an Avian Influenza A Virus Involves Stepwise Changes in NS1. J. Virol. 2018, 92, e01875-17. [Google Scholar] [CrossRef] [Green Version]
  261. Chien, C.Y.; Tejero, R.; Huang, Y.; Zimmerman, D.E.; Ríos, C.B.; Krug, R.M.; Montelione, G.T. A Novel RNA-Binding Motif in Influenza A Virus Non-Structural Protein 1. Nat. Struct. Biol. 1997, 4, 891–895. [Google Scholar] [CrossRef] [PubMed]
  262. Liu, J.; Lynch, P.A.; Chien, C.-; Montelione, G.T.; Krug, R.M.; Berman, H.M. Crystal Structure of the Unique RNA-Binding Domain of the Influenza Virus NS1 Protein. Nat. Struct. Biol. 1997, 4, 896–899. [Google Scholar] [CrossRef] [PubMed]
  263. Wang, W.; Riedel, K.; Lynch, P.; Chien, C.; Montelione, G.T.; Krug, R.M. RNA Binding by the Novel Helical Domain of the Influenza Virus NS1 Protein Requires Its Dimer Structure and a Small Number of Specific Basic Amino Acids. RNA 1999, 5, 195–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Donelan, N.R.; Basler, C.F.; García-Sastre, A. A Recombinant Influenza A Virus Expressing an RNA-Binding-Defective NS1 Protein Induces High Levels of Beta Interferon and Is Attenuated in Mice. J. Virol. 2003, 77, 13257–13266. [Google Scholar] [CrossRef] [Green Version]
  265. Kerry, P.S.; Ayllon, J.; Taylor, M.A.; Hass, C.; Lewis, A.; García-Sastre, A.; Randall, R.E.; Hale, B.G.; Russell, R.J. A Transient Homotypic Interaction Model for the Influenza A Virus NS1 Protein Effector Domain. PLoS ONE 2011, 6, e17946. [Google Scholar] [CrossRef] [Green Version]
  266. Aramini, J.M.; Hamilton, K.; Ma, L.-; Swapna, G.V.T.; Leonard, P.G.; Ladbury, J.E.; Krug, R.M.; Montelione, G.T. F NMR Reveals Multiple Conformations at the Dimer Interface of the Nonstructural Protein 1 Effector Domain from Influenza A Virus. Structure 2014, 22, 515–525. [Google Scholar] [CrossRef] [Green Version]
  267. Aramini, J.M.; Ma, L.C.; Zhou, L.; Schauder, C.M.; Hamilton, K.; Amer, B.R.; Mack, T.R.; Lee, H.W.; Ciccosanti, C.T.; Zhao, L.; et al. Dimer Interface of the Effector Domain of Non-Structural Protein 1 from Influenza A Virus: An Interface with Multiple Functions. J. Biol. Chem. 2011, 286, 26050–26060. [Google Scholar] [CrossRef] [Green Version]
  268. Cheng, A.; Wong, S.M.; Yuan, Y.A. Structural basis for dsRNA recognition by NS1 protein of influenza A virus. Cell Res. 2009, 19, 187–195. [Google Scholar] [CrossRef]
  269. Greenspan, D.; Palese, P.; Krystal, M. Two Nuclear Location Signals in the Influenza Virus NS1 Nonstructural Protein. J. Virol. 1988, 62, 3020–3026. [Google Scholar] [CrossRef] [Green Version]
  270. Melén, K.; Kinnunen, L.; Fagerlund, R.; Ikonen, N.; Twu, K.Y.; Krug, R.M.; Julkunen, I. Nuclear and Nucleolar Targeting of Influenza A Virus NS1 Protein: Striking Differences between Different Virus Subtypes. J. Virol. 2007, 81, 5995–6006. [Google Scholar] [CrossRef] [Green Version]
  271. Li, Y.; Yamakita, Y.; Krug, R.M. Regulation of a Nuclear Export Signal by an Adjacent Inhibitory Sequence: The Effector Domain of the Influenza Virus NS1 Protein. Proc. Natl. Acad. Sci. USA 1998, 95, 4864–4869. [Google Scholar] [CrossRef] [Green Version]
  272. Hale, B.G.; Randall, R.E.; Ortín, J.; Jackson, D. The Multifunctional NS1 Protein of Influenza A Viruses. J. Gen. Virol. 2008, 89, 2359–2376. [Google Scholar] [CrossRef]
  273. Hale, B.G. Conformational Plasticity of the Influenza A Virus NS1 Protein. J. Gen. Virol. 2014, 95, 2099–2105. [Google Scholar] [CrossRef] [PubMed]
  274. Jackson, D.; Hossain, M.J.; Hickman, D.; Perez, D.R.; Lamb, R.A. A New Influenza Virus Virulence Determinant: The NS1 Protein Four C-Terminal Residues Modulate Pathogenicity. Proc. Natl. Acad. Sci. USA 2008, 105, 4381–4386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Zhao, N.; Sebastiano, V.; Moshkina, N.; Mena, N.; Hultquist, J.; Jimenez-Morales, D.; Ma, Y.; Rialdi, A.; Albrecht, R.; Fenouil, R.; et al. Influenza Virus Infection Causes Global RNAPII Termination Defects. Nat. Struct. Mol. Biol. 2018, 25, 885–893. [Google Scholar] [CrossRef] [PubMed]
  276. Klemm, C.; Boergeling, Y.; Ludwig, S.; Ehrhardt, C. Immunomodulatory Nonstructural Proteins of Influenza A Viruses. Trends Microbiol. 2018, 26, 624–636. [Google Scholar] [CrossRef] [PubMed]
  277. García-Sastre, A.; Egorov, A.; Matassov, D.; Brandt, S.; Levy, D.E.; Durbin, J.E.; Palese, P.; Muster, T. Influenza A Virus Lacking the NS1 Gene Replicates in Interferon-Deficient Systems. Virology 1998, 252, 324–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Kochs, G.; Koerner, I.; Thiel, L.; Kothlow, S.; Kaspers, B.; Ruggli, N.; Summerfield, A.; Pavlovic, J.; Stech, J.; Staeheli, P. Properties of H7N7 Influenza A Virus Strain SC35M Lacking Interferon Antagonist NS1 in Mice and Chickens. J. Gen. Virol. 2007, 88, 1403–1409. [Google Scholar] [CrossRef]
  279. Nogales, A.; Chauché, C.; DeDiego, M.L.; Topham, D.J.; Parrish, C.R.; Murcia, P.R.; Martínez-Sobrido, L. The K186E Amino Acid Substitution in the Canine Influenza Virus H3N8 NS1 Protein Restores Its Ability To Inhibit Host Gene Expression. J. Virol. 2017, 91, e00877-17. [Google Scholar] [CrossRef] [Green Version]
  280. Nogales, A.; Baker, S.F.; Ortiz-Riaño, E.; Dewhurst, S.; Topham, D.J.; Martínez-Sobrido, L. Influenza A Virus Attenuation by Codon Deoptimization of the NS Gene for Vaccine Development. J. Virol. 2014, 88, 10525–10540. [Google Scholar] [CrossRef] [Green Version]
  281. DeDiego, M.L.; Nogales, A.; Lambert-Emo, K.; Martinez-Sobrido, L.; Topham, D.J. NS1 Protein Mutation I64T Affects Interferon Responses and Virulence of Circulating H3N2 Human Influenza A Viruses. J. Virol. 2016, 90, 9693–9711. [Google Scholar] [CrossRef] [Green Version]
  282. Nogales, A.; Martinez-Sobrido, L.; Topham, D.J.; DeDiego, M.L. NS1 Protein Amino Acid Changes D189N and V194I Affect Interferon Responses, Thermosensitivity, and Virulence of Circulating H3N2 Human Influenza A Viruses. J. Virol. 2017, 91, e01930-16. [Google Scholar] [CrossRef] [Green Version]
  283. Rajsbaum, R.; Albrecht, R.A.; Wang, M.K.; Maharaj, N.P.; Versteeg, G.A.; Nistal-Villán, E.; García-Sastre, A.; Gack, M.U. Species-Specific Inhibition of RIG-I Ubiquitination and IFN Induction by the Influenza A Virus NS1 Protein. PLoS Pathog. 2012, 8, e1003059. [Google Scholar] [CrossRef]
  284. Qian, W.; Wei, X.; Guo, K.; Li, Y.; Lin, X.; Zou, Z.; Zhou, H.; Jin, M. The C-Terminal Effector Domain of Non-Structural Protein 1 of Influenza A Virus Blocks IFN-β Production by Targeting TNF Receptor-Associated Factor 3. Front. Immunol. 2017, 8, 779. [Google Scholar] [CrossRef] [PubMed]
  285. Gao, S.; Song, L.; Li, J.; Zhang, Z.; Peng, H.; Jiang, W.; Wang, Q.; Kang, T.; Chen, S.; Huang, W. Influenza A Virus-Encoded NS1 Virulence Factor Protein Inhibits Innate Immune Response by Targeting IKK. Cell. Microbiol. 2012, 14, 1849–1866. [Google Scholar] [CrossRef]
  286. Newby, C.M.; Sabin, L.; Pekosz, A. The RNA Binding Domain of Influenza A Virus NS1 Protein Affects Secretion of Tumor Necrosis Factor Alpha, Interleukin-6, and Interferon in Primary Murine Tracheal Epithelial Cells. J. Virol. 2007, 81, 9469–9480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Liu, S.; Liu, L.; Xu, G.; Cao, Z.; Wang, Q.; Li, S.; Peng, N.; Yin, J.; Yu, H.; Li, M.; et al. Epigenetic Modification Is Regulated by the Interaction of Influenza A Virus Nonstructural Protein 1 with the De Novo DNA Methyltransferase DNMT3B and Subsequent Transport to the Cytoplasm for K48-Linked Polyubiquitination. J. Virol. 2019, 93, e01587-18. [Google Scholar] [CrossRef] [Green Version]
  288. Nogales, A.; Martinez-Sobrido, L.; Topham, D.; DeDiego, M. Modulation of Innate Immune Responses by the Influenza A NS1 and PA-X Proteins. Viruses 2018, 10, 708. [Google Scholar] [CrossRef] [Green Version]
  289. Wang, B.X.; Fish, E.N. Interactions Between NS1 of Influenza A Viruses and Interferon-α/β: Determinants for Vaccine Development. J. Interferon Cytokine Res. 2017, 37, 331–341. [Google Scholar] [CrossRef] [PubMed]
  290. Krug, R.M. Functions of the Influenza A Virus NS1 Protein In Antiviral Defense. Curr. Opin. Virol. 2015, 12, 1–6. [Google Scholar] [CrossRef] [Green Version]
  291. Cheong, W.-C.; Kang, H.-R.; Yoon, H.; Kang, S.-J.; Ting, J.P.-Y.; Song, M.J. Influenza A Virus NS1 Protein Inhibits the NLRP3 Inflammasome. PLoS ONE 2015, 10, e0126456. [Google Scholar] [CrossRef]
  292. Park, H.-S.; Liu, G.; Thulasi Raman, S.N.; Landreth, S.L.; Liu, Q.; Zhou, Y. NS1 Protein of 2009 Pandemic Influenza A Virus Inhibits Porcine NLRP3 Inflammasome-Mediated Interleukin-1 Beta Production by Suppressing ASC Ubiquitination. J. Virol. 2018, 92, e00022-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  293. Ehrhardt, C.; Ludwig, S. A New Player in a Deadly Game: Influenza Viruses and the PI3K/Akt Signalling Pathway. Cell Microbiol. 2009, 11, 863–871. [Google Scholar] [CrossRef] [PubMed]
  294. Shin, Y.-K.; Liu, Q.; Tikoo, S.K.; Babiuk, L.A.; Zhou, Y. Influenza A Virus NS1 Protein Activates the Phosphatidylinositol 3-Kinase (PI3K)/Akt Pathway by Direct Interaction with the P85 Subunit of PI3K. J. Gen. Virol. 2007, 88, 13–18. [Google Scholar] [CrossRef]
  295. Ponnuswamy, A.; Hupp, T.; Fåhraeus, R. Concepts in MDM2 Signaling. Genes Cancer 2012, 3, 291–297. [Google Scholar] [CrossRef] [Green Version]
  296. Hale, B.G.; Jackson, D.; Chen, Y.-H.; Lamb, R.A.; Randall, R.E. Influenza A Virus NS1 Protein Binds P85β and Activates Phosphatidylinositol-3-Kinase Signaling. Proc. Natl. Acad. Sci. USA 2006, 103, 14194–14199. [Google Scholar] [CrossRef] [Green Version]
  297. Hale, B.G.; Kerry, P.S.; Jackson, D.; Precious, B.L.; Gray, A.; Killip, M.J.; Randall, R.E.; Russell, R.J. Structural Insights into Phosphoinositide 3-Kinase Activation by the Influenza A Virus NS1 Protein. Proc. Natl. Acad. Sci. USA 2010, 107, 1954–1959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. Lopes, A.M.; Domingues, P.; Zell, R.; Hale, B.G. Structure-Guided Functional Annotation of the Influenza A Virus NS1 Protein Reveals Dynamic Evolution of the P85β-Binding Site during Circulation in Humans. J. Virol. 2017, 91, e01081-17. [Google Scholar] [CrossRef] [Green Version]
  299. Ehrhardt, C.; Wolff, T.; Pleschka, S.; Planz, O.; Beermann, W.; Bode, J.G.; Schmolke, M.; Ludwig, S. Influenza A Virus NS1 Protein Activates the PI3K/Akt Pathway To Mediate Antiapoptotic Signaling Responses. J. Virol. 2007, 81, 3058–3067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Hrincius, E.R.; Dierkes, R.; Anhlan, D.; Wixler, V.; Ludwig, S.; Ehrhardt, C. Phosphatidylinositol-3-Kinase (PI3K) Is Activated by Influenza Virus VRNA via the Pathogen Pattern Receptor Rig-I to Promote Efficient Type I Interferon Production. Cell. Microbiol. 2011, 13, 1907–1919. [Google Scholar] [CrossRef]
  301. Lu, X.; Masic, A.; Liu, Q.; Zhou, Y. Regulation of Influenza A Virus Induced CXCL-10 Gene Expression Requires PI3K/Akt Pathway and IRF3 Transcription Factor. Mol. Immunol. 2011, 48, 1417–1423. [Google Scholar] [CrossRef]
  302. Ampomah, P.B.; Lim, L.H.K. Influenza A Virus-Induced Apoptosis and Virus Propagation. Apoptosis 2020, 25, 1–11. [Google Scholar] [CrossRef] [PubMed]
  303. Wurzer, W.J.; Planz, O.; Ehrhardt, C.; Giner, M.; Silberzahn, T.; Pleschka, S.; Ludwig, S. Caspase 3 Activation Is Essential for Efficient Influenza Virus Propagation. EMBO J. 2003, 22, 2717–2728. [Google Scholar] [CrossRef] [Green Version]
  304. Das, K.; Ma, L.-C.; Xiao, R.; Radvansky, B.; Aramini, J.; Zhao, L.; Marklund, J.; Kuo, R.-L.; Twu, K.Y.; Arnold, E.; et al. Structural Basis for Suppression of a Host Antiviral Response by Influenza A Virus. Proc. Natl. Acad. Sci. USA 2008, 105, 13093–13098. [Google Scholar] [CrossRef] [Green Version]
  305. Noah, D.L.; Twu, K.Y.; Krug, R.M. Cellular Antiviral Responses against Influenza A Virus Are Countered at the Posttranscriptional Level by the Viral NS1A Protein via Its Binding to a Cellular Protein Required for the 3′ End Processing of Cellular Pre-MRNAS. Virology 2003, 307, 386–395. [Google Scholar] [CrossRef]
  306. Nemeroff, M.E.; Barabino, S.M.L.; Li, Y.; Keller, W.; Krug, R.M. Influenza Virus NS1 Protein Interacts with the Cellular 30 KDa Subunit of CPSF and Inhibits 3′ End Formation of Cellular Pre-MRNAs. Mol. Cell 1998, 1, 991–1000. [Google Scholar] [CrossRef]
  307. Levene, R.E.; Gaglia, M.M. Host Shutoff in Influenza A Virus: Many Means to an End. Viruses 2018, 10, 475. [Google Scholar] [CrossRef] [Green Version]
  308. Chan, S.; Choi, E.-A.; Shi, Y. Pre-MRNA 3′-End Processing Complex Assembly and Function. WIREs RNA 2011, 2, 321–335. [Google Scholar] [CrossRef] [Green Version]
  309. Chen, Z. Influenza A Virus NS1 Protein Targets Poly(A)-Binding Protein II of the Cellular 3′-End Processing Machinery. EMBO J. 1999, 18, 2273–2283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  310. Anastasina, M.; Le May, N.; Bugai, A.; Fu, Y.; Söderholm, S.; Gaelings, L.; Ohman, T.; Tynell, J.; Kyttänen, S.; Barboric, M.; et al. Influenza Virus NS1 Protein Binds Cellular DNA to Block Transcription of Antiviral Genes. Biochim. Biophys. Acta Gene Regul. Mech. 2016, 1859, 1440–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  311. Satterly, N.; Tsai, P.-L.; van Deursen, J.; Nussenzveig, D.R.; Wang, Y.; Faria, P.A.; Levay, A.; Levy, D.E.; Fontoura, B.M.A. Influenza Virus Targets the MRNA Export Machinery and the Nuclear Pore Complex. Proc. Natl. Acad. Sci. USA 2007, 104, 1853–1858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  312. Aragón, T. Eukaryotic Translation Initiation Factor 4GI Is a Cellular Target for NS1 Protein, a Translational Activator of Influenza Virus. Mol. Cell. Biol. 2000, 20, 10. [Google Scholar] [CrossRef] [PubMed]
  313. Guo, Z.; Chen, L.; Zeng, H.; Gomez, J.A.; Plowden, J.; Fujita, T.; Katz, J.M.; Donis, R.O.; Sambhara, S. NS1 Protein of Influenza A Virus Inhibits the Function of Intracytoplasmic Pathogen Sensor, RIG-I. Am. J. Respir Cell Mol. Biol. 2007, 36, 263–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Opitz, B.; Rejaibi, A.; Dauber, B.; Eckhard, J.; Vinzing, M.; Schmeck, B.; Hippenstiel, S.; Suttorp, N.; Wolff, T. IFNβ Induction by Influenza A Virus Is Mediated by RIG-I Which Is Regulated by the Viral NS1 Protein. Cell. Microbiol. 2007, 9, 930–938. [Google Scholar] [CrossRef]
  315. Jureka, A.S.; Kleinpeter, A.B.; Tipper, J.L.; Harrod, K.S.; Petit, C.M. The Influenza NS1 Protein Modulates RIG-I Activation via a Strain-Specific Direct Interaction with the Second CARD of RIG-I. J. Biol. Chem. 2020, 295, 1153–1164. [Google Scholar] [CrossRef]
  316. Jureka, A.S.; Kleinpeter, A.B.; Cornilescu, G.; Cornilescu, C.C.; Petit, C.M. Structural Basis for a Novel Interaction between the NS1 Protein Derived from the 1918 Influenza Virus and RIG-I. Structure 2015, 23, 2001–2010. [Google Scholar] [CrossRef] [Green Version]
  317. Kuo, R.-L.; Li, L.-H.; Lin, S.-J.; Li, Z.-H.; Chen, G.-W.; Chang, C.-K.; Wang, Y.-R.; Tam, E.-H.; Gong, Y.-N.; Krug, R.M.; et al. Role of N Terminus-Truncated NS1 Proteins of Influenza A Virus in Inhibiting IRF3 Activation. J. Virol. 2016, 90, 4696–4705. [Google Scholar] [CrossRef] [Green Version]
  318. Kuo, R.-L.; Zhao, C.; Malur, M.; Krug, R.M. Influenza A Virus Strains That Circulate in Humans Differ in the Ability of Their NS1 Proteins to Block the Activation of IRF3 and Interferon-β Transcription. Virology 2010, 408, 146–158. [Google Scholar] [CrossRef]
  319. Marcos-Villar, L.; Nistal-Villan, E.; Zamarreño, N.; Garaigorta, U.; Gastaminza, P.; Nieto, A. Interferon-β Stimulation Elicited by the Influenza Virus Is Regulated by the Histone Methylase Dot1L through the RIG-I-TRIM25 Signaling Axis. Cells 2020, 9, 732. [Google Scholar] [CrossRef] [Green Version]
  320. Catrysse, L.; Vereecke, L.; Beyaert, R.; van Loo, G. A20 in Inflammation and Autoimmunity. Trends Immunol. 2014, 35, 22–31. [Google Scholar] [CrossRef]
  321. Maelfait, J.; Roose, K.; Vereecke, L.; Guire, C.M.; Sze, M.; Schuijs, M.J.; Willart, M.; Ibañez, L.I.; Hammad, H.; Lambrecht, B.N.; et al. A20 Deficiency in Lung Epithelial Cells Protects against Influenza A Virus Infection. PLoS Pathog. 2016, 12, e1005410. [Google Scholar] [CrossRef] [Green Version]
  322. Arguello, M.; Paz, S.; Ferran, C.; Moll, H.P.; Hiscott, J. Anti-Viral Tetris: Modulation of the Innate Anti-Viral Immune Response by A20. Adv. Exp. Med. Biol. 2014, 809, 49–64. [Google Scholar] [CrossRef] [PubMed]
  323. Wertz, I.E.; O’Rourke, K.M.; Zhou, H.; Eby, M.; Aravind, L.; Seshagiri, S.; Wu, P.; Wiesmann, C.; Baker, R.; Boone, D.L.; et al. De-Ubiquitination and Ubiquitin Ligase Domains of A20 Downregulate NF-KappaB Signalling. Nature 2004, 430, 694–699. [Google Scholar] [CrossRef]
  324. Wang, Y.-Y.; Li, L.; Han, K.-J.; Zhai, Z.; Shu, H.-B. A20 Is a Potent Inhibitor of TLR3- and Sendai Virus-Induced Activation of NF-ΚB and ISRE and IFN-β Promoter. FEBS Lett. 2004, 576, 86–90. [Google Scholar] [CrossRef] [Green Version]
  325. Lane, D.; Levine, A. P53 Research: The Past Thirty Years and the Next Thirty Years. Cold Spring Harb. Perspect. Biol. 2010, 2, a000893. [Google Scholar] [CrossRef] [Green Version]
  326. Bieging, K.T.; Mello, S.S.; Attardi, L.D. Unravelling Mechanisms of P53-Mediated Tumour Suppression. Nat. Rev. Cancer 2014, 14, 359–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Turpin, E.; Luke, K.; Jones, J.; Tumpey, T.; Konan, K.; Schultz-Cherry, S. Influenza Virus Infection Increases P53 Activity: Role of P53 in Cell Death and Viral Replication. J. Virol. 2005, 79, 8802–8811. [Google Scholar] [CrossRef] [Green Version]
  328. Zhirnov, O.P.; Klenk, H.-D. Control of Apoptosis in Influenza Virus-Infected Cells by up-Regulation of Akt and P53 Signaling. Apoptosis 2007, 12, 1419–1432. [Google Scholar] [CrossRef] [PubMed]
  329. Shen, Y.; Wang, X.; Guo, L.; Qiu, Y.; Li, X.; Yu, H.; Xiang, H.; Tong, G.; Ma, Z. Influenza A Virus Induces P53 Accumulation in a Biphasic Pattern. Biochem. Biophys Res. Commun. 2009, 382, 331–335. [Google Scholar] [CrossRef]
  330. Terrier, O.; Josset, L.; Textoris, J.; Marcel, V.; Cartet, G.; Ferraris, O.; N’Guyen, C.; Lina, B.; Diaz, J.-J.; Bourdon, J.-C.; et al. Cellular Transcriptional Profiling in Human Lung Epithelial Cells Infected by Different Subtypes of Influenza A Viruses Reveals an Overall Down-Regulation of the Host P53 Pathway. Virol. J. 2011, 8, 285. [Google Scholar] [CrossRef] [Green Version]
  331. Muñoz-Fontela, C.; Mandinova, A.; Aaronson, S.A.; Lee, S.W. Emerging Roles of P53 and Other Tumour-Suppressor Genes in Immune Regulation. Nat. Rev. Immunol. 2016, 16, 741–750. [Google Scholar] [CrossRef] [Green Version]
  332. Lazo, P.A.; Santos, C.R. Interference with P53 Functions in Human Viral Infections, a Target for Novel Antiviral Strategies? Rev. Med. Virol. 2011, 21, 285–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  333. Terrier, O.; Diederichs, A.; Dubois, J.; Cartet, G.; Lina, B.; Bourdon, J.-C.; Rosa-Calatrava, M. Influenza NS1 Interacts with P53 and Alters Its Binding to P53-Responsive Genes, in a Promoter-Dependent Manner. FEBS Lett. 2013, 587, 2965–2971. [Google Scholar] [CrossRef] [Green Version]
  334. Terrier, O.; Marcel, V.; Cartet, G.; Lane, D.P.; Lina, B.; Rosa-Calatrava, M.; Bourdon, J.-C. Influenza A Viruses Control Expression of Proviral Human P53 Isoforms P53β and Δ133p53α. J. Virol. 2012, 86, 8452–8460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Wang, X.; Deng, X.; Yan, W.; Zhu, Z.; Shen, Y.; Qiu, Y.; Shi, Z.; Shao, D.; Wei, J.; Xia, X.; et al. Stabilization of P53 in Influenza A Virus-Infected Cells Is Associated with Compromised MDM2-Mediated Ubiquitination of P53. J. Biol. Chem. 2012, 287, 18366–18375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Pizzorno, A.; Dubois, J.; Machado, D.; Cartet, G.; Traversier, A.; Julien, T.; Lina, B.; Bourdon, J.-C.; Rosa-Calatrava, M.; Terrier, O. Influenza A Viruses Alter the Stability and Antiviral Contribution of Host E3-Ubiquitin Ligase Mdm2 during the Time-Course of Infection. Sci. Rep. 2018, 8, 3746. [Google Scholar] [CrossRef]
  337. Bohlman, S.; Manfredi, J.J. P53-Independent Effects of Mdm2. Subcell Biochem. 2014, 85, 235–246. [Google Scholar] [CrossRef] [Green Version]
  338. Thomasova, D.; Mulay, S.R.; Bruns, H.; Anders, H.-J. P53-Independent Roles of MDM2 in NF-ΚB Signaling: Implications for Cancer Therapy, Wound Healing, and Autoimmune Diseases. Neoplasia 2012, 14, 1097–1101. [Google Scholar] [CrossRef] [Green Version]
  339. Xu, K.; Klenk, C.; Liu, B.; Keiner, B.; Cheng, J.; Zheng, B.-J.; Li, L.; Han, Q.; Wang, C.; Li, T.; et al. Modification of Nonstructural Protein 1 of Influenza A Virus by SUMO1. J. Virol. 2011, 85, 1086–1098. [Google Scholar] [CrossRef] [Green Version]
  340. Santos, A.; Pal, S.; Chacon, J.; Meraz, K.; Gonzalez, J.; Prieto, K.; Rosas-Acosta, G. SUMOylation Affects the Interferon Blocking Activity of the Influenza A Nonstructural Protein NS1 without Affecting Its Stability or Cellular Localization. J. Virol. 2013, 87, 5602–5620. [Google Scholar] [CrossRef] [Green Version]
  341. Way, G.; Xiong, Z.; Wang, G.; Dai, H.; Zheng, S.; García-Sastre, A.; Liao, J. A Novel SUMOylation Site in the Influenza a Virus NS1 Protein Identified with a Highly Sensitive FRET Assay. J. Biotechnol. 2020, 323, 121–127. [Google Scholar] [CrossRef]
  342. Jiang, J.; Li, J.; Fan, W.; Zheng, W.; Yu, M.; Chen, C.; Sun, L.; Bi, Y.; Ding, C.; Gao, G.F.; et al. Robust Lys63-Linked Ubiquitination of RIG-I Promotes Cytokine Eruption in Early Influenza B Virus Infection. J. Virol. 2016, 90, 6263–6275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Versteeg, G.A.; Hale, B.G.; van Boheemen, S.; Wolff, T.; Lenschow, D.J.; García-Sastre, A. Species-Specific Antagonism of Host ISGylation by the Influenza B Virus NS1 Protein. J. Virol. 2010, 84, 5423–5430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Guan, R.; Ma, L.-C.; Leonard, P.G.; Amer, B.R.; Sridharan, H.; Zhao, C.; Krug, R.M.; Montelione, G.T. Structural Basis for the Sequence-Specific Recognition of Human ISG15 by the NS1 Protein of Influenza B Virus. Proc. Natl. Acad. Sci. USA 2011, 108, 13468–13473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Ubiquitin-proteasome system. Ubiquitin (Ub) is first attached to the ubiquitin-activating enzyme E1 in an ATP-dependent way. Ubiquitin is then transferred to the ubiquitin-conjugating enzyme, E2. An isopeptide bound is finally catalyzed between the ubiquitin and a lysine (K) residue on a protein substrate by the ubiquitin ligase, E3. The ubiquitinated protein can subsequently be poly-ubiquitinated through K residues within ubiquitin itself. Depending on the linkage type, the protein will then undergo different fates in various cellular pathways. De-ubiquitinase enzymes (DUB) can recognize and hydrolyze isopeptide bonds formed between ubiquitin and protein substrate and between ubiquitin moieties, leading to the recycling of ubiquitin. K48-linked poly-ubiquitination leads to targeted protein degradation by the 26S proteasome, releasing free ubiquitins, which are reincorporated in the loop or used in other pathways. For each category of enzymes (E1, E2, E3 and DUB), the number of proteins in humans are indicated (source: IUUCD 2.0 database).
Figure 1. Ubiquitin-proteasome system. Ubiquitin (Ub) is first attached to the ubiquitin-activating enzyme E1 in an ATP-dependent way. Ubiquitin is then transferred to the ubiquitin-conjugating enzyme, E2. An isopeptide bound is finally catalyzed between the ubiquitin and a lysine (K) residue on a protein substrate by the ubiquitin ligase, E3. The ubiquitinated protein can subsequently be poly-ubiquitinated through K residues within ubiquitin itself. Depending on the linkage type, the protein will then undergo different fates in various cellular pathways. De-ubiquitinase enzymes (DUB) can recognize and hydrolyze isopeptide bonds formed between ubiquitin and protein substrate and between ubiquitin moieties, leading to the recycling of ubiquitin. K48-linked poly-ubiquitination leads to targeted protein degradation by the 26S proteasome, releasing free ubiquitins, which are reincorporated in the loop or used in other pathways. For each category of enzymes (E1, E2, E3 and DUB), the number of proteins in humans are indicated (source: IUUCD 2.0 database).
Viruses 13 02309 g001
Figure 2. Innate immune response during influenza A virus infection and NS1 hijackings. IFNs activation (left): viral particles are incorporated in the host cell through endosomes formation, leading to vRNPs’ release in the cytoplasm. The RLR sensor RIG-I is then activated by their presence and is K63 poly-ubiquitinated by Riplet and by TRIM25. The DUB OTUB1 also activates RIG-I through inhibition of its K48-linked poly-ubiquitination. In turn, RIG-I activates the NLRP3 inflammasome, that stimulates caspase-1 activation. Caspase-1 then mediates the maturation of interleukins IL-1β and IL-18, which are then secreted to promote inflammatory response. After its activation, RIG-I mainly binds to MAVS and promotes its multimerization at the mitochondrial membrane. Through signaling cascades not depicted here, MAVS mediates the recruitment of TBK1 and IKK kinases, which phosphorylate IRF3 and IRF7 transcription factors, and the NF-κB inhibitor IκB, respectively. IκB is then poly-ubiquitinated and degraded by the 26S proteasome. Activated IRF3 and IRF7, as well as activated NF-κB then translocate into the nucleus to promote type I IFNs (IFNα et IFNβ) expression, and pro-inflammatory cytokines, respectively. Notably, NF-κB also mediates IFNβ expression. ISGs activation (right): type I IFNs are then secreted in an autocrine and a paracrine way and bind to IFNAR1/2 heterodimeric receptor. Upon IFN binding, JAK1 and TYK2 kinases are activated and mediate phosphorylation of STAT1 and STAT2 transcription factors. These proteins then translocate into the nucleus with IRF9 and bind to ISRE sequences in ISG promoters to induce their transcription. Several ISGs are known to particularly counteract IAV replication (examples in the orange box). In turn, the NS1 protein hijacks innate immune response by targeting different proteins (bold black arrows), and notably ubiquitin factors (green).
Figure 2. Innate immune response during influenza A virus infection and NS1 hijackings. IFNs activation (left): viral particles are incorporated in the host cell through endosomes formation, leading to vRNPs’ release in the cytoplasm. The RLR sensor RIG-I is then activated by their presence and is K63 poly-ubiquitinated by Riplet and by TRIM25. The DUB OTUB1 also activates RIG-I through inhibition of its K48-linked poly-ubiquitination. In turn, RIG-I activates the NLRP3 inflammasome, that stimulates caspase-1 activation. Caspase-1 then mediates the maturation of interleukins IL-1β and IL-18, which are then secreted to promote inflammatory response. After its activation, RIG-I mainly binds to MAVS and promotes its multimerization at the mitochondrial membrane. Through signaling cascades not depicted here, MAVS mediates the recruitment of TBK1 and IKK kinases, which phosphorylate IRF3 and IRF7 transcription factors, and the NF-κB inhibitor IκB, respectively. IκB is then poly-ubiquitinated and degraded by the 26S proteasome. Activated IRF3 and IRF7, as well as activated NF-κB then translocate into the nucleus to promote type I IFNs (IFNα et IFNβ) expression, and pro-inflammatory cytokines, respectively. Notably, NF-κB also mediates IFNβ expression. ISGs activation (right): type I IFNs are then secreted in an autocrine and a paracrine way and bind to IFNAR1/2 heterodimeric receptor. Upon IFN binding, JAK1 and TYK2 kinases are activated and mediate phosphorylation of STAT1 and STAT2 transcription factors. These proteins then translocate into the nucleus with IRF9 and bind to ISRE sequences in ISG promoters to induce their transcription. Several ISGs are known to particularly counteract IAV replication (examples in the orange box). In turn, the NS1 protein hijacks innate immune response by targeting different proteins (bold black arrows), and notably ubiquitin factors (green).
Viruses 13 02309 g002
Figure 3. Schematic representation of TRIM25, RIG-I and NS1 protein structures and interactions. Upon infection, TRIM25 CCD first mediates RING domain dimerization (1) and subsequent complete TRIM25 dimerization, allowing interaction between TRIM25 PRY-SPRY domain and RIG-I first CARD (2). TRIM25 RING domain then mediates K63-linked poly-ubiquitination of the lysine 172 (K172) on RIG-I second CARD (3), leading to antiviral response activation. In turn, IAV NS1 protein can inhibit these two proteins (red arrows) (4). NS1 RBD from 1918 H1N1 strain directly binds to and inhibits poly-ubiquitination of RIG-I second CARD, while amino acids 96 and 97 in ED interact with TRIM25 CCD, therefore inhibiting TRIM25 dimerization.
Figure 3. Schematic representation of TRIM25, RIG-I and NS1 protein structures and interactions. Upon infection, TRIM25 CCD first mediates RING domain dimerization (1) and subsequent complete TRIM25 dimerization, allowing interaction between TRIM25 PRY-SPRY domain and RIG-I first CARD (2). TRIM25 RING domain then mediates K63-linked poly-ubiquitination of the lysine 172 (K172) on RIG-I second CARD (3), leading to antiviral response activation. In turn, IAV NS1 protein can inhibit these two proteins (red arrows) (4). NS1 RBD from 1918 H1N1 strain directly binds to and inhibits poly-ubiquitination of RIG-I second CARD, while amino acids 96 and 97 in ED interact with TRIM25 CCD, therefore inhibiting TRIM25 dimerization.
Viruses 13 02309 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lamotte, L.-A.; Tafforeau, L. How Influenza A Virus NS1 Deals with the Ubiquitin System to Evade Innate Immunity. Viruses 2021, 13, 2309. https://doi.org/10.3390/v13112309

AMA Style

Lamotte L-A, Tafforeau L. How Influenza A Virus NS1 Deals with the Ubiquitin System to Evade Innate Immunity. Viruses. 2021; 13(11):2309. https://doi.org/10.3390/v13112309

Chicago/Turabian Style

Lamotte, Laurie-Anne, and Lionel Tafforeau. 2021. "How Influenza A Virus NS1 Deals with the Ubiquitin System to Evade Innate Immunity" Viruses 13, no. 11: 2309. https://doi.org/10.3390/v13112309

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