Next Article in Journal
Japanese Encephalitis Enzootic and Epidemic Risks across Australia
Previous Article in Journal
Longitudinal IgA and IgG Response, and ACE2 Binding Blockade, to Full-Length SARS-CoV-2 Spike Protein Variants in a Population of Black PLWH Vaccinated with ChAdOx1 nCoV-19
Previous Article in Special Issue
RNA-Binding Proteins as Regulators of Internal Initiation of Viral mRNA Translation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pro-Viral and Anti-Viral Roles of the RNA-Binding Protein G3BP1

by
Aravinth Kumar Jayabalan
1,
Diane E. Griffin
2 and
Anthony K. L. Leung
1,3,4,5,*
1
Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
2
W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
3
McKusick-Nathans Department of Genetics Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
4
Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
5
Department of Molecular Biology and Genetics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(2), 449; https://doi.org/10.3390/v15020449
Submission received: 9 December 2022 / Revised: 21 January 2023 / Accepted: 30 January 2023 / Published: 6 February 2023

Abstract

:
Viruses depend on host cellular resources to replicate. Interaction between viral and host proteins is essential for the pathogens to ward off immune responses as well as for virus propagation within the infected cells. While different viruses employ unique strategies to interact with diverse sets of host proteins, the multifunctional RNA-binding protein G3BP1 is one of the common targets for many viruses. G3BP1 controls several key cellular processes, including mRNA stability, translation, and immune responses. G3BP1 also serves as the central hub for the protein–protein and protein–RNA interactions within a class of biomolecular condensates called stress granules (SGs) during stress conditions, including viral infection. Increasing evidence suggests that viruses utilize distinct strategies to modulate G3BP1 function—either by degradation, sequestration, or redistribution—and control the viral life cycle positively and negatively. In this review, we summarize the pro-viral and anti-viral roles of G3BP1 during infection among different viral families.

1. Introduction

Eukaryotic cells reprogram a variety of cellular processes and pathways in response to unfavorable growth conditions, including viral infection. When stressed, cells selectively transcribe and translate stress-responsive genes while arresting global protein synthesis to conserve energy [1,2,3,4]. Upon viral entry, cells trigger innate response pathways to eliminate or prevent viral replication [5]. The first line of host defense against viral infection is the recognition of viral RNA or DNA by pathogen-associated molecular patterns (PAMPs) through different sensors [6]. For example, viral nucleic acids can be detected by retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), Toll-like receptors (TLRs), and cGMP/cAMP synthase (cGAS) [7,8,9,10,11,12]. These proteins detect and distinguish viral vs. cellular nucleic acids based on the structural differences, such as the presence of a 5′-triphosphate (5′-PPP) moiety in viral RNAs [13]. In turn, viruses have developed adaptive mechanisms to counteract host innate responses for successful propagation [14]. Even the most complex virus encodes only a few hundred proteins; thus, viruses must interact with host proteins to exploit their cellular machinery for efficient viral replication [14,15,16].
Among the host proteins, ribosomes and translation factors are critical for viral protein synthesis and mRNA binding proteins to stabilize viral RNA, and some proteins are involved in the viral replication complexes. The Ras GTPase-activating (SH3 domain) protein-binding protein G3BP1 is an RNA-binding protein targeted by many viruses [17]. G3BP1 is highly conserved from yeast to humans and plays a key role in RNA metabolism, such as regulating the stability and translation of a subset of mRNAs, binding to specific transcripts during stress conditions, and activating interferon (IFN)-stimulated gene expression [18,19,20,21]. G3BP1 function is further modulated by post-translational modifications and also implicated in many diseases such as cancer, neurodegeneration, and viral infection—the latter is the focus of this review [22,23,24,25,26].
G3BP1 and its paralog G3BP2 possess similar protein domain architecture composed of regions for protein, RNA and DNA interactions (Figure 1). These domains include N-terminal nuclear transport factor 2-like (NTF2L), followed by an acidic domain, proline-rich region (PxxP), an RNA recognition motif (RRM), and an arginine and glycine-rich region (RGG) at the C-terminus [27,28]. The N-terminal domains contain motifs for protein binding while the C-terminal domains bind nucleic acids. For instance, the NTF2L domain interacts with FGDF sequence motifs present in many proteins and the PxxP domain interacts with SH3 motifs. The RRM domain is essential for RNA binding and RGG domain increases RNA binding as well as mediates protein–protein interactions. The functions of G3BP1 and G3BP2 are redundant in some, but not all cases [29]. For example, knockdown of G3BP1 increases the expression of G3BP2 to compensate for the loss of G3BP1 expression and function, but not vice versa [23,29].
Emergent data suggest that G3BP1 is one of the key proteins that impact the viral life cycle both positively and negatively [30,31]. For example, some viruses require G3BP1 for efficient viral replication, while the replication of other viruses is negatively affected by G3BP1 expression. Well-studied functions of G3BP1 include participation in the innate response to infection through activation of viral nucleic acid sensors RIG-I and cGAS and as an essential component of stress granules (SGs) [29,30,32,33,34,35]. SGs are a class of cytoplasmic biomolecular condensates enriched with mRNA–protein complexes assembled in response to stress, including viral infection. Knockout of both G3BP1 and G3BP2 is necessary to completely inhibit SG formation induced by many stressors, implying their importance to form protein–protein and protein–RNA interactions within SGs [29]. Given that G3BP1 impacts the life cycle of many viruses, it is important to distinguish what specific functional roles are played by G3BP1 at different infection stages. Here, we focus on the role of G3BP1 during viral infection and the various strategies utilized by viruses to exploit or counteract G3BP1 functions.

2. G3BP1 Amplifies Innate Immune Signaling

G3BP1 regulates early innate responses through its ability to bind nucleic acids and proteins in these signaling pathways. G3BP1 facilitates the initial IFN-β response by binding and augmenting the function of viral RNA and DNA sensing proteins in the cytoplasm. For example, the RGG domain of G3BP1 binds the RIG-I helicase domain and viral dsRNA to prevent RIG-I degradation and enhance Ifn-b mRNA expression [30]. G3BP1 also interacts with cGAS to form large complexes that enhance interaction with viral DNA to induce IFN [30,34,36]. The binding of the N-terminal region of cGAS with G3BP1 is required for activation of the RNA-dependent protein kinase PKR leading to co-localization as small cytoplasmic foci [35]. The local concentration of G3BP1, cGAS, and PKR within cytoplasmic foci is proposed to enhance PKR activity. G3BP1 is required for activation of cGAS as depletion of G3BP1 dramatically reduces DNA-induced IFN production [34]. These data suggest that G3BP1-mediated interactions with signaling molecules facilitate the initiation of downstream innate response pathway signaling. G3BP1 can also facilitate the expression and translation of IFN-stimulated genes (ISGs). For example, G3BP1, G3BP2, and their binding partner Caprin1 positively regulate the translation of multiple ISGs in response to IFN (Figure 2a) [21].

3. G3BP1 Forms the Core of Stress Granules

A well-known function of G3BP1 during stress stimuli is its involvement in SG assembly (Figure 2b). SGs assemble upon stress-induced translation arrest that results in the accumulation of free untranslated mRNAs and mRNA binding proteins in the cytoplasm [37]. Upon stress, one of the following five kinases is activated: double-stranded RNA (dsRNA)-dependent PKR, protein kinase R-like endoplasmic reticulum kinase (PERK), heme-regulated inhibitor (HRI), general control nonderepressible-2 (GCN2), or microtubule affinity-regulating kinase 2 (MARK2). The activated kinase phosphorylates the translation initiation factor eIF2α subunit [38,39]. The initiation complex containing the phosphorylated eIF2α becomes unavailable for recycling, which leads to a stall in translation initiation [40]. Once the elongating ribosomes run off from polysomes, the stalled translation initiation results in a sudden influx of untranslated mRNAs in the cytoplasm. These untranslated mRNAs bound with ribonucleoproteins (mRNPs) cluster together with G3BP1, G3BP2, and several other RNA binding proteins to form RNA/protein-rich condensates in the cytoplasm. Several post-translational modifications (PTMs) control the dynamics of SGs [33,41,42,43,44,45,46]. Of note, G3BP1 itself undergoes several post-translational modifications, including phosphorylation, methylation, ubiquitination, and ADP-ribosylation during stress conditions [42,44,47]. G3BP1 and G3BP2 form the core protein–protein interaction network which binds translation factors, mRNA binding proteins, long and translationally incompetent mRNAs as well as specific transcripts during stress conditions [19,48,49]. RNAi-mediated knockdown or genetic knockout of both G3BP1 and G3BP2 results in cells that do not form SGs in response to most stressors, emphasizing the importance of G3BP1/G3BP2 in SG assembly [29]. These data suggest that G3BP1/G3BP2 act as a scaffold that recruits other proteins into SGs [47,50].
Recent studies provided more mechanistic insights into the role of G3BP1 as the scaffold for SG assembly. G3BP1 is the central node of the protein–protein interaction network within SGs, where RNA serves as a molecular trigger for phase separation. In the normal state, G3BP1 adopts a compact, closed auto-inhibitory state with intramolecular interaction between acidic and arginine-rich regions. Increasing local RNA concentrations can increase their binding to G3BP1, relieving G3BP1 from the autoinhibitory state and resulting in more RNA recruitment and less aberrant RNA–RNA interaction [50,51,52]. In light of the viral infection, it may be possible that at the early infection stage, an increase in viral RNA concentration may relieve G3BP1 from its auto-inhibitory state, triggering G3BP1 condensation, with the protein serving as the scaffold to recruit other proteins/RNA required for viral replication.

4. Role of G3BP1 during Viral Infection

At least four distinct temporal patterns of SGs have been observed during infection with different viruses: (1) stable, (2) transient, (3) oscillating, or (4) no SGs (active blocking of SG induction) [53,54,55]. These patterns highlight the importance of modulating SGs upon virus infection (Table 1). As demonstrated by one of the earliest studies using Semliki Forest virus (SFV), infection-induced translation arrest results in the assembly of transient SGs. Given that some of the key anti-viral proteins (such as ADAR1, PARP13/ZAP, OAS, IRF3, IRF7, TBK1, and RNase L) localize to SGs and activate the anti-viral response, SGs may negatively affect viral replication [56,57,58,59,60]. Thus, viruses may target SGs not only to modulate G3BP1 function but also to disassemble SGs to counteract anti-viral mechanisms in the infected cells. On the other hand, the presence of SGs may facilitate the switch from viral translation to genome amplification by sequestering translation factors into the condensed state (Figure 3) [23].
G3BP1 can either promote or inhibit the viral life cycle, but the molecular mechanisms are not completely understood. One way by which G3BP1 may regulate its function during infection is through post-translational modifications. For example, ADP-ribosylated G3BP1 is crucial for SG assembly, given that chikungunya virus (CHIKV) actively reduces G3BP1 ADP-ribosylation, disrupting SGs with translation factors released from the condensed state [47]. However, G3BP1 is also required for establishing CHIKV replication complexes, and post-translational modifications required for this process have not been defined [23]. Thus, post-translationally modified G3BP1 may act as an anti-viral protein, while the unmodified or differently modified G3BP1 could play a pro-viral role during CHIKV infection. Similarly, arginine methylation status of G3BP1 partly recruits Tudor domain-containing protein (TDRD3) to regulate innate immune response against enterovirus species [60]. Using polyinosinic:polycytidylic acid as a mimic for viral replication double-stranded intermediates, Kim et al. showed that G3BP1 is phosphorylated at tyrosine residue 40 by Bruton’s tyrosine kinase (BTK). Phosphorylation of G3BP1 by BTK is critical for dimerization and phase separation to limit viral spread [88]. Further clues are obtained from identifying the strategies that viruses employ to target G3BP1—i.e., sequestration, cleavage, or degradation—and modulate its function during infection (Figure 4). In general, viruses use their genome-encoded proteins to sequester G3BP1 if it is required for viral replication. On the contrary, viruses cleave G3BP1 when it exerts an anti-viral role (Table 2). Here, we describe the role of G3BP1 during specific viral infections and the ways by which G3BP1 is targeted.
G3BP1 controls several key cellular processes, including mRNA metabolism, ribosomal quality control, immune response, and SG assembly. G3BP1 facilitates replication of some viruses whereas it restricts viral spread in other viruses. Depending on the virus type, G3BP1 plays different roles in the viral life cycle, including (1) stabilizing viral RNA; (2) recruiting ribosomes to initiate viral RNA translation; (3) serving as a scaffold to build viral replication complexes; (4) assembling SGs; (5) stabilizing and enhancing the IFN-β response; and (6) activating cGAS and RIG-I pathways. Here, we explore the mechanisms by which different viruses regulate G3BP1 function during infection.

4.1. G3BP1 as an Anti-Viral Factor

G3BP1 enhances the immune response against invading viruses by inducing the expression of IFN and translation of ISGs. In virus-infected cells, G3BP1 binds to dsRNA resulting from the viral replication intermediates through the RGG domain and elevates RIG-I induced IFN-ꞵ mRNA expression [30]. In response to IFN, G3BP1, G3BP2, and Caprin 1 promote ISG translation to synthesize anti-viral factors. G3BP1 also assembles SGs in response to infection-mediated translation arrest and limits the availability of translation factors for viral protein synthesis. To counter these anti-viral functions, viruses target G3BP1—either cleaving or sequestering it during infection. Here we summarize the strategies shared by viruses in the Picornaviridae and Coronaviridae families to counteract host defense.

4.1.1. Picornaviruses Inhibit G3BP1 Function through Protease-Mediated Cleavage

Picornaviruses are small, non-enveloped, positive-sense RNA viruses that include EMCV, EV, PV, FMDV, and CVB3. G3BP1 negatively regulates Picornaviridae infection by binding the 3′ untranslated regions (3′UTRs) or Internal Ribosome Entry Site (IRES) regions of viral RNAs to inhibit viral replication [53,95]. To counteract these effects, the 3C protease or leader protein of these viruses cleaves G3BP1 between Q325 and Q326 at late infection stages. G3BP1 cleavage is essential for the expression of viral proteins, and expression of the non-cleavable G3BP1 Q325E mutant suppresses viral replication [61,62,63,64,65,66,67,68,69].
During Picornaviruses infection, PKR is activated by double-stranded RNA intermediates, phosphorylates eIF2α, and induces SG formation. SGs are observed early in infection but disassembled at late stages of infection. SG disassembly correlates temporally with G3BP1 cleavage, and the expression of the non-cleavable G3BP1 Q325E mutant results in sustained IFN-ꞵ mRNA expression and SG persistence. Consistent with the observed effect on G3BP1 cleavage, knockdown of G3BP1 increases viral protein synthesis and virus production, whereas G3BP1 overexpression shows an inhibitory effect [61,64,65,66].
FMDV, which uses the leader protein to cleave G3BP1, also causes SG disassembly. Catalytic mutant C41A or leaderless FMDV inefficiently inhibits arsenite-induced SG formation in infected cells [63]. FMDV infection also causes degradation of G3BP1 through autophagy [62]. Thus, protease-mediated cleavage or degradation of G3BP1 is the fundamental mechanism employed by Picornaviridae to restrict anti-viral activities of G3BP1.

4.1.2. Coronaviruses Modulate G3BP1 Function Either by Cleavage or Sequestration

Coronaviridae is a family of enveloped viruses with large single-stranded positive-sense RNA genomes and includes PEDV and SARS-CoV-2. Similar to Picornaviridae, G3BP1 negatively regulates coronavirus infection. Overexpression of G3BP1 reduces viral replication, protein synthesis, and virus production, while G3BP1 knockdown enhances viral replication. However, these viruses utilize different strategies to modulate G3BP1 function; for example, PEDV induces cleavage of G3BP1 between D168 and D169 by caspase-8 to regulate SG dynamics in infected cells. Expression of non-cleavable G3BP1 results in persistent SGs and reduces viral replication, but the mechanism behind G3BP1-mediated reduction of viral replication remains unexplored [79,80].
SARS-CoV-2—the virus responsible for the COVID-19 pandemic—modulates G3BP1 function by sequestration and condensation with the nucleocapsid protein. The intrinsically disordered region (IDR) at the N-terminal region of nucleocapsid is required for condensation with G3BP1, and this IDR is also crucial for viral particle production. Though it is unclear whether SARS-CoV-2 induces SGs, G3BP1–nucleocapsid binding blocks the interaction of SG proteins such as PKR and USP10 with G3BP1 in arsenite-treated cells [82]. However, the role of G3BP1 remains unclear given the conflicting reports on the effect of G3BP1 knockdown in structural protein and viral RNA levels [81,82,83,96]. Recent studies highlighted the potential anti-viral role of G3BP1 in the lung tissue of COVID-19 patients and SARS-CoV-2-infected mice. In COVID-19 patients and mice, the N protein interacts with G3BP1, suppresses SG formation, and potentiates viral infection by antagonizing the G3BP1-mediated host innate immune response pathway [97,98].

4.2. G3BP1 as a Pro-Viral Factor

Although G3BP1 synergizes host immune responses to fight viral invasion, some viruses require G3BP1 for efficient infection. Such pro-viral activities have been reported for several viruses including CHIKV, hepatitis C virus, and respiratory syncytial virus [23,74,99]. For example, during CHIKV infection, depletion of both G3BP1 and G3BP2 significantly decreases viral RNA levels, protein expression, and subsequent viral progeny production [23]. Binding of the viral genome by G3BP1 and sequestering it into viral replication complexes may be important to prevent G3BP1-mediated activation of the innate immune response. The proposed pro-viral functions of G3BP1 include: stabilizing viral mRNAs to prevent degradation, facilitating viral protein translation by recruiting translation factors and ribosomes, acting as a scaffold to build viral replication complexes, and amplifying the viral genome. In this section, we will discuss the different strategies employed by viruses that use G3BP1 to promote viral replication.

4.2.1. Togaviruses Sequester G3BP1 to Facilitate Viral Replication and Translation

Togaviridae is a family of enveloped, icosahedral viruses with a positive-sense single-stranded RNA genome including the mosquito-borne alphaviruses that cause rash and arthritis such as CHIKV, SFV, and SINV. Notably, the alphavirus genus can be categorized into the Old World alphaviruses (CHIKV, SINV, SFV) and the New World alphaviruses (VEEV, EEEV) based on their geographical origin. Cells lacking both G3BP1/G3BP2 do not support replication of the Old World alphaviruses but do support replication of the New World encephalitic alphaviruses, indicating a virus-specific importance of G3BP1 for infection. In infected cells, G3BP1 is sequestered through interaction of the NTF2L domain with FGDF motifs in the C-terminal hypervariable region of non-structural protein 3 (nsP3). For initiation of infection, G3BP1 facilitates CHIKV, but not to SFV, translation of genomic RNA by enriching translation factors at cytopathic vacuoles [100]. The binding of nsP3 with G3BP1 is a proposed mechanism for SG disassembly where nsP3-G3BP1 binding inhibits the interaction between other SG components and G3BP1. However, SFV without FGDF motifs still disassembles SGs, but at a slower rate suggesting an additional mechanism for SG disassembly [77]. Recently, our group discovered that the SG disassembly for CHIKV is partly mediated by the macrodomain at the N-terminus of nsP3. The macrodomain possesses ADP-ribosylhydrolase activity (the ability to remove ADP-ribose from conjugated proteins), and this enzymatic activity is critical for replication and virulence of viruses from several viral families [92,101,102,103,104]. G3BP1 ADP-ribosylation, a key driver of SG assembly, is reduced in cells infected by wild-type CHIKV, but not a mutant virus that lacks hydrolase activity. Infection with the mutant virus results in delayed SG disassembly and reduced viral structural protein synthesis in neuronal cells, suggesting that G3BP1 ADP-ribosylation regulates virus production [47,105].
Notably, the New World alphaviruses do not have the FGDF motif and thus do not bind G3BP1. Instead, these viruses sequester other SG components, such as FXR1, FXR2, and FMR1, which bind to ribosomes. Hence, these proteins possibly recruit ribosomes and facilitate translation of New World viral genomic RNA similar to the G3BP1 role for Old World alphaviruses [78,106]. Therefore, although SG components are targeted by both Old World and New World alphaviruses, G3BP1 plays a pro-viral role only for Old World alphaviruses.

4.2.2. Caliciviruses Remodel the G3BP1 Interactome during Infection

The Caliciviridae family comprises small non-enveloped positive-strand RNA viruses, including human norovirus (HuNoV) and the closely related murine norovirus (MNV). MNV remodels G3BP1 interactome in infected cells and induces cytoplasmic G3BP1 granules during infection that are distinct from SGs. The proteome of G3BP1 granules contains several proteins that are present in viral replication complexes, suggesting a pro-viral role of G3BP1 during MNV infection. Consistently, genetic depletion of G3BP1 severely affects viral replication and translation. In addition, knockdown of MNV-induced G3BP1 interactors reduces MNV replication, suggesting that the G3BP1 interactome consists of host factors that are required for efficient viral replication complex formation [84,107]. Similar to alphaviruses, G3BP1 in MNV-infected cells recruit ribosomes to viral RNA and initiates translation [31]. During MNV infection, both PKR and GCN2 phosphorylate eIF2α; however, SG formation is not observed in the infected cells, even though they can still form SGs upon arsenite treatment, suggesting that MNV decouples the SG formation from eIF2α phosphorylation during virus infection. In contrast, feline calicivirus cleaves G3BP1 through the viral 3C-like proteinase NS6Pro [94].

4.3. G3BP1 Is Differentially Utilized within the Flaviviridae Family

Although G3BP1 influences viral propagation of Flaviviridae family, different members utilize G3BP1 at different stages of infection. In this section, we describe how G3BP1 function is modulated by DENV, ZIKV, and HCV.

4.3.1. Dengue Virus (DENV) Modulates G3BP1 Function by Interacting with 3′UTR vRNA

DENV infection induces G3BP1 punctate-like structures at the early stage of infection. Although the phosphorylation of eIF2α by PKR occurs at the late infection stage [73], it is unclear whether infected cells induce SGs. The expression of viral protein is reduced in cells containing G3BP1 granules, whereas knockdown of G3BP1 enhances protein level and viral titers. In DENV-2-infected cells, G3BP1 is also associated with ISG mRNAs and facilitates their translation to inhibit viral replication. As an IFN countermeasure during DENV-2 infection of Huh-7 cells, a long noncoding RNA from the subgenomic region of flaviviral RNA (sfRNA) produced by all flaviviruses binds G3BP1, G3BP2, and Caprin1, and thereby inhibits the translation of ISGs [21]. Not all flavivirus sfRNAs have this property and whether a similar decoy phenomenon may be used by other viruses to counter the production of ISG proteins and promote viral replication is unknown.

4.3.2. ZIKV and HCV Require G3BP1 for Proper Viral Replication

In both ZIKV- and HCV-infected cells, G3BP1 associates with the viral replication complex. G3BP1 depletion reduces viral replication, titer, and viral protein levels, while overexpression of G3BP1 enhances viral titers. In addition, in ZIKV-infected cells, G3BP1 interacts with capsid protein and co-localizes with envelope protein, suggesting a possible role for G3BP1 in virion assembly [70,71,72]. During HCV infection, G3BP1 interacts with the 5′-UTR of viral RNA and is involved in genome amplification [74]. Interestingly, HCV requires only G3BP1, but not G3BP2 whose knockdown does not alter viral replication [74,75].

4.4. Viruses without a Defined Role for G3BP1 in Infection

Though G3BP1 is a predominant target for many viruses, there are some viruses whose replication is unaffected by the expression level of G3BP1.

4.4.1. Depletion or Overexpression of G3BP1 Does Not Affect PRRSV Infection

PRRSV—a single-stranded positive-sense enveloped virus in the Arteriviridae family—does not require G3BP1, yet the protein is closely associated with viral replication complexes. Neither deletion nor overexpression of G3BP1, G3BP2, or both affects viral titers. In contrast to the above-described viruses, PRRSV induces SGs only at late stages of infection (48–72-h post-infection) through PERK-, rather than PKR-mediated eIF2α phosphorylation. The reason for G3BP1 association with viral replication complexes and the mechanism and role of SG assembly at late infection stages are not known [85,86]. Intriguingly, one study suggested a pro-viral function of SG assembly during PRRSV infection. Cells infected with PRRSV induces SGs, recruits G3BP1 and the viral replicase protein nsP1β, thereby inhibiting G3BP1-mediated PKR activation [108].

4.4.2. G3BP1 Is Sequestered within Ebola Virus Inclusions

EBOV, a single-stranded negative-sense RNA virus, does not induce SG assembly but sequesters SG components G3BP1, eEF2, eIF2, and eIF3 through the viral protein VP5 [55,87]. Although infection-mediated SG assembly is not observed, EBOV inhibits arsenite-induced SG assembly. VP5 expression affects IFNɑ/ꞵ expression and PKR kinase activation in infected cells. Although G3BP1 is sequestered within the viral inclusions, the exact role of G3BP1 in EBOV infection is not well understood.

5. Discussion

Viruses employ at least three strategies to target and modulate G3BP1 function during infection: (1) enzymatic activity such as proteolytic cleavage or removing post-translation modification (e.g., poliovirus and alphavirus); (2) binding or sequestration (e.g., alphavirus and coronavirus); and (3) redistribution or remodeling of G3BP1 interaction (e.g., norovirus). These functional modulations of G3BP1 by viruses impacts the viral life cycle either positively or negatively. In general, if G3BP1 exerts anti-viral activity, then viruses cleave or sequester G3BP1 (e.g., polioviruses, flaviviruses). If G3BP1 is required for viral replication, viruses do not cleave G3BP1, but instead recruit it to build viral replication complexes (e.g., alphaviruses). For pro-viral roles, the NTF2L and RGG domains of G3BP1 play a crucial role to bind viral proteins or viral RNA, respectively. Besides, some viruses possess conserved motifs (e.g., FGDF) within their proteins to bind and modulate G3BP1 function [76,109]. Intriguingly, the post-translational modification status of heavily regulated G3BP1 may determine the functional consequences of interaction. G3BP1 methylation recruits TDRD3 into SGs to regulate innate immune response [60]. Alphaviruses and coronaviruses encode an enzyme to remove ADP-ribosylation indicating that the modified form of G3BP1 may possess anti-viral functions. Given that G3BP1 is also modified by phosphorylation and ubiquitylation, it is worth checking the status of G3BP1 for other post-translation modification at different infection stages and determine how these modifications affect a viral life cycle [33,42]. Furthermore, the differences in the role of G3BP1 during infection—pro-viral or anti-viral—may also depend on the type and differentiation status of the cell infected.
G3BP1 plays critical yet distinct roles during infection by different families of the virus. Recent proteomic studies provided some insights into the G3BP1 interactome during infection. In SINV-infected cells, G3BP1 stably interacts with viral proteins throughout the life cycle, whereas some SG components interact in a time-dependent manner. During norovirus infection, the G3BP1 interactome is modulated to inhibit the formation of SGs. Given that the interacting partners of G3BP1 are modulated during infection to form new complexes in a virus-specific manner, different terms have been used to differentiate for these viral-induced protein complexes, including atypical SGs (EV), anti-viral SGs (IBV), or nsP3 foci (alphaviruses). Therefore, not all G3BP1-positive structures are SGs, and should be stained with other markers to ascertain their identities. Unlike RNA viruses, the role of G3BP1 during infection with DNA viruses is poorly understood. For example, in vaccinia virus-infected cells, G3BP1 and other SG components are recruited to cytoplasmic viral factories where transcription and translation occur. However, the precise function of G3BP1 in these viral factories is yet to be characterized. Extensive research is warranted to delineate whether G3BP1 plays a pro-viral or anti-viral role in DNA viruses [110,111].
Finally, it is noteworthy to mention that some studies have been performed in a system by overexpressing or knocking down G3BP1, which may result in ambiguous conclusions. For example, G3BP1 knockdown increases G3BP2 expression, while G3BP1 overexpression induces PKR-mediated eIF2α phosphorylation and SG formation–these non-physiological changes could affect the normal stages of the viral life cycle. To overcome these concerns, many groups recently have begun to apply CRISPR technologies to genetically knock in a fluorescent tag or knock out G3BP1 to delineate the functions of G3BP1 under physiological conditions. Special consideration should also be given to the cell type and viral load, as these factors may influence the outcome of virus infection. Given that these factors also modulate the cellular status and proteome in a spatio-temporal manner, it is critical to analyze at single-cell levels using live-cell imaging, and assays, such as ribopuromycylation, to quantitate the cellular changes only in the infected cells. Given that G3BP1 is a well-described target for many viruses, clarifying the precise functional roles of G3BP1 during infection will not only advance the field of host-virus interaction but may also offer novel therapeutic opportunities.

Author Contributions

Conceptualization, A.K.J. and A.K.L.L.; writing—original draft preparation, A.K.J.; writing—review and editing, A.K.J., D.E.G. and A.K.L.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a Johns Hopkins Catalyst Award (to A.K.L.L.), pilot grants from the Johns Hopkins University School of Medicine Sherrilyn and Ken Fisher Center for Environmental Infectious Disease (to A.K.J., D.E.G. and A.K.L.L.), NIH Grants R56AI137264 (to D.E.G. and A.K.L.L.) and R01GM104135 (to A.K.L.L.).

Conflicts of Interest

D.E.G. is on advisory boards for Takeda Pharmaceuticals, GlaxoSmithKline, Academia Sinica, and GreenLight Biosciences. The other authors declare no conflict of interest.

References

  1. Zid, B.M.; O’Shea, E.K. Promoter Sequences Direct Cytoplasmic Localization and Translation of MRNAs during Starvation in Yeast. Nature 2014, 514, 117–121. [Google Scholar] [CrossRef] [PubMed]
  2. Galluzzi, L.; Yamazaki, T.; Kroemer, G. Linking Cellular Stress Responses to Systemic Homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 731–745. [Google Scholar] [CrossRef]
  3. Jackson, S.P.; Bartek, J. The DNA-Damage Response in Human Biology and Disease. Nature 2009, 461, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  4. Hetz, C. The Unfolded Protein Response: Controlling Cell Fate Decisions under ER Stress and Beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef]
  5. Rouse, B.T.; Sehrawat, S. Immunity and Immunopathology to Viruses: What Decides the Outcome? Nat. Rev. Immunol. 2010, 10, 514–526. [Google Scholar] [CrossRef]
  6. Mogensen, T.H. Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed]
  7. Ni, G.; Ma, Z.; Damania, B. CGAS and STING: At the Intersection of DNA and RNA Virus-Sensing Networks. PLoS Pathog. 2018, 14, e1007148. [Google Scholar] [CrossRef]
  8. Lester, S.N.; Li, K. Toll-like Receptors in Antiviral Innate Immunity. J. Mol. Biol. 2014, 426, 1246–1264. [Google Scholar] [CrossRef] [PubMed]
  9. Katze, M.G.; He, Y.; Gale, M. Viruses and Interferon: A Fight for Supremacy. Nat. Rev. Immunol. 2002, 2, 675–687. [Google Scholar] [CrossRef] [PubMed]
  10. Schoggins, J.W.; Rice, C.M. Interferon-Stimulated Genes and Their Antiviral Effector Functions. Curr. Opin. Virol. 2011, 1, 519–525. [Google Scholar] [CrossRef]
  11. 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]
  12. Rehwinkel, J.; Gack, M.U. RIG-I-like Receptors: Their Regulation and Roles in RNA Sensing. Nat. Rev. Immunol. 2020, 20, 537–551. [Google Scholar] [CrossRef] [PubMed]
  13. Hornung, V.; Ellegast, J.; Kim, S.; Brzózka, K.; Jung, A.; Kato, H.; Poeck, H.; Akira, S.; Conzelmann, K.-K.; Schlee, M.; et al. 5′-Triphosphate RNA Is the Ligand for RIG-I. Science 2006, 314, 994–997. [Google Scholar] [CrossRef]
  14. Payne, S. Virus Interactions With the Cell. Viruses 2017, 23, 23–35. [Google Scholar] [CrossRef]
  15. Raoult, D.; Audic, S.; Robert, C.; Abergel, C.; Renesto, P.; Ogata, H.; La Scola, B.; Suzan, M.; Claverie, J.-M. The 1.2-Megabase Genome Sequence of Mimivirus. Science 2004, 306, 1344–1350. [Google Scholar] [CrossRef]
  16. Moore, C.H.; Farron, F.; Bohnert, D.; Weissmann, C. Possible Origin of a Minor Virus Specific Protein (A1) in Q-Beta Particles. Nat. New Biol. 1971, 234, 204–206. [Google Scholar] [CrossRef]
  17. Tsai, W.-C.; Lloyd, R.E. Cytoplasmic RNA Granules and Viral Infection. Annu. Rev. Virol. 2014, 1, 147–170. [Google Scholar] [CrossRef] [PubMed]
  18. Laver, J.D.; Ly, J.; Winn, A.K.; Karaiskakis, A.; Lin, S.; Nie, K.; Benic, G.; Jaberi-Lashkari, N.; Cao, W.X.; Khademi, A.; et al. The RNA-Binding Protein Rasputin/G3BP Enhances the Stability and Translation of Its Target MRNAs. Cell Rep. 2020, 30, 3353–3367.e7. [Google Scholar] [CrossRef]
  19. Somasekharan, S.P.; Zhang, F.; Saxena, N.; Huang, J.N.; Kuo, I.C.; Low, C.; Bell, R.; Adomat, H.; Stoynov, N.; Foster, L.; et al. G3BP1-Linked MRNA Partitioning Supports Selective Protein Synthesis in Response to Oxidative Stress. Nucleic Acids Res. 2020, 48, 6855–6873. [Google Scholar] [CrossRef]
  20. Fischer, J.W.; Busa, V.F.; Shao, Y.; Leung, A.K.L. Structure-Mediated RNA Decay by UPF1 and G3BP1. Mol. Cell 2020, 78, 70–84.e6. [Google Scholar] [CrossRef]
  21. Bidet, K.; Dadlani, D.; Garcia-Blanco, M.A. G3BP1, G3BP2 and CAPRIN1 Are Required for Translation of Interferon Stimulated MRNAs and Are Targeted by a Dengue Virus Non-Coding RNA. PLoS Pathog. 2014, 10, e1004242. [Google Scholar] [CrossRef]
  22. Sanchez, I.I.; Nguyen, T.B.; England, W.E.; Lim, R.G.; Vu, A.Q.; Miramontes, R.; Byrne, L.M.; Markmiller, S.; Lau, A.L.; Orellana, I.; et al. Huntington’s Disease Mice and Human Brain Tissue Exhibit Increased G3BP1 Granules and TDP43 Mislocalization. J. Clin. Investig. 2021, 131, e140723. [Google Scholar] [CrossRef]
  23. Scholte, F.E.M.; Tas, A.; Albulescu, I.C.; Žusinaite, E.; Merits, A.; Snijder, E.J.; van Hemert, M.J. Stress Granule Components G3BP1 and G3BP2 Play a Proviral Role Early in Chikungunya Virus Replication. J. Virol. 2015, 89, 4457–4469. [Google Scholar] [CrossRef]
  24. Cho, E.; Than, T.T.; Kim, S.-H.; Park, E.-R.; Kim, M.-Y.; Lee, K.H.O.; Shin, H.J. G3BP1 Depletion Increases Radiosensitisation by Inducing Oxidative Stress in Response to DNA Damage. Anticancer Res. 2019, 39, 6087–6095. [Google Scholar] [CrossRef] [PubMed]
  25. Dou, N.; Chen, J.; Yu, S.; Gao, Y.; Li, Y. G3BP1 Contributes to Tumor Metastasis via Upregulation of Slug Expression in Hepatocellular Carcinoma. Am. J. Cancer Res. 2016, 6, 2641–2650. [Google Scholar] [PubMed]
  26. Ge, Y.; Jin, J.; Li, J.; Ye, M.; Jin, X. The Roles of G3BP1 in Human Diseases (Review). Gene 2022, 821, 146294. [Google Scholar] [CrossRef]
  27. Irvine, K.; Stirling, R.; Hume, D.; Kennedy, D. Rasputin, More Promiscuous than Ever: A Review of G3BP. Int. J. Dev. Biol. 2004, 48, 1065–1077. [Google Scholar] [CrossRef]
  28. Matsuki, H.; Takahashi, M.; Higuchi, M.; Makokha, G.N.; Oie, M.; Fujii, M. Both G3BP1 and G3BP2 Contribute to Stress Granule Formation. Genes Cells 2013, 18, 135–146. [Google Scholar] [CrossRef]
  29. Kedersha, N.; Panas, M.D.; Achorn, C.A.; Lyons, S.; Tisdale, S.; Hickman, T.; Thomas, M.; Lieberman, J.; McInerney, G.M.; Ivanov, P.; et al. G3BP-Caprin1-USP10 Complexes Mediate Stress Granule Condensation and Associate with 40S Subunits. J. Cell Biol. 2016, 212, 845–860. [Google Scholar] [CrossRef] [PubMed]
  30. Kim, S.S.Y.; Sze, L.; Lam, K.P. The Stress Granule Protein G3BP1 Binds Viral DsRNA and RIG-I to Enhance Interferon-β Response. J. Biol. Chem. 2019, 294, 6430–6438. [Google Scholar] [CrossRef] [Green Version]
  31. Hosmillo, M.; Lu, J.; McAllaster, M.R.; Eaglesham, J.B.; Wang, X.; Emmott, E.; Domingues, P.; Chaudhry, Y.; Fitzmaurice, T.J.; Tung, M.K.; et al. Noroviruses Subvert the Core Stress Granule Component G3BP1 to Promote Viral VPg-Dependent Translation. Elife 2019, 8, e46681. [Google Scholar] [CrossRef]
  32. Riggs, C.L.; Kedersha, N.; Ivanov, P.; Anderson, P. Mammalian Stress Granules and P Bodies at a Glance. J. Cell Sci. 2020, 133, jcs242487. [Google Scholar] [CrossRef] [PubMed]
  33. Tourrière, H.; Chebli, K.; Zekri, L.; Courselaud, B.; Blanchard, J.M.; Bertrand, E.; Tazi, J. The RasGAP-Associated Endoribonuclease G3BP Assembles Stress Granules. J. Cell Biol. 2003, 160, 823–831. [Google Scholar] [CrossRef]
  34. Liu, Z.S.; Cai, H.; Xue, W.; Wang, M.; Xia, T.; Li, W.J.; Xing, J.Q.; Zhao, M.; Huang, Y.J.; Chen, S.; et al. G3BP1 Promotes DNA Binding and Activation of CGAS. Nat. Immunol. 2019, 20, 18–28. [Google Scholar] [CrossRef] [PubMed]
  35. Hu, S.; Sun, H.; Yin, L.; Li, J.; Mei, S.; Xu, F.; Wu, C.; Liu, X.; Zhao, F.; Zhang, D.; et al. PKR-Dependent Cytosolic CGAS Foci Are Necessary for Intracellular DNA Sensing. Sci. Signal. 2019, 12, eaav7934. [Google Scholar] [CrossRef]
  36. Yang, W.; Ru, Y.; Ren, J.; Bai, J.; Wei, J.; Fu, S.; Liu, X.; Li, D.; Zheng, H. G3BP1 Inhibits RNA Virus Replication by Positively Regulating RIG-I-Mediated Cellular Antiviral Response. Cell Death Dis. 2019, 10, 946. [Google Scholar] [CrossRef] [PubMed]
  37. Ivanov, P.; Kedersha, N.; Anderson, P. Stress Granules and Processing Bodies in Translational Control. Cold Spring Harb. Perspect. Biol. 2019, 11, a032813. [Google Scholar] [CrossRef] [PubMed]
  38. Lu, Y.-N.; Kavianpour, S.; Zhang, T.; Zhang, X.; Nguyen, D.; Thombre, R.; He, L.; Wang, J. MARK2 Phosphorylates EIF2α in Response to Proteotoxic Stress. PLoS Biol. 2021, 19, e3001096. [Google Scholar] [CrossRef]
  39. Taniuchi, S.; Miyake, M.; Tsugawa, K.; Oyadomari, M.; Oyadomari, S. Integrated Stress Response of Vertebrates Is Regulated by Four EIF2α Kinases. Sci. Rep. 2016, 6, 1–11. [Google Scholar] [CrossRef]
  40. Gebauer, F.; Hentze, M.W. Molecular Mechanisms of Translational Control. Nat. Rev. Mol. Cell Biol. 2004, 5, 827–835. [Google Scholar] [CrossRef]
  41. Marmor-Kollet, H.; Siany, A.; Kedersha, N.; Knafo, N.; Rivkin, N.; Danino, Y.M.; Moens, T.G.; Olender, T.; Sheban, D.; Cohen, N.; et al. Spatiotemporal Proteomic Analysis of Stress Granule Disassembly Using APEX Reveals Regulation by SUMOylation and Links to ALS Pathogenesis. Mol. Cell 2020, 80, 876–891.e6. [Google Scholar] [CrossRef]
  42. Gwon, Y.; Maxwell, B.A.; Kolaitis, R.-M.; Zhang, P.; Kim, H.J.; Paul Taylor, J. Ubiquitination of G3BP1 Mediates Stress Granule Disassembly in a Context-Specific Manner. Science 2021, 372, eabf6548. [Google Scholar] [CrossRef] [PubMed]
  43. Ohn, T.; Kedersha, N.; Hickman, T.; Tisdale, S.; Anderson, P. A Functional RNAi Screen Links O-GlcNAc Modification of Ribosomal Proteins to Stress Granule and Processing Body Assembly. Nat. Cell Biol. 2008, 10, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
  44. Tsai, W.C.; Gayatri, S.; Reineke, L.C.; Sbardella, G.; Bedford, M.T.; Lloyd, R.E. Arginine Demethylation of G3BP1 Promotes Stress Granule Assembly. J. Biol. Chem. 2016, 291, 22671–22685. [Google Scholar] [CrossRef]
  45. Leung, A.K.L.; Vyas, S.; Rood, J.E.; Bhutkar, A.; Sharp, P.A.; Chang, P. Poly(ADP-Ribose) Regulates Stress Responses and MicroRNA Activity in the Cytoplasm. Mol. Cell 2011, 42, 489–499. [Google Scholar] [CrossRef]
  46. Jayabalan, A.K.; Sanchez, A.; Park, R.Y.; Yoon, S.P.; Kang, G.Y.; Baek, J.H.; Anderson, P.; Kee, Y.; Ohn, T. NEDDylation Promotes Stress Granule Assembly. Nat. Commun. 2016, 7, 12125. [Google Scholar] [CrossRef]
  47. Jayabalan, A.K.; Adivarahan, S.; Koppula, A.; Abraham, R.; Batish, M.; Zenklusen, D.; Griffin, D.E.; Leung, A.K.L. Stress Granule Formation, Disassembly, and Composition Are Regulated by Alphavirus ADP-Ribosylhydrolase Activity. Proc. Natl. Acad. Sci. USA 2021, 118, e2021719118. [Google Scholar] [CrossRef] [PubMed]
  48. Matheny, T.; Van Treeck, B.; Huynh, T.N.; Parker, R. RNA Partitioning into Stress Granules Is Based on the Summation of Multiple Interactions. Rna 2021, 27, 174–189. [Google Scholar] [CrossRef] [PubMed]
  49. Markmiller, S.; Soltanieh, S.; Server, K.L.; Mak, R.; Jin, W.; Fang, M.Y.; Luo, E.-C.; Krach, F.; Yang, D.; Sen, A.; et al. Context-Dependent and Disease-Specific Diversity in Protein Interactions within Stress Granules. Cell 2018, 172, 590–604.e13. [Google Scholar] [CrossRef]
  50. Yang, P.; Mathieu, C.; Kolaitis, R.M.; Zhang, P.; Messing, J.; Yurtsever, U.; Yang, Z.; Wu, J.; Li, Y.; Pan, Q.; et al. G3BP1 Is a Tunable Switch That Triggers Phase Separation to Assemble Stress Granules. Cell 2020, 181, 325–345.e28. [Google Scholar] [CrossRef] [PubMed]
  51. Sanders, D.W.; Kedersha, N.; Lee, D.S.W.; Strom, A.R.; Drake, V.; Riback, J.A.; Bracha, D.; Eeftens, J.M.; Iwanicki, A.; Wang, A.; et al. Competing Protein-RNA Interaction Networks Control Multiphase Intracellular Organization. Cell 2020, 181, 306–324.e28. [Google Scholar] [CrossRef]
  52. Guillén-Boixet, J.; Kopach, A.; Holehouse, A.S.; Wittmann, S.; Jahnel, M.; Schlüßler, R.; Kim, K.; Trussina, I.R.E.A.; Wang, J.; Mateju, D.; et al. RNA-Induced Conformational Switching and Clustering of G3BP Drive Stress Granule Assembly by Condensation. Cell 2020, 181, 346–361.e17. [Google Scholar] [CrossRef] [PubMed]
  53. Ruggieri, A.; Dazert, E.; Metz, P.; Hofmann, S.; Bergeest, J.-P.; Mazur, J.; Bankhead, P.; Hiet, M.-S.; Kallis, S.; Alvisi, G.; et al. Dynamic Oscillation of Translation and Stress Granule Formation Mark the Cellular Response to Virus Infection. Cell Host Microbe 2012, 12, 71–85. [Google Scholar] [CrossRef] [PubMed]
  54. McInerney, G.M.; Kedersha, N.L.; Kaufman, R.J.; Anderson, P.; Liljeström, P. Importance of EIF2α Phosphorylation and Stress Granule Assembly in Alphavirus Translation Regulation. Mol. Biol. Cell 2005, 16, 3753–3763. [Google Scholar] [CrossRef]
  55. Le Sage, V.; Cinti, A.; McCarthy, S.; Amorim, R.; Rao, S.; Daino, G.L.; Tramontano, E.; Branch, D.R.; Mouland, A.J. Ebola Virus VP35 Blocks Stress Granule Assembly. Virology 2017, 502, 73–83. [Google Scholar] [CrossRef]
  56. Weissbach, R.; Scadden, A.D.J. Tudor-SN and ADAR1 Are Components of Cytoplasmic Stress Granules. RNA 2012, 18, 462–471. [Google Scholar] [CrossRef]
  57. Law, L.M.J.; Razooky, B.S.; Li, M.M.H.; You, S.; Jurado, A.; Rice, C.M.; Macdonald, M.R. ZAP’s Stress Granule Localization Is Correlated with Its Antiviral Activity and Induced by Virus Replication. PLoS Pathog. 2019, 15, e1007798. [Google Scholar] [CrossRef] [PubMed]
  58. Onomoto, K.; Jogi, M.; Yoo, J.-S.; Narita, R.; Morimoto, S.; Takemura, A.; Sambhara, S.; Kawaguchi, A.; Osari, S.; Nagata, K.; et al. Critical Role of an Antiviral Stress Granule Containing RIG-I and PKR in Viral Detection and Innate Immunity. PLoS ONE 2012, 7, e43031. [Google Scholar] [CrossRef]
  59. Reineke, L.C.; Lloyd, R.E. Diversion of Stress Granules and P-Bodies during Viral Infection. Virology 2013, 436, 255–267. [Google Scholar] [CrossRef]
  60. Deater, M.; Tamhankar, M.; Lloyd, R.E. TDRD3 Is an Antiviral Restriction Factor That Promotes IFN Signaling with G3BP1. PLoS Pathog. 2022, 18, e1010249. [Google Scholar] [CrossRef]
  61. Ng, C.S.; Jogi, M.; Yoo, J.-S.; Onomoto, K.; Koike, S.; Iwasaki, T.; Yoneyama, M.; Kato, H.; Fujita, T. Encephalomyocarditis Virus Disrupts Stress Granules, the Critical Platform for Triggering Antiviral Innate Immune Responses. J. Virol. 2013, 87, 9511–9522. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, W.; Li, D.; Ru, Y.; Bai, J.; Ren, J.; Zhang, J.; Li, L.; Liu, X.; Zheng, H. Foot-and-Mouth Disease Virus 3A Protein Causes Upregulation of Autophagy-Related Protein LRRC25 To Inhibit the G3BP1-Mediated RIG-Like Helicase-Signaling Pathway. J. Virol. 2020, 94, e02086-19. [Google Scholar] [CrossRef] [PubMed]
  63. Visser, L.J.; Medina, G.N.; Rabouw, H.H.; de Groot, R.J.; Langereis, M.A.; de Los Santos, T.; van Kuppeveld, F.J.M. Foot-and-Mouth Disease Virus Leader Protease Cleaves G3BP1 and G3BP2 and Inhibits Stress Granule Formation. J. Virol. 2019, 93, e00922-18. [Google Scholar] [CrossRef] [PubMed]
  64. Visser, L.J.; Langereis, M.A.; Rabouw, H.H.; Wahedi, M.; Muntjewerff, E.M.; de Groot, R.J.; van Kuppeveld, F.J.M. Essential Role of Enterovirus 2A Protease in Counteracting Stress Granule Formation and the Induction of Type I Interferon. J. Virol. 2019, 93, e00222-19. [Google Scholar] [CrossRef]
  65. Zhang, Y.; Yao, L.; Xu, X.; Han, H.; Li, P.; Zou, D.; Li, X.; Zheng, L.; Cheng, L.; Shen, Y.; et al. Enterovirus 71 Inhibits Cytoplasmic Stress Granule Formation during the Late Stage of Infection. Virus Res. 2018, 255, 55–67. [Google Scholar] [CrossRef]
  66. Cheng, J.; Gao, S.; Zhu, C.; Liu, S.; Li, J.; Kang, J.; Wang, Z.; Wang, T. Typical Stress Granule Proteins Interact with the 3′ Untranslated Region of Enterovirus D68 To Inhibit Viral Replication. J. Virol. 2020, 94, e02041-19. [Google Scholar] [CrossRef]
  67. White, J.P.; Cardenas, A.M.; Marissen, W.E.; Lloyd, R.E. Inhibition of Cytoplasmic MRNA Stress Granule Formation by a Viral Proteinase. Cell Host Microbe 2007, 2, 295–305. [Google Scholar] [CrossRef]
  68. Wu, S.; Wang, Y.; Lin, L.; Si, X.; Wang, T.; Zhong, X.; Tong, L.; Luan, Y.; Chen, Y.; Li, X.; et al. Protease 2A Induces Stress Granule Formation during Coxsackievirus B3 and Enterovirus 71 Infections. Virol. J. 2014, 11, 192. [Google Scholar] [CrossRef]
  69. Fung, G.; Ng, C.S.; Zhang, J.; Shi, J.; Wong, J.; Piesik, P.; Han, L.; Chu, F.; Jagdeo, J.; Jan, E.; et al. Production of a Dominant-Negative Fragment Due to G3BP1 Cleavage Contributes to the Disruption of Mitochondria-Associated Protective Stress Granules during CVB3 Infection. PLoS ONE 2013, 8, e79546. [Google Scholar] [CrossRef]
  70. Bonenfant, G.; Williams, N.; Netzband, R.; Schwarz, M.C.; Evans, M.J.; Pager, C.T. Zika Virus Subverts Stress Granules To Promote and Restrict Viral Gene Expression. J. Virol. 2019, 93, e00520-19. [Google Scholar] [CrossRef] [Green Version]
  71. Amorim, R.; Temzi, A.; Griffin, B.D.; Mouland, A.J. Zika Virus Inhibits EIF2α-Dependent Stress Granule Assembly. PLoS Negl. Trop. Dis. 2017, 11, e0005775. [Google Scholar] [CrossRef] [PubMed]
  72. Hou, S.; Kumar, A.; Xu, Z.; Airo, A.M.; Stryapunina, I.; Wong, C.P.; Branton, W.; Tchesnokov, E.; Götte, M.; Power, C.; et al. Zika Virus Hijacks Stress Granule Proteins and Modulates the Host Stress Response. J. Virol. 2017, 91, e00474-17. [Google Scholar] [CrossRef] [PubMed]
  73. Xia, J.; Chen, X.; Xu, F.; Wang, Y.; Shi, Y.; Li, Y.; He, J.; Zhang, P. Dengue Virus Infection Induces Formation of G3BP1 Granules in Human Lung Epithelial Cells. Arch. Virol. 2015, 160, 2991–2999. [Google Scholar] [CrossRef]
  74. Yi, Z.; Pan, T.; Wu, X.; Song, W.; Wang, S.; Xu, Y.; Rice, C.M.; Macdonald, M.R.; Yuan, Z. Hepatitis C Virus Co-Opts Ras-GTPase-Activating Protein-Binding Protein 1 for Its Genome Replication. J. Virol. 2011, 85, 6996–7004. [Google Scholar] [CrossRef]
  75. Garaigorta, U.; Heim, M.H.; Boyd, B.; Wieland, S.; Chisari, F.V. Hepatitis C Virus (HCV) Induces Formation of Stress Granules Whose Proteins Regulate HCV RNA Replication and Virus Assembly and Egress. J. Virol. 2012, 86, 11043–11056. [Google Scholar] [CrossRef] [PubMed]
  76. Panas, M.D.; Ahola, T.; McInerney, G.M. The C-Terminal Repeat Domains of NsP3 from the Old World Alphaviruses Bind Directly to G3BP. J. Virol. 2014, 88, 5888–5893. [Google Scholar] [CrossRef] [PubMed]
  77. Panas, M.D.; Varjak, M.; Lulla, A.; Eng, K.E.; Merits, A.; Hedestam, G.B.K.; McInerney, G.M. Sequestration of G3BP Coupled with Efficient Translation Inhibits Stress Granules in Semliki Forest Virus Infection. Mol. Biol. Cell 2012, 23, 4701–4712. [Google Scholar] [CrossRef]
  78. Foy, N.J.; Akhrymuk, M.; Akhrymuk, I.; Atasheva, S.; Bopda-Waffo, A.; Frolov, I.; Frolova, E.I. Hypervariable Domains of NsP3 Proteins of New World and Old World Alphaviruses Mediate Formation of Distinct, Virus-Specific Protein Complexes. J. Virol. 2013, 87, 1997–2010. [Google Scholar] [CrossRef] [PubMed]
  79. Sun, L.; Chen, H.; Ming, X.; Bo, Z.; Shin, H.-J.; Jung, Y.-S.; Qian, Y. Porcine Epidemic Diarrhea Virus Infection Induces Caspase-8-Mediated G3BP1 Cleavage and Subverts Stress Granules To Promote Viral Replication. J. Virol. 2021, 95, e02344-20. [Google Scholar] [CrossRef] [PubMed]
  80. Pandey, K.; Zhong, S.; Diel, D.G.; Hou, Y.; Wang, Q.; Nelson, E.; Wang, X. GTPase-Activating Protein-Binding Protein 1 (G3BP1) Plays an Antiviral Role against Porcine Epidemic Diarrhea Virus. Vet. Microbiol. 2019, 236, 108392. [Google Scholar] [CrossRef]
  81. Zheng, Z.-Q.; Wang, S.-Y.; Xu, Z.-S.; Fu, Y.-Z.; Wang, Y.-Y. SARS-CoV-2 Nucleocapsid Protein Impairs Stress Granule Formation to Promote Viral Replication. Cell Discovery 2021, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
  82. Luo, L.; Li, Z.; Zhao, T.; Ju, X.; Ma, P.; Jin, B.; Zhou, Y.; He, S.; Huang, J.; Xu, X.; et al. SARS-CoV-2 Nucleocapsid Protein Phase Separates with G3BPs to Disassemble Stress Granules and Facilitate Viral Production. Sci. Bull. 2021, 66, 1194–1204. [Google Scholar] [CrossRef] [PubMed]
  83. Lu, S.; Ye, Q.; Singh, D.; Cao, Y.; Diedrich, J.K.; Yates, J.R.; Villa, E.; Cleveland, D.W.; Corbett, K.D. The SARS-CoV-2 Nucleocapsid Phosphoprotein Forms Mutually Exclusive Condensates with RNA and the Membrane-Associated M Protein. Nat. Commun. 2021, 12, 502. [Google Scholar] [CrossRef]
  84. Brocard, M.; Iadevaia, V.; Klein, P.; Hall, B.; Lewis, G.; Lu, J.; Burke, J.; Willcocks, M.M.; Parker, R.; Goodfellow, I.G.; et al. Norovirus Infection Results in EIF2α Independent Host Translation Shut-off and Remodels the G3BP1 Interactome Evading Stress Granule Formation. PLoS Pathog. 2020, 16, e1008250. [Google Scholar] [CrossRef] [PubMed]
  85. Catanzaro, N.; Meng, X.-J. Porcine Reproductive and Respiratory Syndrome Virus (PRRSV)-Induced Stress Granules Are Associated with Viral Replication Complexes and Suppression of Host Translation. Virus Res. 2019, 265, 47–56. [Google Scholar] [CrossRef] [PubMed]
  86. Zhou, Y.; Fang, L.; Wang, D.; Cai, K.; Chen, H.; Xiao, S. Porcine Reproductive and Respiratory Syndrome Virus Infection Induces Stress Granule Formation Depending on Protein Kinase R-like Endoplasmic Reticulum Kinase (PERK) in MARC-145 Cells. Front. Cell. Infect. Microbiol. 2017, 7, 111. [Google Scholar] [CrossRef]
  87. Nelson, E.V.; Schmidt, K.M.; Deflubé, L.R.; Doğanay, S.; Banadyga, L.; Olejnik, J.; Hume, A.J.; Ryabchikova, E.; Ebihara, H.; Kedersha, N.; et al. Ebola Virus Does Not Induce Stress Granule Formation during Infection and Sequesters Stress Granule Proteins within Viral Inclusions. J. Virol. 2016, 90, 7268–7284. [Google Scholar] [CrossRef]
  88. Kim, S.S.-Y.; Sim, D.C.N.; Carissimo, G.; Lim, H.-H.; Lam, K.-P. Bruton’s Tyrosine Kinase Phosphorylates Scaffolding and RNA-Binding Protein G3BP1 to Induce Stress Granule Aggregation during Host Sensing of Foreign Ribonucleic Acids. J. Biol. Chem. 2022, 298, 102231. [Google Scholar] [CrossRef] [PubMed]
  89. Galan, A.; Lozano, G.; Piñeiro, D.; Martinez-Salas, E. G3BP1 Interacts Directly with the FMDV IRES and Negatively Regulates Translation. FEBS J. 2017, 284, 3202–3217. [Google Scholar] [CrossRef]
  90. Dougherty, J.D.; White, J.P.; Lloyd, R.E. Poliovirus-Mediated Disruption of Cytoplasmic Processing Bodies. J. Virol. 2011, 85, 64–75. [Google Scholar] [CrossRef] [Green Version]
  91. Zhai, X.; Wu, S.; Lin, L.; Wang, T.; Zhong, X.; Chen, Y.; Xu, W.; Tong, L.; Wang, Y.; Zhao, W.; et al. Stress Granule Formation Is One of the Early Antiviral Mechanisms for Host Cells Against Coxsackievirus B Infection. Virol. Sin. 2018, 33, 314–322. [Google Scholar] [CrossRef]
  92. 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]
  93. Cristea, I.M.; Carroll, J.-W.N.; Rout, M.P.; Rice, C.M.; Chait, B.T.; MacDonald, M.R. Tracking and Elucidating Alphavirus-Host Protein Interactions. J. Biol. Chem. 2006, 281, 30269–30278. [Google Scholar] [CrossRef]
  94. Humoud, M.N.; Doyle, N.; Royall, E.; Willcocks, M.M.; Sorgeloos, F.; van Kuppeveld, F.; Roberts, L.O.; Goodfellow, I.G.; Langereis, M.A.; Locker, N. Feline Calicivirus Infection Disrupts Assembly of Cytoplasmic Stress Granules and Induces G3BP1 Cleavage. J. Virol. 2016, 90, 6489–6501. [Google Scholar] [CrossRef] [PubMed]
  95. Jaafar, Z.A.; Kieft, J.S. Viral RNA Structure-Based Strategies to Manipulate Translation. Nat. Rev. Microbiol. 2019, 17, 110–123. [Google Scholar] [CrossRef] [PubMed]
  96. Ciccosanti, F.; Di Rienzo, M.; Romagnoli, A.; Colavita, F.; Refolo, G.; Castilletti, C.; Agrati, C.; Brai, A.; Manetti, F.; Botta, L.; et al. Proteomic Analysis Identifies the RNA Helicase DDX3X as a Host Target against SARS-CoV-2 Infection. Antiviral Res. 2021, 190, 105064. [Google Scholar] [CrossRef]
  97. Zheng, Y.; Deng, J.; Han, L.; Zhuang, M.-W.; Xu, Y.; Zhang, J.; Nan, M.-L.; Xiao, Y.; Zhan, P.; Liu, X.; et al. SARS-CoV-2 NSP5 and N Protein Counteract the RIG-I Signaling Pathway by Suppressing the Formation of Stress Granules. Signal Transduct Target Ther 2022, 7, 22. [Google Scholar] [CrossRef]
  98. Liu, H.; Bai, Y.; Zhang, X.; Gao, T.; Liu, Y.; Li, E.; Wang, X.; Cao, Z.; Zhu, L.; Dong, Q.; et al. SARS-CoV-2 N Protein Antagonizes Stress Granule Assembly and IFN Production by Interacting with G3BPs to Facilitate Viral Replication. J. Virol. 2022, 96, e0041222. [Google Scholar] [CrossRef]
  99. Lindquist, M.E.; Lifland, A.W.; Utley, T.J.; Santangelo, P.J.; Crowe, J.E. Respiratory Syncytial Virus Induces Host RNA Stress Granules To Facilitate Viral Replication. J. Virol. 2010, 84, 12274–12284. [Google Scholar] [CrossRef]
  100. Götte, B.; Panas, M.D.; Hellström, K.; Liu, L.; Samreen, B.; Larsson, O.; Ahola, T.; McInerney, G.M. Separate Domains of G3BP Promote Efficient Clustering of Alphavirus Replication Complexes and Recruitment of the Translation Initiation Machinery. PLoS Pathog. 2019, 15, e1007842. [Google Scholar] [CrossRef] [Green Version]
  101. McPherson, R.L.; Abraham, R.; Sreekumar, E.; Ong, S.E.; Cheng, S.J.; Baxter, V.K.; Kistemaker, H.A.V.; Filippov, D.V.; Griffin, D.E.; Leung, A.K.L. 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]
  102. Alhammad, Y.M.O.; Kashipathy, M.M.; Roy, A.; Gagné, J.-P.; McDonald, P.; Gao, P.; Nonfoux, L.; Battaile, K.P.; Johnson, D.K.; Holmstrom, E.D.; et al. The SARS-CoV-2 Conserved Macrodomain Is a Mono-ADP-Ribosylhydrolase. bioRxiv 2020, 95, e01969-20. [Google Scholar] [CrossRef] [PubMed]
  103. Leung, A.K.L.; McPherson, R.L.; Griffin, D.E. Macrodomain ADP-Ribosylhydrolase and the Pathogenesis of Infectious Diseases. PLoS Pathog. 2018, 14, e1006864. [Google Scholar] [CrossRef] [PubMed]
  104. Leung, A.K.L.; Griffin, D.E.; Bosch, J.; Fehr, A.R. The Conserved Macrodomain Is a Potential Therapeutic Target for Coronaviruses and Alphaviruses. Pathogens 2022, 11, 94. [Google Scholar] [CrossRef]
  105. 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]
  106. Kim, D.Y.; Reynaud, J.M.; Rasalouskaya, A.; Akhrymuk, I.; Mobley, J.A.; Frolov, I.; Frolova, E.I. New World and Old World Alphaviruses Have Evolved to Exploit Different Components of Stress Granules, FXR and G3BP Proteins, for Assembly of Viral Replication Complexes. PLoS Pathog. 2016, 12, e1005810. [Google Scholar] [CrossRef]
  107. Fritzlar, S.; Aktepe, T.E.; Chao, Y.-W.; Kenney, N.D.; McAllaster, M.R.; Wilen, C.B.; White, P.A.; Mackenzie, J.M. Mouse Norovirus Infection Arrests Host Cell Translation Uncoupled from the Stress Granule-PKR-EIF2α Axis. MBio 2019, 10, e00960-19. [Google Scholar] [CrossRef]
  108. Gao, P.; Liu, Y.; Wang, H.; Chai, Y.; Weng, W.; Zhang, Y.; Zhou, L.; Ge, X.; Guo, X.; Han, J.; et al. Viral Evasion of PKR Restriction by Reprogramming Cellular Stress Granules. Proc. Natl. Acad. Sci. USA 2022, 119, e2201169119. [Google Scholar] [CrossRef] [PubMed]
  109. Panas, M.D.; Schulte, T.; Thaa, B.; Sandalova, T.; Kedersha, N.; Achour, A.; McInerney, G.M. Viral and Cellular Proteins Containing FGDF Motifs Bind G3BP to Block Stress Granule Formation. PLoS Pathog. 2015, 11, e1004659. [Google Scholar] [CrossRef]
  110. Katsafanas, G.C.; Moss, B. Colocalization of Transcription and Translation within Cytoplasmic Poxvirus Factories Coordinates Viral Expression and Subjugates Host Functions. Cell Host Microbe 2007, 2, 221–228. [Google Scholar] [CrossRef] [Green Version]
  111. Katsafanas, G.C.; Moss, B. Vaccinia Virus Intermediate Stage Transcription Is Complemented by Ras-GTPase-Activating Protein SH3 Domain-Binding Protein (G3BP) and Cytoplasmic Activation/Proliferation-Associated Protein (P137) Individually or as a Heterodimer. J. Biol. Chem. 2004, 279, 52210–52217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Domain architecture of G3BP1 and G3BP2.
Figure 1. Domain architecture of G3BP1 and G3BP2.
Viruses 15 00449 g001
Figure 2. Role of G3BP1 in (a) innate immune signaling and (b) stress granule assembly.
Figure 2. Role of G3BP1 in (a) innate immune signaling and (b) stress granule assembly.
Viruses 15 00449 g002
Figure 3. Patterns of SGs observed during infection by distinct viral types.
Figure 3. Patterns of SGs observed during infection by distinct viral types.
Viruses 15 00449 g003
Figure 4. Strategies employed by different viruses to modulate G3BP1.
Figure 4. Strategies employed by different viruses to modulate G3BP1.
Viruses 15 00449 g004
Table 1. SG status during infection against different viruses.
Table 1. SG status during infection against different viruses.
FamilySpeciesSG DynamicsDuration Monitored for SG PresenceeIF2alphα Phosphorylation StatusResponsible KinaseCell Line TestedReferences
PicornaviridaeEMCVTransient4, 12 hpi*ND*PKRHeLa[61]
FMDVBlock4–6 hpi*ND**ND*PK-15[62,63]
EVTransient0–24 hpi8 hPKRHeLa, RD[64,65,66]
PV0–6 hpi*ND**ND*HeLa, 293T, MCF7, Vero[67]
CVB31–7 hpi6 h*ND*HeLa[68,69]
FlaviviridaeZikaBlock 24 hpi24 hPKRA549, Huh7, Vero[70,71,72]
DENVUnknown6–24 hpi12 hPKRA549, Huh7[21,73]
HCVOscillating0–96 hpi24 hPKRHuh7, HEK293T[53,74,75]
TogaviridaeCHIKVTransient0–12 hpi6 h*ND*U2OS, HEK293, Vero[23,47]
SFVTransient2–8 hpi5 h*ND*MEF[54,76,77]
SINVTransient 6 hPKRMEF[78]
CoronaviridaePEDVTransient0–36 hpi*ND**ND*Vero E6, Vero-76[79,80]
SARS-CoV-2Block0, 10, 24 hpi*ND*PKRHeLa [81,82,83]
CaliciviridaeMNVUnknown9, 12 hpi9 hPKR, GCN2BMDM, BV2, RAW264.7[31,84]
ArteriviridaePRRSVStable12–48 hpi12 hPERKMARK-145[85,86]
FiloviridaeEVDBlock0.5–24 hpi*ND**ND*U2OS, Vero, Huh7[55,87]
*ND*—Not determined. Species: Chikungunya virus (CHIKV); Coxsackie virus B (CVB3); Dengue virus (DENV); Ebola virus disease (EVD); Encephalomyocarditis virus (EMCV); Enterovirus (EV); Foot-and-mouth disease virus (FMDV); Hepatitis C virus (HCV); Murine norovirus (MNV); Porcine epidemic diarrhea virus (PEDV); Porcine reproductive and respiratory syndrome virus (PRRSV); Poliovirus (PV); severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); Semliki Forest virus (SFV); Sindbis virus (SINV); Zika virus (ZIKV), Human norovirus (HuNoV), Feline calicivirus (FCV). Duration of SG presence monitored: the time point identified in each report; also, the time point ranges differently. eIF2⍺ status: the earliest time point at which phosphorylation of eIF2α was observed or tested. Kinases: protein kinase R (PKR); protein kinase R-like ER kinase (PERK); general control nonderepressible 2 (GCN2). Cell lines: A549 (lung carcinoma epithelial cells), BMDM (bone marrow-derived macrophages), BV2 (microglial cell derived from C57/BL6 murine), HEK293T (human embryonic kidney 293 cells expressing SV40 T-antigen), HeLa (cervical cancer cells), Huh7 (human hepatoma-derived cell line), MARK-145 (monkey kidney cells), MCF7 (breast cancer cells), MEF (mouse embryonic fibroblasts), PK-15 (porcine kidney cells), RAW264.7 (monocyte/macrophage-like cells), RD (Rhabdomyosarcoma), U2OS (osteosarcoma), Vero (cells derived from kidney of an African green monkey).
Table 2. Role of G3BP1 and proposed mechanism of viral modulation.
Table 2. Role of G3BP1 and proposed mechanism of viral modulation.
FamilySpeciesG3BP1 StatusRole of G3BP1Effect of Viral Protein/RNA or Titer ValueInteraction with Viral ProteinInteraction with Viral RNAProposed Mechanism of ActionReferences
G3BP1 KDG3BP1 OE
PicornaviridaeEMCVCleavageAnti-viral*ND* 3C protease cleaves G3BP1 at Q325[61]
FMDV3AInteracts with IRESLeader protein cleaves G3BP1, G3BP1 dephosphorylated, G3BP1 binds to FMDV IRES region, 3A protein degrades G3BP1 through autophagy[62,63,89]
EV Interacts with 3′UTR3C proteinase cleaves G3BP1 at Q326[64,65,66]
PV*ND* 3C protease cleaves G3BP1, but not G3BP2, at Q326[67,90]
CVB3 3C protease cleaves G3BP1 at Q325[69,91]
FlaviviridaeZikaSequestrationPro-viralInteracts with Capsid, colocalizes with envelope proteinInteracts with genomic RNA, localize with replication complexesSequester G3BP1 and facilitates viral replication[70,71,72]
DENVAnti-viral*ND* Subgenomic viral RNA binds to G3BP1 and antagonizes its function[21,73]
HCVPro-viral*ND*NS5BLocalizes to vRCG3BP1 requires at early and late stages of infection[53,74,75]
TogaviridaeCHIKVSequestrationPro-viral*ND*nsP3yesBinding through FGDF motif, Reduction G3BP1 ADP-ribosylation[23,47,92]
SFV*ND*nsP3yesBinds through FGDF motif; recruits ribosomal proteins to nsP3[54,76,77]
SINV*ND*nsP3, nsP4Colocalizes with vRNABinding through FGDF motif block SG assembly[78,93]
CoronaviridaePEDVCleavageAnti-viral Caspase-8-mediated G3BP1 cleavage at Asp168 and Asp169 at late infection stages[79,80]
SARS-CoV-2SequestrationBoth proviral & antiviral roles have been reported*ND*N protein, nsP1 N protein interacts/phase separates with G3BP1[81,82,83]
CaliciviridaeMNVSequestrationPro-viral*ND*NS3, VPgColocalizes with vRCsRemodels G3BP1 interactome, doesn’t affect SGs[31,84,94]
FCVCleavage*ND* NS6-mediated G3BP1 cleavage at E405
ArteriviridaePRRSV*ND*Not involvedNo changes G3BP1 closely associated with vRCs [85,86]
FiloviridaeEVDSequestration*ND**ND*VP5 Sequestered within viral inclusions[55,87]
*ND*—Not determined. Interaction with viral protein: identified through physical interactions. Interaction with viral RNA: identified through co-localization studies.
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

Jayabalan, A.K.; Griffin, D.E.; Leung, A.K.L. Pro-Viral and Anti-Viral Roles of the RNA-Binding Protein G3BP1. Viruses 2023, 15, 449. https://doi.org/10.3390/v15020449

AMA Style

Jayabalan AK, Griffin DE, Leung AKL. Pro-Viral and Anti-Viral Roles of the RNA-Binding Protein G3BP1. Viruses. 2023; 15(2):449. https://doi.org/10.3390/v15020449

Chicago/Turabian Style

Jayabalan, Aravinth Kumar, Diane E. Griffin, and Anthony K. L. Leung. 2023. "Pro-Viral and Anti-Viral Roles of the RNA-Binding Protein G3BP1" Viruses 15, no. 2: 449. https://doi.org/10.3390/v15020449

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