Next Article in Journal
Mucorales/Fusarium Mixed Infection in Hematologic Patient with COVID-19 Complications: An Unfortunate Combination
Next Article in Special Issue
Discovery and Development Strategies for SARS-CoV-2 NSP3 Macrodomain Inhibitors
Previous Article in Journal
rMELEISH: A Novel Recombinant Multiepitope-Based Protein Applied to the Serodiagnosis of Both Canine and Human Visceral Leishmaniasis
Previous Article in Special Issue
The DarT/DarG Toxin–Antitoxin ADP-Ribosylation System as a Novel Target for a Rational Design of Innovative Antimicrobial Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

ADP-Ribosylation in Antiviral Innate Immune Response

1
Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
2
Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(2), 303; https://doi.org/10.3390/pathogens12020303
Submission received: 17 January 2023 / Revised: 9 February 2023 / Accepted: 10 February 2023 / Published: 12 February 2023
(This article belongs to the Special Issue ADP-Ribosylation in Pathogens)

Abstract

:
Adenosine diphosphate (ADP)-ribosylation is a reversible post-translational modification catalyzed by ADP-ribosyltransferases (ARTs). ARTs transfer one or more ADP-ribose from nicotinamide adenine dinucleotide (NAD+) to the target substrate and release the nicotinamide (Nam). Accordingly, it comes in two forms: mono-ADP-ribosylation (MARylation) and poly-ADP-ribosylation (PARylation). ADP-ribosylation plays important roles in many biological processes, such as DNA damage repair, gene regulation, and energy metabolism. Emerging evidence demonstrates that ADP-ribosylation is implicated in host antiviral immune activity. Here, we summarize and discuss ADP-ribosylation modifications that occur on both host and viral proteins and their roles in host antiviral response.

1. Introduction

Rapid and appropriate cellular responses are essential for organisms to respond to external stimuli. Post-translational modifications (PTMs) play an important role in this process by modulating intracellular signal transduction pathways [1]. Mechanically, PTMs regulate signaling pathways and gene expression mainly by affecting the catalytic activity of the target protein or the interaction of the target protein with other molecules [2]. Adenosine diphosphate (ADP)-ribosylation is an ancient PTM discovered around the 1960s [3]. ADP-ribosyltransferases (ARTs) are responsible for ADP-ribosylation [4]. ARTs are a superfamily consisting of 23 members, two of which are diphtheria toxin-like ADP-ribosyltransferase (ARTD) and cholera-like ADP-ribosyltransferase (ARTC) [5]. The ARTD family includes PARPs and TNKS (tankyrase), as detailed in Table 1. ARTs transfer one or more ADP-ribose (ADPr) units from nicotinamide adenine dinucleotide (NAD+) to target proteins on a variety of amino acids, including lysine (Lys), arginine (Arg), glutamate (Glu), aspartate (Asp), and cysteine (Cys) [6], leading to mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation) of substrates. The human genome encodes 17 PARPs [7], most of which share a common NAD+ binding motif in their catalytic domain with a similar secondary structure [8,9]. In these NAD+ binding motifs, a histidine (His) residue and a tyrosine (Tyr) residue are essential for positioning NAD+ in a correct orientation, and a conserved Glu residue is crucial for ADP-ribose transfer. The histidine-tyrosine-glutamate motif (H–Y–E motif and variants thereof) is also known as the catalytic triad of PARPs [10,11]. However, not all PARPs reserve these critical residues. For example, PARP13 lacks catalytic activity due to the substitution of a His residue on the key NAD+-binding motif. Therefore, PARP13 is the only PARP family member with a catalytically inactive domain [12,13]. In fact, only PARP1, 2, 3, TNKS1 and TNKS2 retain these residues and catalytic PARylation [14], while the rest of PARPs (except PARP13) possess the MARylation activity due to the substitution of the Glu residue [8,15,16]. In brief, the ability of PARPs to catalyze MARylation or PARylation of their protein substrates depends on conserved structural features such as the catalytic triad and the presence of certain cofactors.
As for degradation of the poly-ADP-ribose chain, poly-ADP-ribose glycohydrolase (PARG) and ADP-ribosyl-acceptor hydrolase (ARH) 3 are responsible for it. PARG can only hydrolyze ribose-ribosyl O-glycosidic bonds, and the final product of hydrolysis is a MARylated protein. While ARH3 can hydrolyze the bond between the amino acid side chain and the ribose, thereby completely reversing ADP-ribosylation. The chemical bond between the amino acid side chain and ribose is different from the ribose-ribose bond in the PAR chain, which explains why different enzymes are required to accomplish the two reactions. Moreover, ARH1 is also an ADP-ribosylhydrolase, which can specifically hydrolyze MARylated proteins at arginine residues [17,18]. In addition, three macrodomain-containing enzymes, MacroD1, MacroD2, and TARG1, can also act as mono-ADP-ribosylhydrolases [19,20,21]. Notably, ARH3 acts primarily at the terminal sites of the PAR chains and the serine residues of MARylated proteins, while MacroD1, D2, and TARG1 are involved in hydrolyzing ADP-ribosylation of acidic residues [2]. In a word, ADP-ribosylation is a fully reversible PTM. The dynamic balance of ADP-ribosylation synthesis and degradation provides plasticity for rapid cellular response to external signals.
ADP-ribosylation occurs in both prokaryotes and eukaryotes and is particularly prevalent in stress responses requiring rapid adaptation [22,23]. MARylation and PARylation are involved in a variety of physiological and pathophysiological processes, including DNA damage recognition, chromatin regulation, and oxidative stress response [24,25,26,27,28,29,30]. During this last decade, several studies uncovered the roles of ADP-ribosylation in antiviral immunity [31,32]. Of note is that there are many PARPs that perform antiviral functions without their catalytic activity, and will not be discussed in detail here [15]. In this review, we mainly summarize and discuss new findings on the role of ADP-ribosylation occurred on the host and viral proteins in antiviral response. As to other substrates, such as nucleotides or other small molecules, we will only give a brief introduction here (Table 1).
Table 1. The enzymes that catalyze ADP-ribosylation and their main functions in antiviral innate immune response.
Table 1. The enzymes that catalyze ADP-ribosylation and their main functions in antiviral innate immune response.
NameADP-
Ribosylation Activity
Other NamesRoles in Antiviral Innate Immune Response
PARP1PARylationARTD1PARylating EBNA1 and LANA [33,34]
Degradation of IFNAR1 [35]
PARP2PARylationARTD2Not known
PARP3PARylationARTD3Regulating PARP1 [31]
PARP4MARylationARTD4Not known
PARP5aPARylationARTD5
Tankyrase 1
PARylating MAVS and promoting its degradation [36]
PARylating EBNA1 [37]
PARP5bPARylationARTD6
Tankyrase 2
PARylating MAVS and promoting its degradation [36]
PARylating EBNA1 [37]
PARP6MARylationARTD17Not known
PARP7MARylationARTD14,
BAL3
MARylating and inhibiting TBK1 [38]
MARylating PARP13 [23]
PARP8MARylationARTD16Not known
PARP9MARylationARTD9,
BAL1
Forming a complex with DTX3L and MARylating ubiquitin at Gly76 [39]
Suppressing PARP14-mediated MARylation of STAT1 [40]
PARP10MARylationARTD10MARylating nsP2 of CHIKV and promoting its degradation [41]
PARP11MARylationARTD11Targeting β-TrCP and promoting IFNAR1 degradation [42]
Enhancing PARP12-mediated NS1 and NS3 ADP-ribosylation [43]
PARP12MARylationARTD12Targeting NS1 and NS3 of ZIKV and promoting its degradation [44]
PARP13InactiveARTD13Restricts viral replication [45,46,47,48]
PARP14MARylationARTD8MARylating STAT1 and preventing its phosphorylation [40]
MARylating PARP13 [49]
PARP15MARylationARTD7Not known
PARP16MARylationARTD15Not known

2. ADP-Ribosylation of Host Proteins

The innate immune system is the main defense mechanism of higher organisms against pathogens such as viruses. It senses and responds to pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs). During this process, interferons (IFNs) are produced [50,51,52], enabling a rapid host response to viral infection.
Interferons are commonly classified into three types according to their receptor complex, designated types I to III [53,54]. Type I IFN (IFN-I) comprises IFNα and IFNβ, and most virus-infected cells are able to produce IFN-I to resist viruses. Type II IFN (IFN-II) consists only of IFN-γ, which is synthesized by certain immune cells. However, it can offer resistance to a wide range of pathogens [55]. Type III IFN (IFN-III) includes IFN-λ, which can be produced by most cell types. IFN-III plays an important role in the innate immune response of the intestinal and respiratory mucosal barriers [56,57,58]. Among the three types of IFNs, IFN-I is well characterized and plays an important role in the host response against viral infection [59,60,61]. After IFN-I is produced, it first binds to its membrane receptors (IFNAR1 and IFNAR2) to induce their dimerization and then initiates the autophosphorylation of Janus kinase 1 (JAK1) and tyrosine kinase 2 (TYK2). Phosphorylated JAK1 and TYK2 recruit and activate the signal transducers and activators of transcription 1 (STAT1) and STAT2. Phosphorylated STAT1 and STAT2, together with IFN-regulatory factor 9 (IRF9), form a well-characterized complex, IFN-stimulated gene factor 3 (ISGF3). Then, ISGF3 translocates to the nucleus and binds to IFN-stimulated response elements (ISRE), the promoter of IFN-stimulated genes (ISGs), leading to the expression of ISGs [62,63,64,65,66]. Eventually, these ISGs perform antiviral functions, and therefore host establishes the antiviral status [59,67,68]. In addition, IFN affects several other processes, including cell growth, differentiation, and apoptosis, as well as immune regulation [69,70,71].
IFN and IFN-induced ISGs are so important for the maintenance of host antiviral status that their production undergoes complex and delicate regulation. ADP-ribosylation plays an important role in this process. Here, we first focus on ADP-ribosylation modification that occurs on proteins associated with IFN-I production and IFN-I-induced JAK-STAT signaling pathway (Figure 1). These include several proteins such as Mitochondrial antiviral signaling protein (MAVS), TANK-binding kinase 1 (TBK1) and STAT1. In addition, lots of ADP-ribosylation that occurs on other host antiviral factors can also affect host antiviral response. For example, PARP11 MARylates β-transducin repeat-containing protein (β-TrCP) and promote IFNAR1 ubiquitination and degradation, resulting in the downregulation of IFN-I signaling and antiviral activity. Next, we will describe these contents in detail.

2.1. ADP-Ribosylation of Proteins Associated with the Signaling Pathway of IFN-I Production

MAVS (also known as virus-induced signal adaptor [VISA]) is a key signaling molecule that mediates antiviral innate immune response initiated by RNA viruses [72,73,74,75]. In this process, retinoic acid-induced gene I (RIG-I) acts as a pattern recognition receptor to recognize and bind viral RNA in response to RNA virus infection [76]. Subsequent conformational change of RIG-I exposes its N-terminal caspase recruitment domain (2CARD), which binds to the N-terminal CARD domain of MAVS and induces MAVS polymerization at the mitochondrial outer membrane [77]. Active MAVS polymers recruit the tumor necrosis factor receptor-associated factor (TRAF) family to synthesize polyubiquitin chains that activate TBK1 and IκB-kinase (IKK). Activated TBK1 and IKK first phosphorylate MAVS so that phosphorylated MAVS can recruit IFN-regulatory factor 3 (IRF3) by binding to its conserved positively charged surface. When TBK1 and IRF3 are in close proximity, IRF3 is phosphorylated by TBK1. Finally, phosphorylated IRF3 dissociates from MAVS and translocates to the nucleus after forming a dimer, where it binds to the promoter of the IFN-I gene and drives type I IFN production [50]. Therefore, the strict and subtle dynamic regulation of MAVS is very important to antiviral immune response. PARP5a/Tankyrase 1 (TNKS1) and its homolog PARP5b/TNKS2 are known to catalyze the PARylation of their substrates. The five ankyrin repeat (ANK) units at the N-terminus of TNKS1 and TNKS2 are the structural basis for their substrate recognition. The C-terminal catalytic domain is responsible for the ADP-ribosylation of their substrates [78]. A recent study showed that TNKS1 and TNKS2 can poly-ADP-ribosylate MAVS at Glu137 residue. After viral infection, TNKS1 and TNKS2 are upregulated and translocate from the cytosol to mitochondria, interacting with MAVS and catalyzing its PARylation [36]. The PARylation of MAVS serves as a signal for the ubiquitin E3 ligase Ring figure protein 146 (RNF146)-mediated K48-linked polyubiquitination and subsequent degradation of MAVS [79], thereby negatively regulating the innate immune response to RNA viruses.
As mentioned above, TBK1 is a key kinase that induces IFN-I production. PARP7 (TIPARP) is an ADP-ribosylase whose expression is upregulated by aryl hydrocarbon receptor (AHR) [38,80]. AHR is a ligand-activated transcription factor that can be activated by a variety of environmental xenobiotics. Therefore, there is a close connection between AHR and innate immune signaling [80,81,82,83]. During viral infection, AHR-induced PARP7 can interact with TBK1 and catalyze its mono-ADP-ribosylation, which suppresses the activation of TBK1 and subsequent phosphorylation of IRF3. Consistent with this, AHR-deficient (Ahr−/−) MEFs and MEFs with PARP7-knockdown by siPARP7 show stronger antiviral effects [38]. Thus, the AHR-PARP7 axis is a negative regulator of interferon signaling. However, the ADP ribosylation site of TBK1 remains to be determined. It is reported that PARP7 mainly modifies cysteines and acidic amino acids (glutamates and aspartates), which also provides a basis for the determination of the ADP-ribosylation site of TBK1 [84,85].

2.2. ADP-Ribosylation of Proteins Associated with IFN-I-Induced JAK-STAT Signaling Pathway

STAT1 stands out as a key functional component of the interferon signaling pathway, and its post-translational modification profoundly affects signal transduction [86]. For example, linear ubiquitination of STAT1 inhibits STAT1 activation and thus lowers the strength of IFN-I antiviral signaling [66]. The ADP-ribosylation of STAT1 was first reported by Iwata et al. [40]. It is PARP14 that catalyzes mono-ADP-ribosylation of STAT1 and prevents its phosphorylation. Consistent with this, PARP14 silencing and STAT1 ribosylation site mutation promote STAT1 phosphorylation and STAT1-driven ISG expression, thus enhancing interferon signaling. Interestingly, PARP9 and PARP14 physically and functionally interact with each other. Protein ribosylation assay and mass spectrometry showed that PARP9 suppresses PARP14-mediated MARylation of STAT1, which sustains STAT1 phosphorylation. In other words, PARP14 and PARP9 play opposite roles in this process [40]. However, STAT1 SUMOylation also affects its phosphorylation, which in turn affects interferon downstream events [87]. The ADP-ribosylation of STAT1 has therefore been questioned and requires further evidence. It is worth noting that in mouse bone marrow-derived macrophages (BMDMs), the mRNA level of PARP14 is significantly upregulated when stimulated by IFNβ or toll-like receptor (TLR) agonists such as polyinosinic-polycytidylic acid (Poly (I: C)). Depletion of PARP14 results in a decrease in IRF3-mediated IFNβ production and ISG expression, thereby impairing the interferon response [88]. Taken together, PARP14 plays a complex role in the process of interferon resistance to pathogens.

2.3. ADP-Ribosylation of Other Host Antiviral Factors

β-TrCP is the ubiquitin E3 ligase of interferon-alpha/beta receptor 1 (IFNAR1) and mediates IFNAR1 ubiquitination and degradation [89]. β-TrCP belongs to the F-box/WD40 repeats family, which contains an F-box motif and seven WD40 motifs [90]. Among them, WD40 repeats are responsible for the binding of β-TrCP to its protein substrate. PARP11 can mono-ADP-ribosylate β-TrCP in the WD40 repeats. The MARylatopn of β-TrCP inhibits its ubiquitin-proteasome degradation and enhances the ability of β-TrCP to interact with IFNAR1, which in turn promotes IFNAR1 ubiquitination and degradation. The above process ultimately results in the downregulation of IFN-I signaling and antiviral activity. Moreover, viral infection could lead to the upregulation of PARP11, thus restricting IFN-I-induced expression of ISGs and promoting ADP-ribosylation-mediated immune evasion of the virus. Similarly, PARP11 knockdown significantly downregulates the protein level of β-TrCP and upregulates the protein level of IFNAR1, therefore enhancing the IFN-I-activated signaling pathway and antiviral activity. Taken together, PARP11 and PARP11-mediate-MARylation are highly effective targets for improving the antiviral efficacy of type-I interferon [42]. Consistent with this, a recent study shows that during Influenza A viruses (IAV) infection, PARP1 activity could facilitate IAV-induced IFNAR1 degradation, thus promoting virus propagation [35]. However, the substrate for this ADP-ribosylation process remains to be determined.
Initially, PARP9 was also thought to be enzymatically inactive. PARP9 was reported to form a heterodimer complex with Deltex E3 ubiquitin ligase 3L (DTX3L) [91]. DTX3L and PARP9 are relatively highly expressed in prostate cancer and breast cancer and share a common promoter. Both genes are responsive to IFNγ and activated in cells that express a dominant active form of STAT1 [92]. In addition, the complex can interact with STAT1 to promote STAT1-mediated ISG expression and thus enhance antiviral response [93]. Interestingly, in 2017, a study found that PARP9 can display mono-ADP-ribosylation activity in the case of DTX3L as its chaperone. DTX3L is an E3 ligase containing a RING domain, and PARP9/DTX3L heterodimer still has an E3 ligase activity. In the presence of high levels of NAD+, E1 and E2 enzymes, PARP9/DTX3L heterodimer exhibits ADP-ribosyltransferase activity and catalyzes mono-ADP-ribosylation of ubiquitin (Ub) at Gly76, which is involved in Ub conjugation to substrates. Therefore, the ADP-ribosylation of the Ub restricts the E3 function of DTX3L. In this process, both the RING domain of DTX3L and the catalytic domain of PARP9 are indispensable [39]. Although it has been reported that PARP9/DTX3L can target host histone H2BJ to enhance ISG expression and target encephalomyocarditis virus (EMCV) 3C protease to disrupt viral assembly as an E3 ubiquitin ligase [93], current evidence shows that PARP9 can only ADP-ribosylate Ub. It highlights the selectivity of ADP-ribosylation. However, the effect of PARP9-mediated MARylation on the antiviral activity of PARP9/DTX3L remains to be further investigated.
In addition, PARP7 can MARylate other proteins that function in antiviral immunity, such as PARP13. Recent studies have shown that PARP7 MARylates PARP13 at cysteines (Cys), rather than glutamates (Glu) or aspartates (Asp) [23]. Of course, there are many examples of PARP7 modifying these two amino acids, such as PARP7 MARylates α-tubulin at glutamic acid and aspartic acid residues [84]. PARP13 is also a member of the PARP family, which is considered to be catalytically inactive [13]. Even so, PARP13 is still known for its antiviral activity [48]. PARP13 has shown antiviral activity against a variety of DNA and RNA viruses, including influenza A virus, alphaviruses, filoviruses, and human immunodeficiency virus 1 (HIV-1) [45,46,47,94]. Mechanistically, PARP13 can promote interferon signaling by interacting with RIG-I and other ISGs. In addition, PARP13 can also target viral RNA to induce its degradation [95,96,97]. Since the Cys residues in the zinc-finger domains of PARP13 are responsible for the coordination of Zn2+ [23,85], the MARylation of these Cys residues may prevent RNA binding. Thus, the Cys MARylation of PARP13 appears to limit the antiviral response. Of note is that the MARylation of Cys in cells is more stable than Glu/Asp, suggesting that the MARylation site of protein targets regulates the duration of the signal. Moreover, PARP14 can also MARylate PARP13 on several Glu/Asp residues, providing a link for crosstalk among PARP family members [49].

3. ADP-Ribosylation of Virus-Encoded Proteins

Recent studies have found that viral proteins can also undergo ADP-ribosylation modification. These include the Epstein-Barr nuclear antigen 1 (EBNA1) of Epstein-Barr virus (EBV), the latency-associated nuclear antigen (LANA) of Kaposi’s sarcoma-associated herpesvirus (KSHV), the nonstructural proteins NS1 and NS3 of Zika virus (ZIKV), the nonstructural polyprotein nsP2 of Chikungunya virus (CHIKV), the nucleocapsid (N) protein of coronavirus (CoVs) and the core proteins of adenovirus.
EBV is a human herpesvirus, and EBNA1 is the only viral protein required for the stable maintenance of the viral genome [98,99]. It is reported that PARP1 and TNKS can interact directly with the EBNA1 protein and catalyze its poly-ADP-ribosylation, resulting in the inhibition of OriP replication [33,37]. In the same way, PARP1 can also PARylate LANA of KSHV. KSHV, another human herpesvirus, is usually in the latent phase of disease as reported for EVB [34,100,101]. In latent replication, the KSHV genome is considered to replicate once per cell cycle, and several potentially pathogenic genes such as LANA, K-cyclin (ORF72), K15, and vFLIP (ORF71) are expressed [102]. Thus, the poly-ADP-ribosylation of LANA appears to affect the maintenance of the viral genome. Taking the above discussion together, PARP1- and TNKS-mediated ADP-ribosylation of viral proteins inhibit viral replication and infection using a similar mechanism.
ZiKV is a mosquito-borne flavivirus with a single-stranded positive RNA genome [103,104], and its nonstructural viral proteins NS1 and NS3 have been reported to be targeted by PARP12 [44]. In this process, PARP12 is responsible for the initial MARylation reaction of NS1 and NS3. Subsequent PARylation is performed by other PARPs. This modification of NS1 and NS3 triggers their K48-linked ubiquitination and proteasome-mediated degradation. Since NS1 and NS3 are involved in viral replication and immune evasion [105,106], the PARP-dependent degradation of NS1 and NS3 suppresses Zika virus infection and immune evasion [44]. In addition, a recent study finds that PARP11 can enhance PARP12-mediated NS1 and NS3 ADP-ribosylation and degradation, thus inhibiting Zika virus infection [43]. However, the exact mechanism remains to be elucidated. Again, the ADP-ribosylation of ZiKV proteins shows the potent antiviral activity of this modification.
CHIKV is a mosquito-borne virus with a genome of approximately 11,800 nucleotides [107]. Early in infection, CHIKV encodes a nonstructural polyprotein (nsP). Subsequently, nsP is cleaved into four separate nsP1–4 that together form a replication complex. In this process, nsP2 functions as a key protease. Therefore, it is an important target for antiviral drugs [108,109,110,111]. PARP10 can MARylate nsP2 and its protease domain and inhibit its proteolytic activity, thus restricting the processing of nsP and suppressing CHIKV replication. Interestingly, nsP3 is responsible for MAR hydrolase activity and can remove the MARylation modification from nsP2, thereby reactivating its proteolytic activity and promoting CHIKV infection [41,112,113]. In summary, ADP-ribosylation plays an indispensable role in host antiviral response.
In addition, it has been reported that some viral proteins can undergo ADP-ribosylation, but which PARP catalyzes this process, and the effect of ADP-ribosylation on viral infection are still unclear. For example, the coronavirus (CoVs) nucleocapsid (N) protein is ADP-ribosylated in cells during coronavirus infection, and the nsp3 macrodomain does not affect ADP-ribosylation of the N protein. N protein ADP-ribosylation can only be detected in the context of viral infection and cannot be detected in mock-infected cells. Interestingly, nucleocapsid protein ADP-ribosylation is conserved in both α- and β-coronaviruses [114]. This suggests that it is important for viral replication in the host or host resistance to the virus. Future experiments are needed to explore the function of N protein ADP-ribosylation. Another example is that adenovirus core proteins also undergo ADP-ribosylation during viral infection and may play a role in virus decapsidation [115]. However, more details remain to be explored.

4. ADP-Ribosylation of Nucleic Acid Molecules

It is of interest to mention that in addition to proteins, many nucleic acid molecules can also undergo MARylation or PARylation modification [116,117,118,119,120], but we still know relatively little about them. The first nucleic acid discovered to undergo ADP-ribosylation was bacterial DNA, which results in an inhibition of bacterial DNA replication. The antitoxin DarG, a macrodomain protein, catalyzes this process [121]. This suggests that ADP-ribosylation of nucleic acids may also be present in mammals. In fact, it has been shown that PARP1 and PARP2 can PARylate the phosphorylated ends of double-stranded or single-stranded DNA in vitro [122,123]. Similarly, PARP3 can MARylate DNA substrates [124]. In addition, RNA can also serve as a substrate for ADP-ribosylation. In vitro experiments showed that PARP10 can modify the 5′-phosphorylated termini of RNA [119]. Post-translational modifications of these nucleic acid molecules provide new insights into the molecular mechanisms of ADP-ribosylation modifications mediated by PARPs. However, these in vitro experiments still need more validation, especially in vivo validation. We do not yet know whether PARPs are also sequence-specific when recognizing nucleic acid molecules, which still requires more exploration. The next step is to explore whether ADP-ribosylation occurs on viral genomes and whether it is related to antiviral immunity response. This might provide a seed for subsequent studies.

5. Conclusions and Perspectives

As discussed above, ADP-ribosylation plays a complex function in antiviral response. ADP-ribosylation of proteins involved in IFN-I signaling pathways generally weakens antiviral responses, whereas inhibition of this process enhances antiviral signaling. Interestingly, several of the rapidly evolving PARP genes, including PARP9, PARP10, PARP12, PARP13, and PARP14, are upregulated by IFN, and overexpression of them upregulates several antiviral effectors. Therefore, these PARPs are considered as ISGs [125,126,127,128]. ADP-ribosylation of viral proteins often promotes their degradation and thus enhances the host antiviral response, while viruses have also evolved strategies to reverse ADP-ribosylation modifications. One common strategy is that viral macrodomains can degrade ADP-ribosylation modification. As previously described, the nsP3 macrodomain of CHIKV is able to remove the MARylation modification of nsP2. In addition, severe acute respiratory syndrome coronavirus (SARS-CoV) macrodomain mutations significantly increase the virus’s sensitivity to interferon. Similarly, mutations in the ADP-ribose binding region of the Sindbis virus (SINV) macrodomain impair viral replication [129,130]. All these suggest that viral macrodomains may play an important role in antiviral immune escape. It appears that viral macrodomains are phylogenetically and structurally closely related to cellular macrodomains. It is, therefore, not surprising that viral macrodomains can hydrolyze ADP-ribosylation [41,131,132,133]. Together, these observations offer mechanisms of how ADP-ribosylation functions in host-virus conflicts. It seems that ADP-ribosylation can both enhance and restrict the antiviral response, depending on the specificity of the substrate. Therefore, a thorough understanding of the role of ADP-ribosylation in the antiviral response is crucial for the prevention and treatment of virus-associated diseases.
ADP-ribosylation plays such an important role in many biological processes, such as innate immunity, that PARP inhibitors (PARPi) have also become a research hotspot in recent years. For example, rucaparib inhibits PARP11-induced β-TrCP ADP-ribosylation, leading to a decrease in β-TrCP level and an increase in IFNAR1 level [42]. Therefore, rucaparib can effectively promote type-I IFN signaling pathway transduction and enhance host antiviral activity. Other PARP inhibitors, such as olaparib, veliparib, and niraparib, are still under experimental or clinical investigation with rapid progress [134,135]. However, there is still a long way to go in the development of PARP inhibitors due to the difficulty of developing specific inhibitors for individual PARPs and the drug resistance of PARP inhibitors.
Despite these notions, we would like to point out that only a few direct substrates of ADP-ribosylation have been identified so far. Thus far, we do not have a deep understanding of how PARPs select and interact with target substrates and how they are activated [116]. It might be interesting to define more broadly relevant substrates (both host and viral factors), which requires further exploration. Certainly, the identification of specific sites of ADP-ribosylation is equally important. With the development of mass spectrometry techniques and detection tools, it is possible to identify specific substrates for ADP-ribosylation and map modification sites. This will contribute to a further understanding of the role of ADP-ribosylation in antiviral response.

Author Contributions

Conceptualization, Q.D. and H.Z.; writing—review and editing, Q.D., H.Z., Y.M. and W.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by the National Natural Science Foundation of China (32241009, 31970846) and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, J.; Qian, C.; Cao, X. Post-Translational Modification Control of Innate Immunity. Immunity 2016, 45, 15–30. [Google Scholar] [CrossRef] [PubMed]
  2. Luscher, B.; Butepage, M.; Eckei, L.; Krieg, S.; Verheugd, P.; Shilton, B.H. ADP-Ribosylation, a Multifaceted Posttranslational Modification Involved in the Control of Cell Physiology in Health and Disease. Chem. Rev. 2018, 118, 1092–1136. [Google Scholar] [CrossRef]
  3. Chambon, P.; Weill, J.D.; Mandel, P. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. BioChem. Biophys Res. Commun. 1963, 11, 39–43. [Google Scholar] [CrossRef]
  4. Schreiber, V.; Dantzer, F.; Ame, J.C.; de Murcia, G. Poly(ADP-ribose): Novel functions for an old molecule. Nat. Rev. Mol. Cell Biol. 2006, 7, 517–528. [Google Scholar] [CrossRef] [PubMed]
  5. Lüscher, B.; Ahel, I.; Altmeyer, M.; Ashworth, A.; Bai, P.; Chang, P.; Cohen, M.; Corda, D.; Dantzer, F.; Daugherty, M.D.; et al. ADP-ribosyltransferases, an update on function and nomenclature. FEBS J. 2022, 289, 7399–7410. [Google Scholar] [CrossRef] [PubMed]
  6. Gibson, B.A.; Kraus, W.L. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat. Rev. Mol. Cell Biol. 2012, 13, 411–424. [Google Scholar] [CrossRef] [PubMed]
  7. Hassa, P.O.; Hottiger, M.O. The diverse biological roles of mammalian PARPS, a small but powerful family of poly-ADP-ribose polymerases. Front. Biosci. 2008, 13, 3046–3082. [Google Scholar] [CrossRef] [PubMed]
  8. Hottiger, M.O.; Hassa, P.O.; Luscher, B.; Schuler, H.; Koch-Nolte, F. Toward a unified nomenclature for mammalian ADP-ribosyltransferases. Trends BioChem. Sci. 2010, 35, 208–219. [Google Scholar] [CrossRef]
  9. Gagne, J.P.; Ethier, C.; Defoy, D.; Bourassa, S.; Langelier, M.F.; Riccio, A.A.; Pascal, J.M.; Moon, K.M.; Foster, L.J.; Ning, Z.; et al. Quantitative site-specific ADP-ribosylation profiling of DNA-dependent PARPs. DNA Repair. (Amst.) 2015, 30, 68–79. [Google Scholar] [CrossRef]
  10. Otto, H.; Reche, P.A.; Bazan, F.; Dittmar, K.; Haag, F.; Koch-Nolte, F. In silico characterization of the family of PARP-like poly(ADP-ribosyl)transferases (pARTs). BMC Genom. 2005, 6, 139. [Google Scholar] [CrossRef]
  11. Bell, C.E.; Eisenberg, D. Crystal structure of diphtheria toxin bound to nicotinamide adenine dinucleotide. Biochemistry 1996, 35, 1137–1149. [Google Scholar] [CrossRef]
  12. Todorova, T.; Bock, F.J.; Chang, P. Poly(ADP-ribose) polymerase-13 and RNA regulation in immunity and cancer. Trends Mol. Med. 2015, 21, 373–384. [Google Scholar] [CrossRef]
  13. Karlberg, T.; Klepsch, M.; Thorsell, A.G.; Andersson, C.D.; Linusson, A.; Schuler, H. Structural basis for lack of ADP-ribosyltransferase activity in poly(ADP-ribose) polymerase-13/zinc finger antiviral protein. J. Biol. Chem. 2015, 290, 7336–7344. [Google Scholar] [CrossRef]
  14. Rippmann, J.F.; Damm, K.; Schnapp, A. Functional characterization of the poly(ADP-ribose) polymerase activity of tankyrase 1, a potential regulator of telomere length. J. Mol. Biol. 2002, 323, 217–224. [Google Scholar] [CrossRef]
  15. Lüscher, B.; Verheirstraeten, M.; Krieg, S.; Korn, P. Intracellular mono-ADP-ribosyltransferases at the host-virus interphase. Cell Mol. Life Sci. 2022, 79, 288. [Google Scholar] [CrossRef]
  16. Kleine, H.; Poreba, E.; Lesniewicz, K.; Hassa, P.O.; Hottiger, M.O.; Litchfield, D.W.; Shilton, B.H.; Luscher, B. Substrate-assisted catalysis by PARP10 limits its activity to mono-ADP-ribosylation. Mol. Cell 2008, 32, 57–69. [Google Scholar] [CrossRef]
  17. Moss, J.; Stanley, S.J.; Nightingale, M.S.; Murtagh, J.J., Jr.; Monaco, L.; Mishima, K.; Chen, H.C.; Williamson, K.C.; Tsai, S.C. Molecular and immunological characterization of ADP-ribosylarginine hydrolases. J. Biol. Chem. 1992, 267, 10481–10488. [Google Scholar] [CrossRef]
  18. Kato, J.; Zhu, J.; Liu, C.; Moss, J. Enhanced sensitivity to cholera toxin in ADP-ribosylarginine hydrolase-deficient mice. Mol. Cell Biol. 2007, 27, 5534–5543. [Google Scholar] [CrossRef]
  19. Bonicalzi, M.E.; Haince, J.F.; Droit, A.; Poirier, G.G. Regulation of poly(ADP-ribose) metabolism by poly(ADP-ribose) glycohydrolase: Where and when? Cell Mol. Life Sci. 2005, 62, 739–750. [Google Scholar] [CrossRef]
  20. Oka, S.; Kato, J.; Moss, J. Identification and characterization of a mammalian 39-kDa poly(ADP-ribose) glycohydrolase. J. Biol. Chem. 2006, 281, 705–713. [Google Scholar] [CrossRef]
  21. Mashimo, M.; Kato, J.; Moss, J. Structure and function of the ARH family of ADP-ribosyl-acceptor hydrolases. DNA Repair. (Amst.) 2014, 23, 88–94. [Google Scholar] [CrossRef]
  22. Cho, C.C.; Chien, C.Y.; Chiu, Y.C.; Lin, M.H.; Hsu, C.H. Structural and biochemical evidence supporting poly ADP-ribosylation in the bacterium Deinococcus radiodurans. Nat. Commun. 2019, 10, 1491. [Google Scholar] [CrossRef]
  23. Rodriguez, K.M.; Buch-Larsen, S.C.; Kirby, I.T.; Siordia, I.R.; Hutin, D.; Rasmussen, M.; Grant, D.M.; David, L.L.; Matthews, J.; Nielsen, M.L.; et al. Chemical genetics and proteome-wide site mapping reveal cysteine MARylation by PARP-7 on immune-relevant protein targets. Elife 2021, 10, e60480. [Google Scholar] [CrossRef]
  24. Audebert, M.; Salles, B.; Calsou, P. Involvement of poly(ADP-ribose) polymerase-1 and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J. Biol. Chem. 2004, 279, 55117–55126. [Google Scholar] [CrossRef]
  25. Langelier, M.F.; Planck, J.L.; Roy, S.; Pascal, J.M. Structural basis for DNA damage-dependent poly(ADP-ribosyl)ation by human PARP-1. Science 2012, 336, 728–732. [Google Scholar] [CrossRef]
  26. Kraus, W.L.; Lis, J.T. PARP goes transcription. Cell 2003, 113, 677–683. [Google Scholar] [CrossRef]
  27. Luo, X.; Kraus, W.L. On PAR with PARP: Cellular stress signaling through poly(ADP-ribose) and PARP-1. Genes Dev. 2012, 26, 417–432. [Google Scholar] [CrossRef]
  28. Hassa, P.O.; Hottiger, M.O. The functional role of poly(ADP-ribose)polymerase 1 as novel coactivator of NF-kappaB in inflammatory disorders. Cell Mol. Life Sci. 2002, 59, 1534–1553. [Google Scholar] [CrossRef]
  29. Bai, P.; Canto, C.; Oudart, H.; Brunyanszki, A.; Cen, Y.; Thomas, C.; Yamamoto, H.; Huber, A.; Kiss, B.; Houtkooper, R.H.; et al. PARP-1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metab. 2011, 13, 461–468. [Google Scholar] [CrossRef]
  30. Pellegrino, S.; Altmeyer, M. Interplay between Ubiquitin, SUMO, and Poly(ADP-Ribose) in the Cellular Response to Genotoxic Stress. Front. Genet. 2016, 7, 63. [Google Scholar] [CrossRef]
  31. Rosado, M.M.; Pioli, C. ADP-ribosylation in evasion, promotion and exacerbation of immune responses. Immunology 2021, 164, 15–30. [Google Scholar] [CrossRef] [PubMed]
  32. Zhu, H.; Zheng, C. When PARPs Meet Antiviral Innate Immunity. Trends MicroBiol. 2021, 29, 776–778. [Google Scholar] [CrossRef]
  33. Tempera, I.; Deng, Z.; Atanasiu, C.; Chen, C.J.; D’Erme, M.; Lieberman, P.M. Regulation of Epstein-Barr virus OriP replication by poly(ADP-ribose) polymerase 1. J. Virol. 2010, 84, 4988–4997. [Google Scholar] [CrossRef] [PubMed]
  34. Ohsaki, E.; Ueda, K.; Sakakibara, S.; Do, E.; Yada, K.; Yamanishi, K. Poly(ADP-ribose) polymerase 1 binds to Kaposi’s sarcoma-associated herpesvirus (KSHV) terminal repeat sequence and modulates KSHV replication in latency. J. Virol. 2004, 78, 9936–9946. [Google Scholar] [CrossRef]
  35. Xia, C.; Wolf, J.J.; Sun, C.; Xu, M.; Studstill, C.J.; Chen, J.; Ngo, H.; Zhu, H.; Hahm, B. PARP1 Enhances Influenza A Virus Propagation by Facilitating Degradation of Host Type I Interferon Receptor. J. Virol. 2020, 94, e01572-19. [Google Scholar] [CrossRef]
  36. Xu, Y.R.; Shi, M.L.; Zhang, Y.; Kong, N.; Wang, C.; Xiao, Y.F.; Du, S.S.; Zhu, Q.Y.; Lei, C.Q. Tankyrases inhibit innate antiviral response by PARylating VISA/MAVS and priming it for RNF146-mediated ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2022, 119, e2122805119. [Google Scholar] [CrossRef]
  37. Deng, Z.; Atanasiu, C.; Zhao, K.; Marmorstein, R.; Sbodio, J.I.; Chi, N.W.; Lieberman, P.M. Inhibition of Epstein-Barr virus OriP function by tankyrase, a telomere-associated poly-ADP ribose polymerase that binds and modifies EBNA1. J. Virol. 2005, 79, 4640–4650. [Google Scholar] [CrossRef]
  38. Yamada, T.; Horimoto, H.; Kameyama, T.; Hayakawa, S.; Yamato, H.; Dazai, M.; Takada, A.; Kida, H.; Bott, D.; Zhou, A.C.; et al. Constitutive aryl hydrocarbon receptor signaling constrains type I interferon-mediated antiviral innate defense. Nat. Immunol. 2016, 17, 687–694. [Google Scholar] [CrossRef]
  39. Yang, C.S.; Jividen, K.; Spencer, A.; Dworak, N.; Ni, L.; Oostdyk, L.T.; Chatterjee, M.; Kusmider, B.; Reon, B.; Parlak, M.; et al. Ubiquitin Modification by the E3 Ligase/ADP-Ribosyltransferase Dtx3L/Parp9. Mol. Cell 2017, 66, 503–516.e5. [Google Scholar] [CrossRef]
  40. Iwata, H.; Goettsch, C.; Sharma, A.; Ricchiuto, P.; Goh, W.W.; Halu, A.; Yamada, I.; Yoshida, H.; Hara, T.; Wei, M.; et al. PARP9 and PARP14 cross-regulate macrophage activation via STAT1 ADP-ribosylation. Nat. Commun. 2016, 7, 12849. [Google Scholar] [CrossRef]
  41. Krieg, S.; Pott, F.; Eckei, L.; Verheirstraeten, M.; Bütepage, M.; Lippok, B.; Goffinet, C.; Lüscher, B. Mono-ADP-ribosylation by ARTD10 restricts Chikungunya virus replication by interfering with the proteolytic activity of nsP2. bioRxiv 2020. [Google Scholar] [CrossRef]
  42. Guo, T.; Zuo, Y.; Qian, L.; Liu, J.; Yuan, Y.; Xu, K.; Miao, Y.; Feng, Q.; Chen, X.; Jin, L.; et al. ADP-ribosyltransferase PARP11 modulates the interferon antiviral response by mono-ADP-ribosylating the ubiquitin E3 ligase β-TrCP. Nat. MicroBiol. 2019, 4, 1872–1884. [Google Scholar] [CrossRef] [PubMed]
  43. Li, L.; Shi, Y.; Li, S.; Liu, J.; Zu, S.; Xu, X.; Gao, M.; Sun, N.; Pan, C.; Peng, L.; et al. ADP-ribosyltransferase PARP11 suppresses Zika virus in synergy with PARP12. Cell Biosci. 2021, 11, 116. [Google Scholar] [CrossRef] [PubMed]
  44. Li, L.; Zhao, H.; Liu, P.; Li, C.; Quanquin, N.; Ji, X.; Sun, N.; Du, P.; Qin, C.F.; Lu, N.; et al. PARP12 suppresses Zika virus infection through PARP-dependent degradation of NS1 and NS3 viral proteins. Sci. Signal. 2018, 11, eaas9332. [Google Scholar] [CrossRef] [PubMed]
  45. Bick, M.J.; Carroll, J.W.; Gao, G.; Goff, S.P.; Rice, C.M.; MacDonald, M.R. Expression of the zinc-finger antiviral protein inhibits alphavirus replication. J. Virol. 2003, 77, 11555–11562. [Google Scholar] [CrossRef] [PubMed]
  46. Müller, S.; Möller, P.; Bick, M.J.; Wurr, S.; Becker, S.; Günther, S.; Kümmerer, B.M. Inhibition of filovirus replication by the zinc finger antiviral protein. J. Virol. 2007, 81, 2391–2400. [Google Scholar] [CrossRef] [PubMed]
  47. Tang, Q.; Wang, X.; Gao, G. The Short Form of the Zinc Finger Antiviral Protein Inhibits Influenza A Virus Protein Expression and Is Antagonized by the Virus-Encoded NS1. J. Virol. 2017, 91, e01909-16. [Google Scholar] [CrossRef]
  48. Xue, G.; Braczyk, K.; Gonçalves-Carneiro, D.; Dawidziak, D.M.; Sanchez, K.; Ong, H.; Wan, Y.; Zadrozny, K.K.; Ganser-Pornillos, B.K.; Bieniasz, P.D.; et al. Poly(ADP-ribose) potentiates ZAP antiviral activity. PLoS Pathog. 2022, 18, e1009202. [Google Scholar] [CrossRef]
  49. Carter-O’Connell, I.; Vermehren-Schmaedick, A.; Jin, H.; Morgan, R.K.; David, L.L.; Cohen, M.S. Combining Chemical Genetics with Proximity-Dependent Labeling Reveals Cellular Targets of Poly(ADP-ribose) Polymerase 14 (PARP14). ACS Chem. Biol. 2018, 13, 2841–2848. [Google Scholar] [CrossRef]
  50. Liu, S.; Cai, X.; Wu, J.; Cong, Q.; Chen, X.; Li, T.; Du, F.; Ren, J.; Wu, Y.T.; Grishin, N.V.; et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 2015, 347, aaa2630. [Google Scholar] [CrossRef]
  51. Park, A.; Iwasaki, A. Type I and Type III Interferons—Induction, Signaling, Evasion, and Application to Combat COVID-19. Cell Host Microbe 2020, 27, 870–878. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, H.G.; Wang, B.; Yang, Y.; Liu, X.; Wang, J.; Xin, N.; Li, S.; Miao, Y.; Wu, Q.; Guo, T.; et al. Depression compromises antiviral innate immunity via the AVP-AHI1-Tyk2 axis. Cell Res. 2022, 32, 897–913. [Google Scholar] [CrossRef] [PubMed]
  53. Pestka, S.; Krause, C.D.; Walter, M.R. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, S.Y.; Boisson-Dupuis, S.; Chapgier, A.; Yang, K.; Bustamante, J.; Puel, A.; Picard, C.; Abel, L.; Jouanguy, E.; Casanova, J.L. Inborn errors of interferon (IFN)-mediated immunity in humans: Insights into the respective roles of IFN-alpha/beta, IFN-gamma, and IFN-lambda in host defense. Immunol. Rev. 2008, 226, 29–40. [Google Scholar] [CrossRef] [PubMed]
  55. Muller, U.; Steinhoff, U.; Reis, L.F.; Hemmi, S.; Pavlovic, J.; Zinkernagel, R.M.; Aguet, M. Functional role of type I and type II interferons in antiviral defense. Science 1994, 264, 1918–1921. [Google Scholar] [CrossRef] [PubMed]
  56. Ye, L.; Schnepf, D.; Staeheli, P. Interferon-λ orchestrates innate and adaptive mucosal immune responses. Nat. Rev. Immunol. 2019, 19, 614–625. [Google Scholar] [CrossRef]
  57. Kotenko, S.V.; Rivera, A.; Parker, D.; Durbin, J.E. Type III IFNs: Beyond antiviral protection. Semin. Immunol. 2019, 43, 101303. [Google Scholar] [CrossRef]
  58. Ank, N.; West, H.; Bartholdy, C.; Eriksson, K.; Thomsen, A.R.; Paludan, S.R. Lambda interferon (IFN-lambda), a type III IFN, is induced by viruses and IFNs and displays potent antiviral activity against select virus infections in vivo. J. Virol. 2006, 80, 4501–4509. [Google Scholar] [CrossRef]
  59. Schneider, W.M.; Chevillotte, M.D.; Rice, C.M. Interferon-Stimulated Genes: A Complex Web of Host Defenses. Annu. Rev. Immunol. 2014, 32, 513–545. [Google Scholar] [CrossRef]
  60. Sadler, A.J.; Williams, B.R. Interferon-inducible antiviral effectors. Nat. Rev. Immunol. 2008, 8, 559–568. [Google Scholar] [CrossRef]
  61. Zuo, Y.; He, J.; Liu, S.; Xu, Y.; Liu, J.; Qiao, C.; Zang, L.; Sun, W.; Yuan, Y.; Zhang, H.; et al. LATS1 is a central Signal. transmitter for achieving full type-I interferon activity. Sci. Adv. 2022, 8, eabj3887. [Google Scholar] [CrossRef] [PubMed]
  62. Mohr, A.; Chatain, N.; Domoszlai, T.; Rinis, N.; Sommerauer, M.; Vogt, M.; Muller-Newen, G. Dynamics and non-canonical aspects of JAK/STAT signalling. Eur. J. Cell Biol. 2012, 91, 524–532. [Google Scholar] [CrossRef] [PubMed]
  63. Levy, D.E.; Kessler, D.S.; Pine, R.; Reich, N.; Darnell, J.E., Jr. Interferon-induced nuclear factors that bind a shared promoter element correlate with positive and negative transcriptional control. Genes Dev. 1988, 2, 383–393. [Google Scholar] [CrossRef] [PubMed]
  64. Yuan, Y.; Miao, Y.; Qian, L.; Zhang, Y.; Liu, C.; Liu, J.; Zuo, Y.; Feng, Q.; Guo, T.; Zhang, L.; et al. Targeting UBE4A Revives Viperin Protein in Epithelium to Enhance Host Antiviral Defense. Mol. Cell 2020, 77, 734–747.e737. [Google Scholar] [CrossRef]
  65. Yuan, Y.; Miao, Y.; Ren, T.; Huang, F.; Qian, L.; Chen, X.; Zuo, Y.; Zhang, H.G.; He, J.; Qiao, C.; et al. High salt activates p97 to reduce host antiviral immunity by restricting Viperin induction. EMBO Rep. 2022, 23, e53466. [Google Scholar] [CrossRef]
  66. Zuo, Y.; Feng, Q.; Jin, L.; Huang, F.; Miao, Y.; Liu, J.; Xu, Y.; Chen, X.; Zhang, H.; Guo, T.; et al. Regulation of the linear ubiquitination of STAT1 controls antiviral interferon signaling. Nat. Commun. 2020, 11, 1146. [Google Scholar] [CrossRef]
  67. Liu, S.Y.; Aliyari, R.; Chikere, K.; Li, G.; Marsden, M.D.; Smith, J.K.; Pernet, O.; Guo, H.; Nusbaum, R.; Zack, J.A.; et al. Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol. Immunity 2013, 38, 92–105. [Google Scholar] [CrossRef]
  68. Raftery, N.; Stevenson, N.J. Advances in anti-viral immune defence: Revealing the importance of the IFN JAK/STAT pathway. Cell Mol. Life Sci. 2017, 74, 2525–2535. [Google Scholar] [CrossRef]
  69. Minegishi, Y.; Saito, M.; Morio, T.; Watanabe, K.; Agematsu, K.; Tsuchiya, S.; Takada, H.; Hara, T.; Kawamura, N.; Ariga, T.; et al. Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple cytokine signals involved in innate and acquired immunity. Immunity 2006, 25, 745–755. [Google Scholar] [CrossRef]
  70. Essers, M.A.; Offner, S.; Blanco-Bose, W.E.; Waibler, Z.; Kalinke, U.; Duchosal, M.A.; Trumpp, A. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature 2009, 458, 904–908. [Google Scholar] [CrossRef]
  71. González-Navajas, J.M.; Lee, J.; David, M.; Raz, E. Immunomodulatory functions of type I interferons. Nat. Rev. Immunol. 2012, 12, 125–135. [Google Scholar] [CrossRef] [PubMed]
  72. Seth, R.B.; Sun, L.; Ea, C.K.; Chen, Z.J. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 2005, 122, 669–682. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Liu, S.; Chen, J.; Cai, X.; Wu, J.; Chen, X.; Wu, Y.T.; Sun, L.; Chen, Z.J. MAVS recruits multiple ubiquitin E3 ligases to activate antiviral signaling cascades. Elife 2013, 2, e00785. [Google Scholar] [CrossRef]
  75. 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]
  76. Kawasaki, T.; Kawai, T.; Akira, S. Recognition of Nucleic. acids by pattern-recognition receptors and its relevance in autoimmunity. Immunol. Rev. 2011, 243, 61–73. [Google Scholar] [CrossRef] [PubMed]
  77. Ren, Z.; Ding, T.; Zuo, Z.; Xu, Z.; Deng, J.; Wei, Z. Regulation of MAVS Expression and Signaling Function in the Antiviral Innate Immune Response. Front. Immunol. 2020, 11, 1030. [Google Scholar] [CrossRef]
  78. Guettler, S.; LaRose, J.; Petsalaki, E.; Gish, G.; Scotter, A.; Pawson, T.; Rottapel, R.; Sicheri, F. Structural basis and sequence rules for substrate recognition by Tankyrase explain the basis for cherubism disease. Cell 2011, 147, 1340–1354. [Google Scholar] [CrossRef]
  79. DaRosa, P.A.; Wang, Z.; Jiang, X.; Pruneda, J.N.; Cong, F.; Klevit, R.E.; Xu, W. Allosteric activation of the RNF146 ubiquitin ligase by a poly(ADP-ribosyl)ation signal. Nature 2015, 517, 223–226. [Google Scholar] [CrossRef] [PubMed]
  80. Diani-Moore, S.; Ram, P.; Li, X.; Mondal, P.; Youn, D.Y.; Sauve, A.A.; Rifkind, A.B. Identification of the aryl hydrocarbon receptor target gene TiPARP as a mediator of suppression of hepatic gluconeogenesis by 2,3,7,8-tetrachlorodibenzo-p-dioxin and of nicotinamide as a corrective agent for this effect. J. Biol. Chem. 2010, 285, 38801–38810. [Google Scholar] [CrossRef]
  81. Linden, J.; Lensu, S.; Tuomisto, J.; Pohjanvirta, R. Dioxins, the aryl hydrocarbon receptor and the central regulation of energy balance. Front. Neuroendocr. 2010, 31, 452–478. [Google Scholar] [CrossRef]
  82. Denison, M.S.; Nagy, S.R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharm. Toxicol. 2003, 43, 309–334. [Google Scholar] [CrossRef]
  83. Stockinger, B.; Di Meglio, P.; Gialitakis, M.; Duarte, J.H. The aryl hydrocarbon receptor: Multitasking in the immune system. Annu. Rev. Immunol. 2014, 32, 403–432. [Google Scholar] [CrossRef]
  84. Palavalli Parsons, L.H.; Challa, S.; Gibson, B.A.; Nandu, T.; Stokes, M.S.; Huang, D.; Lea, J.S.; Kraus, W.L. Identification of PARP-7 substrates reveals a role for MARylation in microtubule control in ovarian cancer cells. Elife 2021, 10, e60481. [Google Scholar] [CrossRef]
  85. Gomez, A.; Bindesbøll, C.; Satheesh, S.V.; Grimaldi, G.; Hutin, D.; MacPherson, L.; Ahmed, S.; Tamblyn, L.; Cho, T.; Nebb, H.I.; et al. Characterization of TCDD-inducible poly-ADP-ribose polymerase (TIPARP/ARTD14) catalytic activity. BioChem. J. 2018, 475, 3827–3846. [Google Scholar] [CrossRef]
  86. Stark, G.R.; Darnell, J.E., Jr. The JAK-STAT pathway at twenty. Immunity 2012, 36, 503–514. [Google Scholar] [CrossRef]
  87. Begitt, A.; Cavey, J.; Droescher, M.; Vinkemeier, U. On the role of STAT1 and STAT6 ADP-ribosylation in the regulation of macrophage activation. Nat. Commun. 2018, 9, 2144. [Google Scholar] [CrossRef]
  88. Caprara, G.; Prosperini, E.; Piccolo, V.; Sigismondo, G.; Melacarne, A.; Cuomo, A.; Boothby, M.; Rescigno, M.; Bonaldi, T.; Natoli, G. PARP14 Controls the Nuclear Accumulation of a Subset of Type I IFN-Inducible Proteins. J. Immunol. 2018, 200, 2439–2454. [Google Scholar] [CrossRef]
  89. Kumar, K.G.; Krolewski, J.J.; Fuchs, S.Y. Phosphorylation and specific ubiquitin acceptor sites are required for ubiquitination and degradation of the IFNAR1 subunit of type I interferon receptor. J. Biol. Chem. 2004, 279, 46614–46620. [Google Scholar] [CrossRef]
  90. Fuchs, S.Y.; Spiegelman, V.S.; Kumar, K.G. The many faces of beta-TrCP E3 ubiquitin ligases: Reflections in the magic mirror of cancer. Oncogene 2004, 23, 2028–2036. [Google Scholar] [CrossRef]
  91. Takeyama, K.; Aguiar, R.C.; Gu, L.; He, C.; Freeman, G.J.; Kutok, J.L.; Aster, J.C.; Shipp, M.A. The BAL-binding protein BBAP and related Deltex family members exhibit ubiquitin-protein isopeptide ligase activity. J. Biol. Chem. 2003, 278, 21930–21937. [Google Scholar] [CrossRef]
  92. Juszczynski, P.; Kutok, J.L.; Li, C.; Mitra, J.; Aguiar, R.C.; Shipp, M.A. BAL1 and BBAP are regulated by a gamma interferon-responsive bidirectional promoter and are overexpressed in diffuse large B-cell lymphomas with a prominent inflammatory infiltrate. Mol. Cell Biol. 2006, 26, 5348–5359. [Google Scholar] [CrossRef]
  93. Zhang, Y.; Mao, D.; Roswit, W.T.; Jin, X.; Patel, A.C.; Patel, D.A.; Agapov, E.; Wang, Z.; Tidwell, R.M.; Atkinson, J.J.; et al. PARP9-DTX3L ubiquitin ligase targets host histone H2BJ. and viral 3C protease to enhance interferon signaling and control viral infection. Nat. Immunol. 2015, 16, 1215–1227. [Google Scholar] [CrossRef]
  94. Zhu, Y.; Chen, G.; Lv, F.; Wang, X.; Ji, X.; Xu, Y.; Sun, J.; Wu, L.; Zheng, Y.T.; Gao, G. Zinc-finger antiviral protein inhibits HIV-1 infection by selectively targeting multiply spliced viral mRNAs for degradation. Proc. Natl. Acad. Sci. USA 2011, 108, 15834–15839. [Google Scholar] [CrossRef]
  95. Luo, X.; Wang, X.; Gao, Y.; Zhu, J.; Liu, S.; Gao, G.; Gao, P. Molecular Mechanism of RNA Recognition by Zinc-Finger Antiviral Protein. Cell Rep. 2020, 30, 46–52.e4. [Google Scholar] [CrossRef]
  96. Hayakawa, S.; Shiratori, S.; Yamato, H.; Kameyama, T.; Kitatsuji, C.; Kashigi, F.; Goto, S.; Kameoka, S.; Fujikura, D.; Yamada, T.; et al. ZAPS is a potent stimulator of signaling mediated by the RNA helicase RIG-I during antiviral responses. Nat. Immunol. 2011, 12, 37–44. [Google Scholar] [CrossRef]
  97. Guo, X.; Ma, J.; Sun, J.; Gao, G. The zinc-finger antiviral protein recruits the RNA processing exosome to degrade the target mRNA. Proc. Natl. Acad. Sci. USA 2007, 104, 151–156. [Google Scholar] [CrossRef]
  98. Kennedy, G.; Sugden, B. EBNA-1, a bifunctional transcriptional activator. Mol. Cell Biol. 2003, 23, 6901–6908. [Google Scholar] [CrossRef]
  99. Hayman, I.R.; Temple, R.M.; Burgess, C.K.; Ferguson, M.; Liao, J.; Meyers, C.; Sample, C.E. New insight into Epstein-Barr Virus infection using models of stratified epithelium. PLoS Pathog. 2023, 19, e1011040. [Google Scholar] [CrossRef]
  100. Imai, S.; Koizumi, S.; Sugiura, M.; Tokunaga, M.; Uemura, Y.; Yamamoto, N.; Tanaka, S.; Sato, E.; Osato, T. Gastric carcinoma: Monoclonal epithelial malignant cells expressing Epstein-Barr virus latent infection protein. Proc. Natl. Acad. Sci. USA 1994, 91, 9131–9135. [Google Scholar] [CrossRef]
  101. Hu, J.; Garber, A.C.; Renne, R. The latency-associated nuclear antigen of Kaposi’s sarcoma-associated herpesvirus supports latent DNA replication in dividing cells. J. Virol. 2002, 76, 11677–11687. [Google Scholar] [CrossRef]
  102. Chang, Y.; Moore, P.S. Kaposi’s Sarcoma (KS)-associated herpesvirus and its role in KS. Infect. Agents Dis. 1996, 5, 215–222. [Google Scholar]
  103. Campos, G.C.; Sardi, S.I.; Sarno, M.; Brites, C. Zika virus infection, a new public health challenge. Braz. J. Infect. Dis. 2016, 20, 227–228. [Google Scholar] [CrossRef]
  104. Ong, C.W. Zika virus: An emerging infectious threat. Intern. Med. J. 2016, 46, 525–530. [Google Scholar] [CrossRef]
  105. Akey, D.L.; Brown, W.C.; Dutta, S.; Konwerski, J.; Jose, J.; Jurkiw, T.J.; DelProposto, J.; Ogata, C.M.; Skiniotis, G.; Kuhn, R.J.; et al. Flavivirus NS1 structuRes. reveal surfaces for associations with membranes and the immune system. Science 2014, 343, 881–885. [Google Scholar] [CrossRef]
  106. Sironi, M.; Forni, D.; Clerici, M.; Cagliani, R. Nonstructural Proteins Are Preferential Positive Selection Targets in Zika Virus and Related Flaviviruses. PLoS Negl. Trop. Dis. 2016, 10, e0004978. [Google Scholar] [CrossRef]
  107. Silva, L.A.; Dermody, T.S. Chikungunya virus: Epidemiology, replication, disease mechanisms, and prospective intervention strategies. J. Clin. Investig. 2017, 127, 737–749. [Google Scholar] [CrossRef]
  108. Kril, V.; Aïqui-Reboul-Paviet, O.; Briant, L.; Amara, A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu. Rev. Virol. 2021, 8, 327–347. [Google Scholar] [CrossRef]
  109. Göertz, G.P.; McNally, K.L.; Robertson, S.J.; Best, S.M.; Pijlman, G.P.; Fros, J.J. The Methyltransferase-Like Domain of Chikungunya Virus nsP2 Inhibits the Interferon Response by Promoting the Nuclear Export of STAT1. J. Virol. 2018, 92. [Google Scholar] [CrossRef]
  110. Utt, A.; Das, P.K.; Varjak, M.; Lulla, V.; Lulla, A.; Merits, A. Mutations conferring a noncytotoxic phenotype on chikungunya virus replicons compromise enzymatic properties of nonstructural protein 2. J. Virol. 2015, 89, 3145–3162. [Google Scholar] [CrossRef]
  111. Webb, L.G.; Veloz, J.; Pintado-Silva, J.; Zhu, T.; Rangel, M.V.; Mutetwa, T.; Zhang, L.; Bernal-Rubio, D.; Figueroa, D.; Carrau, L.; et al. Chikungunya virus antagonizes cGAS-STING mediated type-I interferon responses by degrading cGAS. PLoS Pathog. 2020, 16, e1008999. [Google Scholar] [CrossRef] [PubMed]
  112. Abraham, R.; Hauer, D.; McPherson, R.L.; Utt, A.; Kirby, I.T.; Cohen, M.S.; Merits, A.; Leung, A.K.L.; Griffin, D.E. ADP-ribosyl-binding and hydrolase activities of the alphavirus nsP3 macrodomain are critical for initiation of virus replication. Proc. Natl. Acad. Sci. USA 2018, 115, E10457–E10466. [Google Scholar] [CrossRef] [PubMed]
  113. McPherson, R.L.; Abraham, R.; Sreekumar, E.; Ong, S.E.; Cheng, S.J.; Baxter, V.K.; Kistemaker, H.A.; Filippov, D.V.; Griffin, D.E.; Leung, A.K. ADP-ribosylhydrolase activity of Chikungunya virus macrodomain is critical for virus replication and virulence. Proc. Natl. Acad. Sci. USA 2017, 114, 1666–1671. [Google Scholar] [CrossRef] [PubMed]
  114. Grunewald, M.E.; Fehr, A.R.; Athmer, J.; Perlman, S. The coronavirus nucleocapsid protein is ADP-ribosylated. Virology 2018, 517, 62–68. [Google Scholar] [CrossRef]
  115. Déry, C.V.; de Murcia, G.; Lamarre, D.; Morin, N.; Poirier, G.G.; Weber, J. Possible role of ADP-ribosylation of adenovirus core proteins in virus infection. Virus Res. 1986, 4, 313–329. [Google Scholar] [CrossRef]
  116. Suskiewicz, M.J.; Palazzo, L.; Hughes, R.; Ahel, I. Progress and outlook in studying the substrate specificities of PARPs and related enzymes. FEBS J. 2021, 288, 2131–2142. [Google Scholar] [CrossRef]
  117. Weixler, L.; Schäringer, K.; Momoh, J.; Lüscher, B.; Feijs, K.L.H.; Žaja, R. ADP-ribosylation of RNA and DNA: From in vitro characterization to in vivo function. Nucleic. Acids Res. 2021, 49, 3634–3650. [Google Scholar] [CrossRef]
  118. Schuller, M.; Butler, R.E.; Ariza, A.; Tromans-Coia, C.; Jankevicius, G.; Claridge, T.D.W.; Kendall, S.L.; Goh, S.; Stewart, G.R.; Ahel, I. Molecular basis for DarT ADP-ribosylation of a DNA base. Nature 2021, 596, 597–602. [Google Scholar] [CrossRef]
  119. Munnur, D.; Bartlett, E.; Mikolčević, P.; Kirby, I.T.; Rack, J.G.M.; Mikoč, A.; Cohen, M.S.; Ahel, I. Reversible ADP-ribosylation of RNA. Nucleic. Acids Res. 2019, 47, 5658–5669. [Google Scholar] [CrossRef]
  120. Kim, D.S.; Challa, S.; Jones, A.; Kraus, W.L. PARPs and ADP-ribosylation in RNA biology: From RNA expression and processing to protein translation and proteostasis. Genes Dev. 2020, 34, 302–320. [Google Scholar] [CrossRef]
  121. Jankevicius, G.; Ariza, A.; Ahel, M.; Ahel, I. The Toxin-Antitoxin System DarTG Catalyzes Reversible ADP-Ribosylation of DNA. Mol. Cell 2016, 64, 1109–1116. [Google Scholar] [CrossRef] [PubMed]
  122. Matta, E.; Kiribayeva, A.; Khassenov, B.; Matkarimov, B.T.; Ishchenko, A.A. Insight into DNA substrate specificity of PARP1-catalysed DNA poly(ADP-ribosyl)ation. Sci. Rep. 2020, 10, 3699. [Google Scholar] [CrossRef]
  123. Talhaoui, I.; Lebedeva, N.A.; Zarkovic, G.; Saint-Pierre, C.; Kutuzov, M.M.; Sukhanova, M.V.; Matkarimov, B.T.; Gasparutto, D.; Saparbaev, M.K.; Lavrik, O.I.; et al. Poly(ADP-ribose) polymerases covalently modify strand break termini in DNA fragments in vitro. Nucleic. Acids Res. 2016, 44, 9279–9295. [Google Scholar] [CrossRef] [PubMed]
  124. Zarkovic, G.; Belousova, E.A.; Talhaoui, I.; Saint-Pierre, C.; Kutuzov, M.M.; Matkarimov, B.T.; Biard, D.; Gasparutto, D.; Lavrik, O.I.; Ishchenko, A.A. Characterization of DNA ADP-ribosyltransferase activities of PARP2 and PARP3: New insights into DNA ADP-ribosylation. Nucleic. Acids Res. 2018, 46, 2417–2431. [Google Scholar] [CrossRef]
  125. Shaw, A.E.; Hughes, J.; Gu, Q.; Behdenna, A.; Singer, J.B.; Dennis, T.; Orton, R.J.; Varela, M.; Gifford, R.J.; Wilson, S.J.; et al. Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses. PLoS Biol. 2017, 15, e2004086. [Google Scholar] [CrossRef]
  126. Eckei, L.; Krieg, S.; Bütepage, M.; Lehmann, A.; Gross, A.; Lippok, B.; Grimm, A.R.; Kümmerer, B.M.; Rossetti, G.; Lüscher, B.; et al. The conserved macrodomains of the non-structural proteins of Chikungunya virus and other pathogenic positive strand RNA viruses function as mono-ADP-ribosylhydrolases. Sci. Rep. 2017, 7, 41746. [Google Scholar] [CrossRef] [PubMed]
  127. Schoggins, J.W. Interferon-stimulated genes: Roles in viral pathogenesis. Curr. Opin. Virol. 2014, 6, 40–46. [Google Scholar] [CrossRef]
  128. Karki, S.; Li, M.M.; Schoggins, J.W.; Tian, S.; Rice, C.M.; MacDonald, M.R. Multiple interferon stimulated genes synergize with the zinc finger antiviral protein to mediate anti-alphavirus activity. PLoS ONE 2012, 7, e37398. [Google Scholar] [CrossRef]
  129. Kuri, T.; Eriksson, K.K.; Putics, A.; Züst, R.; Snijder, E.J.; Davidson, A.D.; Siddell, S.G.; Thiel, V.; Ziebuhr, J.; Weber, F. The ADP-ribose-1″-monophosphatase domains of severe acute respiratory syndrome coronavirus and human coronavirus 229E mediate resistance to antiviral interferon responses. J. Gen. Virol. 2011, 92, 1899–1905. [Google Scholar] [CrossRef]
  130. Park, E.; Griffin, D.E. The nsP3 macro domain is important for Sindbis virus replication in neurons and neurovirulence in mice. Virology 2009, 388, 305–314. [Google Scholar] [CrossRef]
  131. Fu, W.; Yao, H.; Butepage, M.; Zhao, Q.; Luscher, B.; Li, J. The search for inhibitors of macrodomains for targeting the readers and erasers of mono-ADP-ribosylation. Drug. Discov. Today 2021, 26, 2547–2558. [Google Scholar] [CrossRef] [PubMed]
  132. Alhammad, Y.M.O.; Fehr, A.R. The Viral Macrodomain Counters Host Antiviral ADP-Ribosylation. Viruses 2020, 12, 384. [Google Scholar] [CrossRef] [PubMed]
  133. Grunewald, M.E.; Chen, Y.; Kuny, C.; Maejima, T.; Lease, R.; Ferraris, D.; Aikawa, M.; Sullivan, C.S.; Perlman, S.; Fehr, A.R. The coronavirus macrodomain is required to prevent PARP-mediated inhibition of virus replication and enhancement of IFN expression. PLoS Pathog. 2019, 15, e1007756. [Google Scholar] [CrossRef] [PubMed]
  134. Li, H.; Liu, Z.Y.; Wu, N.; Chen, Y.C.; Cheng, Q.; Wang, J. PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol. Cancer 2020, 19, 107. [Google Scholar] [CrossRef]
  135. Curtin, N.J.; Szabo, C. Poly(ADP-ribose) polymerase inhibition: Past, present and future. Nat. Rev. Drug. Discov. 2020, 19, 711–736. [Google Scholar] [CrossRef]
Figure 1. ADP-ribosylation in IFN-I production and IFN-I-induced JAK-STAT signaling pathway. During viral infection, (1) TNKS1 and TNKS2 interact with MAVS and catalyze its PARylation, leading to the degradation of MAVS and impairment of IFN production. (2) PARP7 targets TBK1 and catalyzes it mono-ADP-ribosylation, which suppresses the activation of TBK1 and downstream events. (3) PARP11 mono-ADP-ribosylates β-TrCP and mediates IFNAR1 ubiquitination and degradation, thus acting as a negative regulator of IFN-I response. (4) PARP14 catalyzes mono-ADP-ribosylation of STAT1 and prevents its phosphorylation, thus repressing the IFN-I response. PARP9 suppresses PARP14-mediated MARylation of STAT1, which sustains STAT1 phosphorylation. (Created with BioRender.com (accessed on 16 January 2023).
Figure 1. ADP-ribosylation in IFN-I production and IFN-I-induced JAK-STAT signaling pathway. During viral infection, (1) TNKS1 and TNKS2 interact with MAVS and catalyze its PARylation, leading to the degradation of MAVS and impairment of IFN production. (2) PARP7 targets TBK1 and catalyzes it mono-ADP-ribosylation, which suppresses the activation of TBK1 and downstream events. (3) PARP11 mono-ADP-ribosylates β-TrCP and mediates IFNAR1 ubiquitination and degradation, thus acting as a negative regulator of IFN-I response. (4) PARP14 catalyzes mono-ADP-ribosylation of STAT1 and prevents its phosphorylation, thus repressing the IFN-I response. PARP9 suppresses PARP14-mediated MARylation of STAT1, which sustains STAT1 phosphorylation. (Created with BioRender.com (accessed on 16 January 2023).
Pathogens 12 00303 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Du, Q.; Miao, Y.; He, W.; Zheng, H. ADP-Ribosylation in Antiviral Innate Immune Response. Pathogens 2023, 12, 303. https://doi.org/10.3390/pathogens12020303

AMA Style

Du Q, Miao Y, He W, Zheng H. ADP-Ribosylation in Antiviral Innate Immune Response. Pathogens. 2023; 12(2):303. https://doi.org/10.3390/pathogens12020303

Chicago/Turabian Style

Du, Qian, Ying Miao, Wei He, and Hui Zheng. 2023. "ADP-Ribosylation in Antiviral Innate Immune Response" Pathogens 12, no. 2: 303. https://doi.org/10.3390/pathogens12020303

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