Next Article in Journal
Viruses and Cajal Bodies: A Critical Cellular Target in Virus Infection?
Next Article in Special Issue
The Novel Porcine Parvoviruses: Current State of Knowledge and Their Possible Implications in Clinical Syndromes in Pigs
Previous Article in Journal
COVID-19 Genomic Surveillance in Bangui (Central African Republic) Reveals a Landscape of Circulating Variants Linked to Validated Antiviral Targets of SARS-CoV-2 Proteome
Previous Article in Special Issue
Pathogenicity Studies of NADC34-like Porcine Reproductive and Respiratory Syndrome Virus LNSY-GY and NADC30-like Porcine Reproductive and Respiratory Syndrome Virus GXGG-8011 in Piglets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress of Porcine Reproductive and Respiratory Syndrome Virus NSP2 Protein

by
Benjin Liu
1,†,
Lingzhi Luo
1,†,
Ziqi Shi
1,
Houbin Ju
2,
Lingxue Yu
3,*,
Guoxin Li
3 and
Jin Cui
1,*
1
College of Veterinary Medicine, Northeast Agricultural University, Harbin 150038, China
2
Shanghai Animal Disease Prevention and Control Center, Shanghai 201103, China
3
Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2023, 15(12), 2310; https://doi.org/10.3390/v15122310
Submission received: 30 October 2023 / Revised: 21 November 2023 / Accepted: 22 November 2023 / Published: 24 November 2023
(This article belongs to the Special Issue Porcine Viruses 2023)

Abstract

:
Porcine reproductive and respiratory syndrome virus (PRRSV) is globally prevalent and seriously harms the economic efficiency of pig farming. Because of its immunosuppression and high incidence of mutant recombination, PRRSV poses a great challenge for disease prevention and control. Nonstructural protein 2 (NSP2) is the most variable functional protein in the PRRSV genome and can generate NSP2N and NSP2TF variants due to programmed ribosomal frameshifts. These variants are broad and complex in function and play key roles in numerous aspects of viral protein maturation, viral particle assembly, regulation of immunity, autophagy, apoptosis, cell cycle and cell morphology. In this paper, we review the structural composition, programmed ribosomal frameshift and biological properties of NSP2 to facilitate basic research on PRRSV and to provide theoretical support for disease prevention and control and therapeutic drug development.

1. Introduction

1.1. Epidemiology of PRRSV

Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative pathogen of porcine reproductive and respiratory syndrome (PRRS) and is characterized by reproductive disorders in sows and respiratory disorders in pigs of all ages. The disease has been prevalent around the world for more than 30 years and has seriously harmed the global pig breeding industry. In 1992, the World Organization for Animal Health (OIE) classified PRRSV as a Category B infectious disease [1]. Currently, PRRSV is divided into PRRSV-1 and PRRSV-2 [2,3], of which PRRSV-2 is the dominant strain in China. The first PRRSV strain, CH-1a, was isolated in China in 1996 [4]. In 2006, HP-PRRSV (a highly pathogenic PRRS virus) broke out in China, and its virus genome is characterized by 30 amino acid deletions in the NSP2 hypervariable region [5]. Subsequently, China discovered the main prevalent NADC30 strain in China in 2015, and the potentially prevalent NADC34 strain in China in 2018 [6,7,8,9] (Table 1). Beginning in 2020, a severe PRRSV outbreak in northeastern Spain was characterized by a high rate of abortion and increased mortality among weaners. Virus isolation and sequencing analysis showed that they were caused by a mosaic PRRSV strain, derived from the PR40 strain reported by Canelli, after drifting and undergoing several recombination events with other PRRSV-1.1 isolates. The strains quickly changed from an acute course to a slow form in evolution, and the virulence was weakened [10,11]. In addition, research has shown that all respiratory and intestinal mixed infections lead to increased farm losses, which is exacerbated by secondary infections with Streptococcus suis and intestinal bacteria [12]. PRRSV continues to evolve new variants with increasingly complex differentiation and virulence phenotypes [13], posing enormous challenges to disease prevention and control.

1.2. PRRSV Viral Genome Structure and Function

PRRSV is a member of the Arteriviridae family and is an enveloped single-stranded positive-sense RNA virus [21]. Its genome is approximately 15 kb and contains at least 10 known open reading frames, ORF1a, ORF1b, ORF2a, ORF2b, ORF3-7 and ORF5a, as well as flanking 5′ and 3′-untranslated regions (Figure 1) [22]. Except for ORF1, the other open reading frames are translated into viral structural proteins and participate in virus assembly; ORF1 is approximately 12 kb long, accounting for approximately 80% of the viral genome and encodes two larger subreading frames, namely ORFla and ORFlb. The polypeptides produced by translation are hydrolyzed and processed by autoencoded proteases to produce at least 16 nonstructural proteins (NSP1-12), which together with structural proteins, are involved in regulating host cells (Table 2) [21,23].
PRRSV has the characteristics of easy mutation, strain diversity, immunosuppression, difficulty in producing neutralizing antibodies and ADCC effects. In recent years, China has approved many attenuated vaccine strains for use, making the virus prone to mutation, resulting in frequent recombination between field virus strains and vaccine strains and the emergence of a series of new PRRSV mutant strains, such as NADC30-like or NADC34-like strains [6,7,9]. The emergence of new strains and the characteristics of the virus itself have made the prevention and control of PRRSV very challenging.

2. NSP2 Structural Composition and Ribosome Frameshifting

2.1. Structural Composition and Genetic Variation of NSP2

NSP2 is the largest replicase cleavage product of PRRSV and is a large multidomain protein of 1066–1197 aa. The structural composition consists of six domains, including the N-terminal hypervariable region HV-Ⅰ, cysteine protease PL2 region, Peptidase_C33 structure, 500–700 amino acid hypervariable region HV-Ⅱ and transmembrane domain TM. NSP2 has a hydrophilic cytoplasmic domain tail composed of approximately 200 amino acids at the C-terminus. Because NSP2 has a hypervariable region, the amino acid similarity of NSP2 in strains of the same subtype is 66–70%, but the similarity in strains of different subtypes is only approximately 30% [20]. Therefore, NSP2 has the largest variation in the genome [69], and NSP2 also has the ability to recombine between strains: in vivo recombination experiments found that the NSP2, ORF3 and ORF5 genes will recombine and exchange when different strains infect SPF pigs at the same time [70].
The PLP2 domain of NSP2 has cis and trans cleavage activities. At least six NSP2 isoforms of different lengths can be generated through enzymatic cleavage: NSP2a, NSP2b, NSP2c, NSP2d, NSP2e and NSP2f. Their expression levels are relatively stable. Among these isoforms, NSP2d, NSP2e and NSP2f cleavage sites are all located in the hypervariable region. There are no predictable conserved enzyme cleavage sites, and they have no obvious impact on PRRSV replication [27]. Studies have confirmed that the PLP2 domain can inactivate cis and trans cleavage activities through C55A and H124C [71]. In addition, the plp2 domain also has deubiquitination and deacylation functions, which can be lost through site mutations such as D85R or D91R [72].
The hypervariable region of NSP2 is flexible and contains multiple B-cell epitopes. Jun Han et al. [73] used VR2332 as the backbone through a reverse genetic system to construct a series of mutant strains and indicated that NSP2 can be randomly deleted between 324 and 813 (aa), and it was found that a maximum of 400 aa (NSP2 324–726) can be deleted and 13–35 aa at the N end can be deleted but other positions cannot be deleted. When the mutant strain has fewer deletions, it has growth kinetics and RNA expression profiles similar to those of the parent virus, but the lytic activity of the NSP2 324–726 mutant on MARC-145 cells is reduced, and no visible plaques appear. This finding indicates that the caspase domain PL2 of NSP2, its downstream Peptidase_C33 structure, the putative transmembrane domain and the C-terminal tail region are necessary for replication. The deletable positions were mainly concentrated in the B-cell epitope region in the NSP2 hypervariable region. This region may be used as a bait epitope to deviate the host immune response from key viral proteins and to interfere with the host’s immune response. However, this region cannot be completely deleted. It may be crucial to maintain the spatial structure of the entire protein. In addition, Jun Han et al. also found that by inserting the gene encoding GFP into a NSP2 deletion mutant, the constructed recombinant virus was damaged and unstable and could gradually gain parental growth ability by losing most of the GFP gene [73]. The ability of the virus to quickly be lost is amazing.
Reviewing previous studies, the characteristics of mutable deletions in the hypervariable region of NSP2 are consistent with the HP-PRRSV outbreak in China in 2006, which had a 30-amino-acid deletion in the B-cell epitope region of NSP2, but the deletion in this region has been shown not to be associated with changes in virulence [74]. This is also consistent with the structural understanding of NSP2. Little is known about other structures of NSP2, especially the function of its C-terminus, and more important biological functions of NSP2 need to be elucidated.

2.2. NSP2 and Ribosome Frameshifting

Programmed ribosomal frameshift (PRF) is a mechanism by which host cells or viruses produce multiple proteins through overlapping reading frames. PRF was first proposed in sarcoid viruses, which generate gag–pol fusion proteins from transiently overlapping gag and pol ORFS by sliding the host cell ribosome backward one position, −1 PRF, during translation of the viral RNA genome [75,76,77]. Arter viruses such as PRRSV have evolved a −2/−1 PRF mechanism. PRRSV has two PRF sites, one of which is located at the end of ORF1a and generates ORF1ab through −1 PRF; the other PRF site is clever, the −1 and −2 PRF events occurs at the same site in the NSP2 coding region of ORF1a, resulting in two NSP2 variants. Respectively, it occurs in −1 PRF and generates a truncated structure compared to that of the full-length NSP2 called NSP2N, while the structure generated in −2 PRF contains an alternative C-terminal domain called NSP2TF [78,79].
At the core of most −1 PRFs are ribosomes interacting with a stimulatory mRNA structure (stem-loop or RNA pseudoknot), which promotes translational frameshifting of a stretch of polybases called a slippery sequence. PRRSV −2/−1 PRF is a highly efficient −2/−1 PRF event that functions without identifiable stimulating RNA secondary structure [78,79]. Specifically, pcbp interacts with PRRSV mRNA through KH1 and KH3, with each domain linked to one of two C-patches (CCCA and CUCC) in the C-rich region, which is located in the sliding sequence (GG GUU UUU) approximately 10 bp downstream. The KH2 domain binds to NSP1b instead of viral RNA, and the complex disrupts the normal sliding of ribosomes. The NSP1b-PCBP complex causes ribosome stalling, possibly as a result of direct interaction with the unwinding center of the small subunit, in a manner similar to that described for structured RNA stimuli [80]. An increasing number of similar mechanisms have been discovered, including Stm1 of Saccharomyces cerevisiae, which is a fragile, thereby inhibiting translation [81,82,83].
The −1/−2 PRF of NSP2 has important biological significance for PRRSV. The recombinant virus that silences NSP2TF severely affects virus replication and produces small plaques but is not lethal [78,84]. Traditionally, it has been shown that approximately 20% of ribosomes frameshift the translation of NSP2 into the −2 reading frame, thereby producing a transframe fusion protein (NSP2TF), and 7% of ribosomes enter the −1 reading frame, where they immediately encounter a stop codon, thus synthesizing a truncated version of NSP2 called NSP2N [79]. Georgia M. Cook found, through RNA seq methods, that −2 PRF efficiency, which may be promoted by PRF stimulating the accumulation of the viral protein NSP1β during infection, increased over time [85]. Before 2011, almost all PRRSV would immediately encounter the stop codon of NSP2 −1 PRF and stop transcription. However, the percentage of stop codon mutant strains increased rapidly after 2011. From 2011 to 2021, as many as 50% of the sequences lacked stop codons at this position. These dominant mutant strains came from farms where PRRSV outbreaks had occurred and were on the rise, such as pig farms [84]. Reverse genetics approaches were used to construct a mutant strain using SD9521 as the viral framework and VR2332 sliding cassette. The results of the Slippage to Construct box experiment showed that mutant strains carrying UGG or CGG at the −1 PRF site of the NSP2 gene had higher virus titers and higher −2 PRF efficiency. Surprisingly, two −1 PRF stop codon mutation patterns were identified to be predominantly prevalent in the field. These mutation patterns show higher growth kinetics than other mutant strains, indicating that the most dominant −1 PRF stop codon mutation pattern may enhance the growth adaptability of the virus [84], and we speculate that the mechanism may be different for base pair effects on the different binding abilities and gliding efficiencies of the NSP1b-PCBP complex and the small subunit. This strategy of PRRSV using PRF to produce polyproteins is important as it allows for viruses with smaller genomes to produce more proteins to regulate viral replication.

3. The Function of NSP2

3.1. The Relationship between NSP2 and Virus Assembly

NSP2 plays an important role in viral assembly [86,87]. PRRSV ORF1 translationally produces long-chain polypeptides, which need to be sheared to form polysaccharides to function and complete viral assembly. The plp2 functional domain of NSP2, which has both cis and trans cleavage activity, cleaves both its own and nonentropic polypeptides to form multiple NSP2 subunits (NSP2a, NSP2b, NSP2c, NSP2d, NSP2e and np2f) or is responsible for cleavage between NSP2 and NSP3. In addition, NSP2 cooperates with NSP4 to complete cleavage between NSP4 and NSP5. In addition, the NSP2 essential variant, NSP2TF, interacts with two major viral envelope proteins (GP5 glycoprotein and membrane (M) protein) to drive key processes in arterivirus assembly and outgrowth, while NSP2TF targets the cytosolic pathway to reduce the proteasome-driven turnover of the GP5/M protein, thereby facilitating the formation of the GP5-M dimer, which is essential for arterivirus assembly [86]. NSP2N and NSP2 may not share this function due to differences in subcellular localization or three-dimensional structure.
PRRSV is a positive-stranded RNA virus, and a key step in its replication is the assembly of the replication and transcription complex (RTC) [88], while the virus modifies the intracellular environment to generate bilayer vesicles that provide a barrier for itself. In addition, viruses also utilize aggregates (aggresomes), which provide a rich material base for viral replication and assembly, while being able to evade recognition by the immune system. In the arteritis virus EAV, NSP2 and NSP3 are involved in the formation of the RTC, a multiprotein complex that facilitates viral replication to modify cell membranes. NSP2 interacts with NSP3 to form replicative bilayer vesicles, which promote the formation of aggresomes [89,90]. PRRSV NSP2 and NSP3 have also been shown to interact [91]. It has been shown that PRRSV NSP2 recruits the Bcl2-associated antiapoptotic gene 6 (BAG6) protein to help localize in the endoplasmic reticulum, thereby promoting the formation of bilayer membrane vesicle structures. In addition, NSP2 binds to the cytosolic protein 14-3-3, an important protein involved in aggregate formation, through the highly variable region or the C-terminal tail region to promote aggresome formation, enhancing viral replication [87]. In addition, NSP2 has also been shown to be present among purified virus particles, but its specific function has not been elucidated. Therefore, NSP2 may be involved in PRRSV replication as a structural protein in addition to being a nonstructural protein.

3.2. The Relationship between NSP2 and Immunology

3.2.1. The Relationship between NSP2 and Innate Immunity

Natural immunity is important as the first line of host defense against viral infection. The intracellular RLR receptor RIG-I/MDA5 is an important molecule for the recognition of RNA viruses (Figure 2). RIG-I senses and binds to viral 5′ppp-RNA through the C-terminal CTD structural domain, undergoes a conformational change and releases the CARD structural domain, which in turn activates RIG-I via catalytic polyubiquitination of the K63 linkage of the CARD and CTD by ubiquitin ligases such as TRIM25, TRIM4 and Riplet (RNF135). RIG-I is activated by catalyzing the polyubiquitination of the K63 linkage of CARD and CTD. The zinc finger protein ZCCHC3 can act as a coreceptor for RLR to facilitate the binding of RIG-I and MDA5 to viral RNA and can promote polyubiquitination and activation of the K63 linkage of the CARD structural domains of RIG-I and MDA5 by TRIM25 [92]. The PRRSV NSP2 protein inhibits the ubiquitination activation of RIG-I (DDX58) by means of TRIM25 through plp2 deubiquitination activity, and ZCCHC3 knockdown also promotes PRRSV replication [92].
The structural domains of CARDs form tetramers after RIG-I activation and recruit MAVS proteins located in the outer mitochondrial membrane through CARD-CARD interactions. MAVS dimerizes and forms a complex with DDX3X and TRAF3, which enhances the activation of intrinsic immunity to anti-RNA viruses and in turn activates TBK1 and the IRF3 protein through the phosphorylation of TBK1. Phosphorylated IRF3 forms a dimer, enters the nucleus, binds to the promoter region of the IFN gene and induces the production of interferon; in addition, activated MAVS can also degrade IKBα through ubiquitination, and P65 enters the nucleus to induce the expression of antiviral proteins. The N-terminal cysteine protease domain of NSP2 attenuates the interaction of MAVS with the deconvolution enzyme DDX3X by interacting with the N-terminal DExDc and C-terminal HELICc domains of the enzyme. The interaction with MAVS is weakened by NSP2, which affects the level of MAVS-mediated IRF3 phosphorylation and prevents activation of the IFN promoter, thus antagonizing the antiviral effect of DDX3X. In addition, NSP2 inhibits the NF-KB pathway and inflammatory factor expression by deubiquitinating K48-linked IKBα, preventing its ubiquitination and degradation, and thereby retaining P65 in the cytoplasm [93]. Whether NSP2N and NSP2TF bind to DDX3X to inhibit IRF3 phosphorylation or deubiquitinate IKBα to suppress the expression of inflammatory factors has not been reported.
After interferon production, P65 is secreted outside the cell and binds to the surface receptor JAK in neighboring cells. The cytoplasmic region of the JAK kinase is phosphorylated and activated. STAT is transferred to a position near the JAK kinase and undergoes tyrosine phosphorylation. The tyrosine phosphorylated STAT starts to detach from the IFN receptor and forms a homodimer or a heterodimer, and the STAT1-2 heterodimer binds to IRF9 to form the activated transcriptional complex ISGF3. The STAT homodimer itself is a transcriptional activator, and the dimer transfers to the nucleus and induces the production of the antiviral protein ISG. ISG15 is an important antiviral protein that can exert antiviral effects through multimerization of the substrate, and it can also promote STAT1/STAT2 activation and ISGF3 formation through the interaction of free ISG15 with STAT2, which in turn promotes ISGF3-induced ISRE activation to exert free-state ISG15 antiviral effects [94]. NSP2 inhibits the expression and coupling of the antiviral protein ISG15 through the PLP2-DUB structural domain [93], resulting in the premature detachment of ISG15 from key signaling molecules. This mechanism acts as an emergency immune evasion mode for PRRSV, silencing the inflammatory response and IFN response [95], which in turn inhibits the amplification of IFN to adjacent cells. Recently, it was found that ISG15 acts as a substrate for TRIM21 glycosylation to enhance p62 ubiquitination to prevent autophagosome formation [96], and it is interesting to see whether NSP2 removes the inhibitory effect of autophagy via this pathway, although autophagy has been reported to be favorable in some reports [97]. There are no relevant reports on the role of NSP2N and NSP2TF in ISG15.

3.2.2. The Relationship between NSP2 and Acquired Immunity

NSP2, which contains B-cell epitopes and predicted T-cell epitopes, is an immunogenic protein of PRRSV [98], and high levels of antibodies comparable to or even higher than those against the N protein have been measured in porcine blood from one week to several months after viral infection [99,100,101,102]. Six of the ten linear B-cell epitopes contained in type 1 PRRSV were found to be present in NSP2 using phage display [98], and eighteen B-cell epitopes were found in NSP2 using peptide scanning, two of which are located at the position of deletions in the highly variable region of HP-PRRSV NSP2 [100]. Some researchers used GOLDKEY (IBMS, AMMS, Ürümqi, China) and DNASTAR software (Inc., Madison, WI, USA) to predict the dominant epitopes of NSP2 in BJ-4 and used peptide scanning to find the presence of six B-cellular epitopes at amino acids 73-567, of which SP4, SP5, SP6 and SP8 are immunodominant epitopes [103]. The same T-cell epitopes were found to exist for NSP2 by analysis and prediction [69]. Since NSP2 can be detected in purified virus particles, NSP2 may be present in virus particles as a structural protein, which may be the key reason why NSP2 antibodies are present at high levels in infected pig sera for a long period of time [104]. NSP2N and NSP2TF share the N-terminal region of NSP2 and therefore share all of the above characteristics as well.
The major histocompatibility complex (MHC) is a group of tightly interlinked genes encoding major histocompatibility antigens, i.e., leukocyte antigens. Porcine SLA-DR, the porcine leukocyte antigen, plays an important role in immunity, and the OTU domain of PRRSV NSP2 acts as a deubiquitinating enzyme, blocking ubiquitination and proteasome-mediated degradation of the α- and β-chains of SLA-DR and leading to the accumulation of SLA-DR proteins in PRRSV-infected cells. Naturally, PRRSV infections often result in SLA-DR-mediated NSP protein presentation and corresponding antibodies. NSP protein presentation and corresponding antibody production, thus, affect organism-specific immunity [93]. This ability of NSP2 may be responsible for the massive production of non-neutralized antibodies against PRRSV.
Since the emergence of PRRS in 1987, even though scientists have made continuous efforts to understand the pathogenesis and vaccinology of PRRSV, vaccines have not been successfully developed to prevent PRRSV widely and effectively, MLV is still the vaccine mainly used for prevention and control [105]. NSP2 is closely related to virulence and immunity and is one of the focuses of PRRSV vaccine research [106,107,108]. Earlier, the PRRSV TJM vaccine strain was found to have a sequential 120-amino-acid deletion (NSP2 628–747 aa) after a 30-amino-acid deletion in viral culture, which was thought to be related to its decreased virulence [102]. Subsequently, residues 500–596 and 658–777 of HP-PRRSV NSP2 were found to be required for the up-regulation of COX-2 expression and PGE2 production. The mutant HP-PRRS virus with residue 500–596 or 658–777 deletions had impaired ability to up-regulate COX-2 and PGE2 production in vitro and in vivo. Importantly, pigs infected with the variant virus can alleviate fever, clinical symptoms and mortality, providing clues for the development of an attenuated HP-PRRSV vaccine [109]. In fact, as early as 2009, some authors tried to construct a mutant NSP2 attenuated vaccine and proved that NSP2 is an important target for the development of tagged vaccines and virus attenuation [110]. Subsequently, two recombinant viruses that knocked out the ribosomal frameshift variants of NSP2, NSP2TF and NSP2N were shown to promote IFN-α responses, improve NK cell cytotoxicity and enhance T cell immune responses in infected pigs [105]. In addition, peptides 562–627 (m1B) and 749–813 (m2B) of the NSP2 protein were recently found to have no significant effect on viral replication. Therefore, m1B and m2B have the potential to be molecular markers for PRRSV vaccines [111]. Recently, the papasin-like protease 2 (PLP2) domain of PRRSV NSP2 was shown to be a target for neutralizing antibodies, and genetic variation in NSP2 resulted in poor cross-neutralization. When animals were immunized with the NSP2 chimeric strain, they were resistant to challenges with the NSP2 parent strain, but not with the backbone virus. In addition, neither the immune strain nor the challenge strain could be detected in vaccine-immunized animals, highlighting the important role of HP-PRRSV NSP2 in promoting viral clearance [112]. In conclusion, with a better understanding of NSP2 function, partial loss or retention of NSP2 will present opportunities for MLV vaccines.

3.3. The Relationship between NSP2 and Autophagy

Autophagy is a physiological process of material degradation–recycling through the lysosomal degradation of damaged organelles or proteins in eukaryotic cells [113], and it is also involved in the infection process of many viruses [114,115,116]. PRRSV can promote its own replication by inducing autophagy (Figure 3) [117,118].
Autophagy as an important physiological process in eukaryotic cells is negatively regulated through the PI3K-AKT-mTOR and MAPK-ERK-mTOR inactivation of ULK1 [119,120,121]. Therefore, autophagy is negatively regulated, or positively regulated through AMPK-ULK1. The researchers found that PRRSV infection induced autophagy in the following ways: PRRSV infection activates the opening of endoplasmic reticulum IP3R channels, allows Ca2+ into the cytoplasm, causes Ca2+ imbalance in the ER, induces endoplasmic reticulum stress, increases endoplasmic reticulum stress marker protein GRP78, activates the MAPK and PI3K signaling pathways, and promotes autophagy formation. On the other hand, endoplasmic reticulum calcium ion levels were reduced by the endoplasmic reticulum Ca2+ receptor STIM1, which accumulates and translocates to the vicinity of the plasma membrane [117]. The accumulated STIM1 protein then activates Orai1, prompting the opening of the CRAC calcium channel pore, extracellular calcium inward flow to the endoplasmic reticulum, endoplasmic reticulum Ca2+ influx into the cytoplasm via IP3R again and cytoplasmic Ca2+ level increases [122]. Calcium is a ubiquitous intracellular messenger involved in a variety of signaling processes, such as gene transcription, differentiation, proliferation and kinase activation [13]. Ca2+ and Ca2+ receptor proteins play important roles in the regulation of autophagy, and excessive release of Ca2+ from the endoplasmic reticulum activates the calmodulin-dependent protein kinase ii (CaMKII) and adenosine monophosphate-activated protein kinase (AMPK) signaling cascades and increases the lipidation of LC3II, leading to the inhibition of mTOR signaling and the induction of autophagy [123,124]. In addition, the ER is an important organelle for Ca2+ storage and protein processing and is highly sensitive to viral infection [125,126]. When endoplasmic reticulum function is disrupted, misfolded and unfolded, proteins accumulate in the lumen of the endoplasmic reticulum, interfering with Ca2+ homeostasis and leading to endoplasmic reticulum stress, which in turn induces autophagy [126]. PRRSV infection induces endoplasmic reticulum stress and disrupts Ca2+ homeostasis by means of a mechanism that ultimately activates autophagy through the CaMKII-AMPK-mTOR pathway in order to promote autophagy for self-replication [117]. CaMKII (a multifunctional Ca2+-dependent kinase) is activated in response to elevated Ca2+ levels, which in turn phosphorylates the downstream effector AMPK and enhances the lipidation of LC3II to promote autophagy. This process can be inhibited by 4-PBA, which suppresses PRRSV replication by inhibiting endoplasmic reticulum activation, and by ML-9 HCL, which suppresses viral replication by inhibiting SOCE channels [127]. The authors found that NSP2 interacts with STIM1 and GRP78 to induce autophagy by overexpressing Marc145 cells through a single gene [117]. The effect of NSP2 on the STIM1 and GRP78 proteins has not yet been demonstrated. Meanwhile, PRRSV-induced endoplasmic reticulum stress opens the SOCE channel to allow extracellular Ca2+ to enter the intracellular space. The SOCE channel is the main calcium channel on the plasma membrane of the cell, which mediates Ca2+ entry from the extracellular medium. Orai1 on the plasma membrane and STIM1 on the endoplasmic reticulum are the proteins responsible for SOCE channel activation [117].
In addition, it has been shown that NSP2 can activate the endoplasmic reticulum stress pathway by decreasing BAG6 expression through the interaction with BAG6, which affects the localization of NSP2 in the cytoplasm [128]. NSP2 binds to BAG6 and protects itself from ubiquitinated degradation through plp2 activity, and mutations in plp2(CH/AA) activity on NSP2 are ubiquitinated and degraded by BAG6. PLP2 activity also inhibits the ubiquitinated degradation of error proteins by BAG6, which in turn activates endoplasmic reticulum stress [128]. Interestingly, endoplasmic reticulum Ca2+ levels showed a decreasing trend at 2–4 h of viral infection, whereas NSP2 was gradually expressed after 4 h [129]. Therefore, the initial activation of IP3R channels may not be induced by NSP2, such as lymphocytes being activated by antigens and consequently Ca2+ translocation to the cytoplasm [130]. The reason why NSP2 expression alone can also induce endoplasmic reticulum stress may be closely related to the fact that NSP2 is rich in B-cell and T-cell epitopes. Recently, it has been found that cells can use autophagy to inhibit PRRSV NSP1α protein expression to suppress viral replication [97], which is contrary to the conclusion that autophagy promotes PRRSV replication, and it is clear that this game of virus–host interaction is complex and seemingly contradictory.

3.4. The Relationship between NSP2 and Apoptosis

Apoptosis is the programmed cell death of multicellular organisms that occurs in an autonomous and orderly manner, under certain physiological and pathological conditions, and is controlled by their own genes to maintain the stability of their own internal environment [131]. Viral infection and apoptosis have been hot topics of research, and PRRSV infection can cause apoptosis in a variety of organs, tissues and cells in pigs, such as the lungs, lymphatic system, testes, placenta, etc. The infected cells are mainly macrophages, lymphocytes, germ cells and dendritic cells [132,133,134,135,136]. Recently, it has been reported in the literature that NSP2 induces apoptosis, which is second only to NSP4 in the intensity of apoptosis induced, but the mechanism is not clear [137]. It has been shown that NSP2 can inhibit the ubiquitin proteasome pathway degradation of AIF1, which in turn causes no caspase-dependent apoptosis through the endoplasmic reticulum stress pathway, and its binding ability can be abolished through mutation of the plp2 active site, in addition to the caspase-dependent apoptosis induced by NSP2 [128]. It has been shown that PRRSV-1 cannot cause caspase-independent apoptosis, which may be related to the large difference in NSP2 between the two [138].
CD2AP and SH3KBP1 belong to a group of “scaffolding molecules” that regulate the actin cytoskeleton and can affect endocytosis, cell death, cell adhesion and division through anchoring and remodeling of actin [139]. It was found that NSP2 binds to CD2AP and SH3KBP1 [87]. The biological significance of NSP2 binding to SH3KBP1 has not yet been clearly demonstrated. The NSP2 highly variable region proxylate motif (PXXXPR motif) interacts with the SH3 domain region of CD2AP, resulting in the aggregation of CD2AP around the nucleus, altered subcellular localization and induced CatL-dependent degradation of CD2AP [87]. On the one hand, the degradation of CD2AP TGF-b1 secretion is also promoted, which can inactivate the anti-regulatory signal TGF-b1-CD2AP-PI3K-ATK and enhance the pro-apoptotic signal TGF-b1-Smad3-P38 [140], but interestingly, PI3K-ATK is in an inhibited state only in the middle stage of PRRSV infection [141], which may be the result of the virus in the middle stage of infection in order to breaking the barrier effect of alveolar epithelial cells for invasion of the organism and cell-to-cell spreading. On the other hand, the degradation of CD2AP disrupts the cytoskeleton, leading to defective cell division and more cells arresting in the S phase [142]. In addition, it has been found that, in inflammation, macrophages deficient in CD2AP can lose their motility [143], which may attenuate the phagocytosis of PRRSV by macrophages. Thus, NSP2 plays a regulatory role in various aspects, such as apoptosis.

4. Conclusions

PRRSV, a virus with a long history and a serious threat to the global pig farming industry, has been the subject of extensive basic research. Important progress has been made in the treatment and prevention of PRRSV, also known as blue ear disease, but there is still no highly effective, cross-protective vaccine for clinical prevention. NSP2 plays an important role in the maturation of PRRSV proteins, assembly of viral particles, modulation of the host immune response, cellular autophagy, apoptosis, alteration of the cytoskeleton and regulation of the cell cycle (Figure 4). In addition, NSP2 uses −1/2 PRF to translate its own nucleic acid sequence into the NSP2N and NSP2TF variants, which have the same or similar functions in many aspects while retaining their own unique functions because all three of these proteins have identical amino-terminal domains, especially the PLP2 domain. It seems that −1/2 PRF can also be regarded as an opportunity to inhibit viral replication by blocking the −1/2 PRF site to achieve therapeutic effects. NSP2 of PRRSV is characterized by recombination, deletion and high mutation, and the mechanism of its −1/2 PRF site makes the study of the protein challenging. The complex and varied structural composition of NSP2 and its wide range of functional characteristics make the protein a mystery.
In this paper, we provide a more detailed depiction of the PRRSV NSP2 structure, function and genetic variation to provide a reference for basic research on PRRSV and to facilitate a deeper exploration of NSP2 function. We found that NSP2 functions are many and varied, but there are few studies related to the comparison of the functional variability of NSP2, NSP2N and NSP2TF. What are the roles played by each of them or synergistically, what are the functions unique to each of them and what are the shared functions, and what determines these functions? In addition, some studies have shown that NSP2 can also exist as a structural protein, but what is its specific functional significance? There are many unanswered questions that scientists need to continue to explore to solidify basic research on PRRSV and to fight for the prevention and control of blue ear disease and for the development of the global pig farming industry.

Author Contributions

J.C., B.L., L.L. and Z.S. contributed to the study’s conception, the research design and manuscript writing. B.L., Z.S., H.J. and L.L. performed the data curation and analysis. L.Y., G.L. and J.C. supervised this study and contributed to the manuscript revision and funding acquisition. All authors have read and agreed to the published version of the manuscript provided.

Funding

This research was funded by Shanghai Municipal Agriculture Science and Technology Project (Grant No. 2022-02-08-00-12-F01115) and the Northeast Agricultural university high level university funds (Grant No. 54917612).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Shi Fu for her assistance with the preparation of this manuscript.

Conflicts of Interest

The authors declare that there are no conflict of interest regarding the publication of this paper.

References

  1. Rossow, K.D. Porcine reproductive and respiratory syndrome. Vet. Pathol. 1998, 35, 1–20. [Google Scholar] [CrossRef] [PubMed]
  2. Brinton, M.A.; Gulyaeva, A.A.; Balasuriya, U.B.R.; Dunowska, M.; Faaberg, K.S.; Goldberg, T.; Leung, F.C.C.; Nauwynck, H.J.; Snijder, E.J.; Stadejek, T.; et al. ICTV Virus Taxonomy Profile: Arteriviridae 2021. J. Gen. Virol. 2021, 102, 001632. [Google Scholar] [CrossRef] [PubMed]
  3. Kuhn, J.H.; Lauck, M.; Bailey, A.L.; Shchetinin, A.M.; Vishnevskaya, T.V.; Bào, Y.; Ng, T.F.; LeBreton, M.; Schneider, B.S.; Gillis, A.; et al. Reorganization and expansion of the nidoviral family Arteriviridae. Arch. Virol. 2016, 161, 755–768. [Google Scholar] [CrossRef] [PubMed]
  4. Karniychuk, U.U.; Geldhof, M.; Vanhee, M.; Van Doorsselaere, J.; Saveleva, T.A.; Nauwynck, H.J. Pathogenesis and antigenic characterization of a new East European subtype 3 porcine reproductive and respiratory syndrome virus isolate. BMC Vet. Res. 2010, 6, 30. [Google Scholar] [CrossRef] [PubMed]
  5. Tian, K.; Yu, X.; Zhao, T.; Feng, Y.; Cao, Z.; Wang, C.; Hu, Y.; Chen, X.; Hu, D.; Tian, X.; et al. Emergence of fatal PRRSV variants: Unparalleled outbreaks of atypical PRRS in China and molecular dissection of the unique hallmark. PLoS ONE 2007, 2, e526. [Google Scholar] [CrossRef] [PubMed]
  6. Gao, J.C.; Xiong, J.Y.; Ye, C.; Chang, X.B.; Guo, J.C.; Jiang, C.G.; Zhang, G.H.; Tian, Z.J.; Cai, X.H.; Tong, G.Z.; et al. Genotypic and geographical distribution of porcine reproductive and respiratory syndrome viruses in mainland China in 1996–2016. Vet. Microbiol. 2017, 208, 164–172. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, H.L.; Zhang, W.L.; Xiang, L.R.; Leng, C.L.; Tian, Z.J.; Tang, Y.D.; Cai, X.H. Emergence of novel porcine reproductive and respiratory syndrome viruses (ORF5 RFLP 1-7-4 viruses) in China. Vet. Microbiol. 2018, 222, 105–108. [Google Scholar] [CrossRef] [PubMed]
  8. Xu, H.; Song, S.; Zhao, J.; Leng, C.; Fu, J.; Li, C.; Tang, Y.D.; Xiang, L.; Peng, J.; Wang, Q.; et al. A potential endemic strain in China: NADC34-like porcine reproductive and respiratory syndrome virus. Transbound. Emerg. Dis. 2020, 67, 1730–1738. [Google Scholar] [CrossRef]
  9. Zhou, L.; Wang, Z.; Ding, Y.; Ge, X.; Guo, X.; Yang, H. NADC30-like Strain of Porcine Reproductive and Respiratory Syndrome Virus, China. Emerg. Infect. Dis. 2015, 21, 2256–2257. [Google Scholar] [CrossRef]
  10. Canelli, E.; Catella, A.; Borghetti, P.; Ferrari, L.; Ogno, G.; De Angelis, E.; Corradi, A.; Passeri, B.; Bertani, V.; Sandri, G.; et al. Phenotypic characterization of a highly pathogenic Italian porcine reproductive and respiratory syndrome virus (PRRSV) type 1 subtype 1 isolate in experimentally infected pigs. Vet. Microbiol. 2017, 210, 124–133. [Google Scholar] [CrossRef]
  11. Martín-Valls, G.E.; Cortey, M.; Allepuz, A.; Illas, F.; Tello, M.; Mateu, E. Introduction of a PRRSV-1 strain of increased virulence in a pig production structure in Spain: Virus evolution and impact on production. Porc. Health Manag. 2023, 9, 1. [Google Scholar] [CrossRef] [PubMed]
  12. Romeo, C.; Parisio, G.; Scali, F.; Tonni, M.; Santucci, G.; Maisano, A.M.; Barbieri, I.; Boniotti, M.B.; Stadejek, T.; Alborali, G.L. Complex interplay between PRRSV-1 genetic diversity, coinfections and antimicrobial use influences performance parameters in post-weaning pigs. Vet. Microbiol. 2023, 284, 109830. [Google Scholar] [CrossRef] [PubMed]
  13. Xu, H.; Li, C.; Li, W.; Zhao, J.; Gong, B.; Sun, Q.; Tang, Y.D.; Xiang, L.; Leng, C.; Peng, J.; et al. Novel characteristics of Chinese NADC34-like PRRSV during 2020–2021. Transbound. Emerg. Dis. 2022, 69, e3215–e3224. [Google Scholar] [CrossRef] [PubMed]
  14. Russell, P.; Atkinson, K.; Krishnan, L. Recurrent reproductive failure due to severe placental villitis of unknown etiology. J. Reprod. Med. 1980, 24, 93–98. [Google Scholar] [PubMed]
  15. Wensvoort, G.; Terpstra, C.; Pol, J.M.; ter Laak, E.A.; Bloemraad, M.; de Kluyver, E.P.; Kragten, C.; van Buiten, L.; den Besten, A.; Wagenaar, F.; et al. Mystery swine disease in The Netherlands: The isolation of Lelystad virus. Vet. Q. 1991, 13, 121–130. [Google Scholar] [CrossRef]
  16. Kim, H.S.; Kwang, J.; Yoon, I.J.; Joo, H.S.; Frey, M.L. Enhanced replication of porcine reproductive and respiratory syndrome (PRRS) virus in a homogeneous subpopulation of MA-104 cell line. Arch. Virol. 1993, 133, 477–483. [Google Scholar] [CrossRef] [PubMed]
  17. Kuwahara, H.; Nunoya, T.; Tajima, M.; Kato, A.; Samejima, T. An outbreak of porcine reproductive and respiratory syndrome in Japan. J. Vet. Med. Sci. 1994, 56, 901–909. [Google Scholar] [CrossRef] [PubMed]
  18. Meng, X.J.; Paul, P.S.; Halbur, P.G.; Lum, M.A. Characterization of a high-virulence US isolate of porcine reproductive and respiratory syndrome virus in a continuous cell line, ATCC CRL11171. J. Vet. Diagn. Investig. 1996, 8, 374–381. [Google Scholar] [CrossRef]
  19. Mengeling, W.L.; Lager, K.M.; Vorwald, A.C. Clinical consequences of exposing pregnant gilts to strains of porcine reproductive and respiratory syndrome (PRRS) virus isolated from field cases of “atypical” PRRS. Am. J. Vet. Res. 1998, 59, 1540–1544. [Google Scholar]
  20. Han, J.; Wang, Y.; Faaberg, K.S. Complete genome analysis of RFLP 184 isolates of porcine reproductive and respiratory syndrome virus. Virus Res. 2006, 122, 175–182. [Google Scholar] [CrossRef]
  21. Snijder, E.J.; Kikkert, M.; Fang, Y. Arterivirus molecular biology and pathogenesis. J. Gen. Virol. 2013, 94 Pt 10, 2141–2163. [Google Scholar] [CrossRef] [PubMed]
  22. Music, N.; Gagnon, C.A. The role of porcine reproductive and respiratory syndrome (PRRS) virus structural and non-structural proteins in virus pathogenesis. Anim. Health Res. Rev. 2010, 11, 135–163. [Google Scholar] [CrossRef]
  23. Kappes, M.A.; Miller, C.L.; Faaberg, K.S. Porcine reproductive and respiratory syndrome virus nonstructural protein 2 (nsp2) topology and selective isoform integration in artificial membranes. Virology 2015, 481, 51–62. [Google Scholar] [CrossRef] [PubMed]
  24. Darwich, L.; Gimeno, M.; Sibila, M.; Diaz, I.; de la Torre, E.; Dotti, S.; Kuzemtseva, L.; Martin, M.; Pujols, J.; Mateu, E. Genetic and immunobiological diversities of porcine reproductive and respiratory syndrome genotype I strains. Vet. Microbiol. 2011, 150, 49–62. [Google Scholar] [CrossRef] [PubMed]
  25. Kroese, M.V.; Zevenhoven-Dobbe, J.C.; Bos-de Ruijter, J.N.A.; Peeters, B.P.H.; Meulenberg, J.J.M.; Cornelissen, L.; Snijder, E.J. The nsp1alpha and nsp1 papain-like autoproteinases are essential for porcine reproductive and respiratory syndrome virus RNA synthesis. J. Gen. Virol. 2008, 89 Pt 2, 494–499. [Google Scholar] [CrossRef] [PubMed]
  26. Tijms, M.A.; van Dinten, L.C.; Gorbalenya, A.E.; Snijder, E.J. A zinc finger-containing papain-like protease couples subgenomic mRNA synthesis to genome translation in a positive-stranded RNA virus. Proc. Natl. Acad. Sci. USA 2001, 98, 1889–1894. [Google Scholar] [CrossRef] [PubMed]
  27. Han, J.; Rutherford, M.S.; Faaberg, K.S. Proteolytic products of the porcine reproductive and respiratory syndrome virus nsp2 replicase protein. J. Virol. 2010, 84, 10102–10112. [Google Scholar] [CrossRef]
  28. Yun, S.I.; Lee, Y.M. Overview: Replication of porcine reproductive and respiratory syndrome virus. J. Microbiol. 2013, 51, 711–723. [Google Scholar] [CrossRef]
  29. Zhang, W.; Chen, K.; Guo, Y.; Chen, Y.; Liu, X. Involvement of PRRSV NSP3 and NSP5 in the autophagy process. Virol. J. 2019, 16, 13. [Google Scholar] [CrossRef]
  30. Huang, C.; Du, Y.; Yu, Z.; Zhang, Q.; Liu, Y.; Tang, J.; Shi, J.; Feng, W.H. Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Nsp4 Cleaves VISA to Impair Antiviral Responses Mediated by RIG-I-like Receptors. Sci. Rep. 2016, 6, 28497. [Google Scholar] [CrossRef]
  31. Huang, C.; Zhang, Q.; Guo, X.K.; Yu, Z.B.; Xu, A.T.; Tang, J.; Feng, W.H. Porcine reproductive and respiratory syndrome virus nonstructural protein 4 antagonizes beta interferon expression by targeting the NF-κB essential modulator. J. Virol. 2014, 88, 10934–10945. [Google Scholar] [CrossRef] [PubMed]
  32. Tao, R.; Fang, L.; Bai, D.; Ke, W.; Zhou, Y.; Wang, D.; Xiao, S. Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 4 Cleaves Porcine DCP1a To Attenuate Its Antiviral Activity. J. Immunol. 2018, 201, 2345–2353. [Google Scholar] [CrossRef]
  33. Tian, X.; Lu, G.; Gao, F.; Peng, H.; Feng, Y.; Ma, G.; Bartlam, M.; Tian, K.; Yan, J.; Hilgenfeld, R.; et al. Structure and cleavage specificity of the chymotrypsin-like serine protease (3CLSP/nsp4) of Porcine Reproductive and Respiratory Syndrome Virus (PRRSV). J. Mol. Biol. 2009, 392, 977–993. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, Y.; Song, Z.; Bai, J.; Liu, X.; Nauwynck, H.; Jiang, P. Porcine reproductive and respiratory syndrome virus Nsp4 cleaves ZAP to antagonize its antiviral activity. Vet. Microbiol. 2020, 250, 108863. [Google Scholar] [CrossRef]
  35. Lunney, J.K.; Fang, Y.; Ladinig, A.; Chen, N.; Li, Y.; Rowland, B.; Renukaradhya, G.J. Porcine Reproductive and Respiratory Syndrome Virus (PRRSV): Pathogenesis and Interaction with the Immune System. Annu. Rev. Anim. Biosci. 2016, 4, 129–154. [Google Scholar] [CrossRef]
  36. Mokhtar, H.; Pedrera, M.; Frossard, J.P.; Biffar, L.; Hammer, S.E.; Kvisgaard, L.K.; Larsen, L.E.; Stewart, G.R.; Somavarapu, S.; Steinbach, F.; et al. The Non-structural Protein 5 and Matrix Protein Are Antigenic Targets of T Cell Immunity to Genotype 1 Porcine Reproductive and Respiratory Syndrome Viruses. Front. Immunol. 2016, 7, 40. [Google Scholar] [CrossRef] [PubMed]
  37. Kwon, B.; Ansari, I.H.; Pattnaik, A.K.; Osorio, F.A. Identification of virulence determinants of porcine reproductive and respiratory syndrome virus through construction of chimeric clones. Virology 2008, 380, 371–378. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, M.; Cao, Z.; Xie, J.; Zhu, W.; Zhou, P.; Gu, H.; Sun, L.; Su, S.; Zhang, G. Mutagenesis analysis of porcine reproductive and respiratory syndrome virus nonstructural protein 7. Virus Genes. 2013, 47, 467–477. [Google Scholar] [CrossRef]
  39. Beerens, N.; Selisko, B.; Ricagno, S.; Imbert, I.; van der Zanden, L.; Snijder, E.J.; Canard, B. De novo initiation of RNA synthesis by the arterivirus RNA-dependent RNA polymerase. J. Virol. 2007, 81, 8384–8395. [Google Scholar] [CrossRef]
  40. Li, Y.; Zhou, L.; Zhang, J.; Ge, X.; Zhou, R.; Zheng, H.; Geng, G.; Guo, X.; Yang, H. Nsp9 and Nsp10 contribute to the fatal virulence of highly pathogenic porcine reproductive and respiratory syndrome virus emerging in China. PLoS Pathog. 2014, 10, e1004216. [Google Scholar] [CrossRef]
  41. Posthuma, C.C.; Te Velthuis, A.J.W.; Snijder, E.J. Nidovirus RNA polymerases: Complex enzymes handling exceptional RNA genomes. Virus Res. 2017, 234, 58–73. [Google Scholar] [CrossRef] [PubMed]
  42. Xu, L.; Zhou, L.; Sun, W.; Zhang, P.; Ge, X.; Guo, X.; Han, J.; Yang, H. Nonstructural protein 9 residues 586 and 592 are critical sites in determining the replication efficiency and fatal virulence of the Chinese highly pathogenic porcine reproductive and respiratory syndrome virus. Virology 2018, 517, 135–147. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, K.; Gao, J.C.; Xiong, J.Y.; Guo, J.C.; Yang, Y.B.; Jiang, C.G.; Tang, Y.D.; Tian, Z.J.; Cai, X.H.; Tong, G.Z.; et al. Two Residues in NSP9 Contribute to the Enhanced Replication and Pathogenicity of Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus. J. Virol. 2018, 92, e02209-17. [Google Scholar] [CrossRef] [PubMed]
  44. Bautista, E.M.; Faaberg, K.S.; Mickelson, D.; McGruder, E.D. Functional properties of the predicted helicase of porcine reproductive and respiratory syndrome virus. Virology 2002, 298, 258–270. [Google Scholar] [CrossRef] [PubMed]
  45. Fang, Y.; Snijder, E.J. The PRRSV replicase: Exploring the multifunctionality of an intriguing set of nonstructural proteins. Virus Res. 2010, 154, 61–76. [Google Scholar] [CrossRef] [PubMed]
  46. Nedialkova, D.D.; Ulferts, R.; van den Born, E.; Lauber, C.; Gorbalenya, A.E.; Ziebuhr, J.; Snijder, E.J. Biochemical characterization of arterivirus nonstructural protein 11 reveals the nidovirus-wide conservation of a replicative endoribonuclease. J. Virol. 2009, 83, 5671–5682. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, D.; Fan, J.; Fang, L.; Luo, R.; Ouyang, H.; Ouyang, C.; Zhang, H.; Chen, H.; Li, K.; Xiao, S. The nonstructural protein 11 of porcine reproductive and respiratory syndrome virus inhibits NF-κB signaling by means of its deubiquitinating activity. Mol. Immunol. 2015, 68 Pt A, 357–366. [Google Scholar] [CrossRef]
  48. Bai, Y.; Li, L.; Shan, T.; Zhang, Y.; Chen, X.; Gao, F.; Jiang, Y.; Zhou, Y.; Li, G.; Yu, L.; et al. Proteasomal degradation of nonstructural protein 12 by RNF114 suppresses porcine reproductive and respiratory syndrome virus replication. Vet. Microbiol. 2020, 246, 108746. [Google Scholar] [CrossRef]
  49. Li, L.; Bai, Y.; Zhou, Y.; Jiang, Y.; Tong, W.; Li, G.; Zheng, H.; Gao, F.; Tong, G. PSMB1 Inhibits the Replication of Porcine Reproductive and Respiratory Syndrome Virus by Recruiting NBR1 To Degrade Nonstructural Protein 12 by Autophagy. J. Virol. 2023, 97, e0166022. [Google Scholar] [CrossRef]
  50. Wang, T.Y.; Fang, Q.Q.; Cong, F.; Liu, Y.G.; Wang, H.M.; Zhang, H.L.; Tian, Z.J.; Tang, Y.D.; Cai, X.H. The Nsp12-coding region of type 2 PRRSV is required for viral subgenomic mRNA synthesis. Emerg. Microbes Infect. 2019, 8, 1501–1510. [Google Scholar] [CrossRef]
  51. Shi, X.; Fan, X.; Nie, S.; Kou, L.; Zhang, X.; Liu, H.; Ji, S.; Deng, R.; Wang, A.; Zhang, G. Identification of a linear B-cell epitope on glycoprotein (GP) 2a of porcine reproductive and respiratory syndrome virus (PRRSV). Int. J. Biol. Macromol. 2019, 139, 1288–1294. [Google Scholar] [CrossRef]
  52. Liu, L.; Tian, J.; Nan, H.; Tian, M.; Li, Y.; Xu, X.; Huang, B.; Zhou, E.; Hiscox, J.A.; Chen, H. Porcine Reproductive and Respiratory Syndrome Virus Nucleocapsid Protein Interacts with Nsp9 and Cellular DHX9 To Regulate Viral RNA Synthesis. J. Virol. 2016, 90, 5384–5398. [Google Scholar] [CrossRef] [PubMed]
  53. Snijder, E.J.; van Tol, H.; Pedersen, K.W.; Raamsman, M.J.; de Vries, A.A. Identification of a novel structural protein of arteriviruses. J. Virol. 1999, 73, 6335–6345. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, C.; Yoo, D. The small envelope protein of porcine reproductive and respiratory syndrome virus possesses ion channel protein-like properties. Virology 2006, 355, 30–43. [Google Scholar] [CrossRef] [PubMed]
  55. Ding, G.; Liu, J.; Shao, Q.; Wang, B.; Feng, J.; Li, Y.; Li, L.; Cao, S.; Cong, F.; Zhao, Y.; et al. Porcine Reproductive and Respiratory Syndrome Virus Structural Protein GP3 Regulates Claudin 4 To Facilitate the Early Stages of Infection. J. Virol. 2020, 94, e00124-20. [Google Scholar] [CrossRef]
  56. Vu, H.L.; Kwon, B.; Yoon, K.J.; Laegreid, W.W.; Pattnaik, A.K.; Osorio, F.A. Immune evasion of porcine reproductive and respiratory syndrome virus through glycan shielding involves both glycoprotein 5 as well as glycoprotein 3. J. Virol. 2011, 85, 5555–5564. [Google Scholar] [CrossRef] [PubMed]
  57. Costers, S.; Vanhee, M.; Van Breedam, W.; Van Doorsselaere, J.; Geldhof, M.; Nauwynck, H.J. GP4-specific neutralizing antibodies might be a driving force in PRRSV evolution. Virus Res. 2010, 154, 104–113. [Google Scholar] [CrossRef] [PubMed]
  58. Prather, R.S.; Rowland, R.R.; Ewen, C.; Trible, B.; Kerrigan, M.; Bawa, B.; Teson, J.M.; Mao, J.; Lee, K.; Samuel, M.S.; et al. An intact sialoadhesin (Sn/SIGLEC1/CD169) is not required for attachment/internalization of the porcine reproductive and respiratory syndrome virus. J. Virol. 2013, 87, 9538–9546. [Google Scholar] [CrossRef]
  59. Ostrowski, M.; Galeota, J.A.; Jar, A.M.; Platt, K.B.; Osorio, F.A.; Lopez, O.J. Identification of neutralizing and nonneutralizing epitopes in the porcine reproductive and respiratory syndrome virus GP5 ectodomain. J. Virol. 2002, 76, 4241–4250. [Google Scholar] [CrossRef]
  60. Bautista, E.M.; Molitor, T.W. Cell-mediated immunity to porcine reproductive and respiratory syndrome virus in swine. Viral Immunol. 1997, 10, 83–94. [Google Scholar] [CrossRef]
  61. Cancel-Tirado, S.M.; Evans, R.B.; Yoon, K.J. Monoclonal antibody analysis of porcine reproductive and respiratory syndrome virus epitopes associated with antibody-dependent enhancement and neutralization of virus infection. Vet. Immunol. Immunopathol. 2004, 102, 249–262. [Google Scholar] [CrossRef] [PubMed]
  62. Jeong, H.J.; Song, Y.J.; Lee, S.W.; Lee, J.B.; Park, S.Y.; Song, C.S.; Ha, G.W.; Oh, J.S.; Oh, Y.K.; Choi, I.S. Comparative measurement of cell-mediated immune responses of swine to the M and N proteins of porcine reproductive and respiratory syndrome virus. Clin. Vaccine Immunol. 2010, 17, 503–512. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, L.; Frey, M.L.; Yoon, K.J.; Zimmerman, J.J.; Platt, K.B. Categorization of North American porcine reproductive and respiratory syndrome viruses: Epitopic profiles of the N, M, GP5 and GP3 proteins and susceptibility to neutralization. Arch. Virol. 2000, 145, 1599–1619. [Google Scholar] [CrossRef] [PubMed]
  64. Loemba, H.D.; Mounir, S.; Mardassi, H.; Archambault, D.; Dea, S. Kinetics of humoral immune response to the major structural proteins of the porcine reproductive and respiratory syndrome virus. Arch. Virol. 1996, 141, 751–761. [Google Scholar] [CrossRef] [PubMed]
  65. Van Breedam, W.; Van Gorp, H.; Zhang, J.Q.; Crocker, P.R.; Delputte, P.L.; Nauwynck, H.J. The M/GP(5) glycoprotein complex of porcine reproductive and respiratory syndrome virus binds the sialoadhesin receptor in a sialic acid-dependent manner. PLoS Pathog. 2010, 6, e1000730. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, Y.; Yu, Z.; Yi, H.; Wei, Y.; Han, X.; Li, Q.; Ji, C.; Huang, J.; Deng, Q.; Liu, Y.; et al. The phosphorylation of the N protein could affect PRRSV virulence in vivo. Vet. Microbiol. 2019, 231, 226–231. [Google Scholar] [CrossRef]
  67. Das, P.B.; Dinh, P.X.; Ansari, I.H.; de Lima, M.; Osorio, F.A.; Pattnaik, A.K. The minor envelope glycoproteins GP2a and GP4 of porcine reproductive and respiratory syndrome virus interact with the receptor CD163. J. Virol. 2010, 84, 1731–1740. [Google Scholar] [CrossRef]
  68. Veit, M.; Matczuk, A.K.; Sinhadri, B.C.; Krause, E.; Thaa, B. Membrane proteins of arterivirus particles: Structure, topology, processing and function. Virus Res. 2014, 194, 16–36. [Google Scholar] [CrossRef]
  69. Fang, Y.; Kim, D.Y.; Ropp, S.; Steen, P.; Christopher-Hennings, J.; Nelson, E.A.; Rowland, R.R. Heterogeneity in Nsp2 of European-like porcine reproductive and respiratory syndrome viruses isolated in the United States. Virus Res. 2004, 100, 229–235. [Google Scholar] [CrossRef]
  70. Liu, D.; Zhou, R.; Zhang, J.; Zhou, L.; Jiang, Q.; Guo, X.; Ge, X.; Yang, H. Recombination analyses between two strains of porcine reproductive and respiratory syndrome virus in vivo. Virus Res. 2011, 155, 473–486. [Google Scholar] [CrossRef]
  71. Han, J.; Rutherford, M.S.; Faaberg, K.S. The porcine reproductive and respiratory syndrome virus nsp2 cysteine protease domain possesses both trans- and cis-cleavage activities. J. Virol. 2009, 83, 9449–9463. [Google Scholar] [CrossRef] [PubMed]
  72. Zhou, S.; Ge, X.; Kong, C.; Liu, T.; Liu, A.; Gao, P.; Song, J.; Zhou, L.; Guo, X.; Han, J.; et al. Characterizing the PRRSV nsp2 Deubiquitinase Reveals Dispensability of Cis-Activity for Replication and a Link of nsp2 to Inflammation Induction. Viruses 2019, 11, 896. [Google Scholar] [CrossRef] [PubMed]
  73. Han, J.; Liu, G.; Wang, Y.; Faaberg, K.S. Identification of nonessential regions of the nsp2 replicase protein of porcine reproductive and respiratory syndrome virus strain VR-2332 for replication in cell culture. J. Virol. 2007, 81, 9878–9890. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, L.; Zhang, J.; Zeng, J.; Yin, S.; Li, Y.; Zheng, L.; Guo, X.; Ge, X.; Yang, H. The 30-amino-acid deletion in the Nsp2 of highly pathogenic porcine reproductive and respiratory syndrome virus emerging in China is not related to its virulence. J. Virol. 2009, 83, 5156–5167. [Google Scholar] [CrossRef]
  75. Jacks, T.; Madhani, H.D.; Masiarz, F.R.; Varmus, H.E. Signals for ribosomal frameshifting in the Rous sarcoma virus gag-pol region. Cell 1988, 55, 447–458. [Google Scholar] [CrossRef] [PubMed]
  76. Jacks, T.; Townsley, K.; Varmus, H.E.; Majors, J. Two efficient ribosomal frameshifting events are required for synthesis of mouse mammary tumor virus gag-related polyproteins. Proc. Natl. Acad. Sci. USA 1987, 84, 4298–4302. [Google Scholar] [CrossRef] [PubMed]
  77. Jacks, T.; Varmus, H.E. Expression of the Rous sarcoma virus pol gene by ribosomal frameshifting. Science 1985, 230, 1237–1242. [Google Scholar] [CrossRef]
  78. Fang, Y.; Treffers, E.E.; Li, Y.; Tas, A.; Sun, Z.; van der Meer, Y.; de Ru, A.H.; van Veelen, P.A.; Atkins, J.F.; Snijder, E.J.; et al. Efficient -2 frameshifting by mammalian ribosomes to synthesize an additional arterivirus protein. Proc. Natl. Acad. Sci. USA 2012, 109, E2920–E2928. [Google Scholar] [CrossRef]
  79. Li, Y.; Treffers, E.E.; Napthine, S.; Tas, A.; Zhu, L.; Sun, Z.; Bell, S.; Mark, B.L.; van Veelen, P.A.; van Hemert, M.J.; et al. Transactivation of programmed ribosomal frameshifting by a viral protein. Proc. Natl. Acad. Sci. USA 2014, 111, E2172–E2181. [Google Scholar] [CrossRef]
  80. Napthine, S.; Treffers, E.E.; Bell, S.; Goodfellow, I.; Fang, Y.; Firth, A.E.; Snijder, E.J.; Brierley, I. A novel role for poly(C) binding proteins in programmed ribosomal frameshifting. Nucleic Acids Res. 2016, 44, 5491–5503. [Google Scholar] [CrossRef]
  81. Balagopal, V.; Parker, R. Stm1 modulates translation after 80S formation in Saccharomyces cerevisiae. RNA 2011, 17, 835–842. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, E.; Sharma, M.R.; Shi, X.; Agrawal, R.K.; Joseph, S. Fragile X mental retardation protein regulates translation by binding directly to the ribosome. Mol. Cell 2014, 54, 407–417. [Google Scholar] [CrossRef] [PubMed]
  83. Darnell, J.C.; Van Driesche, S.J.; Zhang, C.; Hung, K.Y.; Mele, A.; Fraser, C.E.; Stone, E.F.; Chen, C.; Fak, J.J.; Chi, S.W.; et al. FMRP stalls ribosomal translocation on mRNAs linked to synaptic function and autism. Cell 2011, 146, 247–261. [Google Scholar] [CrossRef] [PubMed]
  84. Yan, X.; Shang, P.; Yim-Im, W.; Sun, Y.; Zhang, J.; Firth, A.E.; Lowe, J.F.; Fang, Y. Molecular characterization of emerging variants of PRRSV in the United States: New features of the -2/-1 programmed ribosomal frameshifting signal in the nsp2 region. Virology 2022, 573, 39–49. [Google Scholar] [CrossRef] [PubMed]
  85. Cook, G.M.; Brown, K.; Shang, P.; Li, Y.; Soday, L.; Dinan, A.M.; Tumescheit, C.; Mockett, A.P.A.; Fang, Y.; Firth, A.E.; et al. Ribosome profiling of porcine reproductive and respiratory syndrome virus reveals novel features of viral gene expression. Elife 2022, 11, e75668. [Google Scholar] [CrossRef] [PubMed]
  86. Guo, R.; Yan, X.; Li, Y.; Cui, J.; Misra, S.; Firth, A.E.; Snijder, E.J.; Fang, Y. A swine arterivirus deubiquitinase stabilizes two major envelope proteins and promotes production of viral progeny. PLoS Pathog. 2021, 17, e1009403. [Google Scholar] [CrossRef] [PubMed]
  87. Xiao, Y.; Wu, W.; Gao, J.; Smith, N.; Burkard, C.; Xia, D.; Zhang, M.; Wang, C.; Archibald, A.; Digard, P.; et al. Characterization of the Interactome of the Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 2 Reveals the Hyper Variable Region as a Binding Platform for Association with 14-3-3 Proteins. J. Proteome Res. 2016, 15, 1388–1401. [Google Scholar] [CrossRef]
  88. Hu, Y.; Ke, P.; Gao, P.; Zhang, Y.; Zhou, L.; Ge, X.; Guo, X.; Han, J.; Yang, H. Identification of an Intramolecular Switch That Controls the Interaction of Helicase nsp10 with Membrane-Associated nsp12 of Porcine Reproductive and Respiratory Syndrome Virus. J. Virol. 2021, 95, e0051821. [Google Scholar] [CrossRef]
  89. Posthuma, C.C.; Pedersen, K.W.; Lu, Z.; Joosten, R.G.; Roos, N.; Zevenhoven-Dobbe, J.C.; Snijder, E.J. Formation of the arterivirus replication/transcription complex: A key role for nonstructural protein 3 in the remodeling of intracellular membranes. J. Virol. 2008, 82, 4480–4491. [Google Scholar] [CrossRef]
  90. Snijder, E.J.; van Tol, H.; Roos, N.; Pedersen, K.W. Non-structural proteins 2 and 3 interact to modify host cell membranes during the formation of the arterivirus replication complex. J. Gen. Virol. 2001, 82 Pt 5, 985–994. [Google Scholar] [CrossRef]
  91. Nan, H.; Lan, J.; Tian, M.; Dong, S.; Tian, J.; Liu, L.; Xu, X.; Chen, H. The Network of Interactions Among Porcine Reproductive and Respiratory Syndrome Virus Non-structural Proteins. Front. Microbiol. 2018, 9, 970. [Google Scholar] [CrossRef] [PubMed]
  92. Lian, H.; Zang, R.; Wei, J.; Ye, W.; Hu, M.M.; Chen, Y.D.; Zhang, X.N.; Guo, Y.; Lei, C.Q.; Yang, Q.; et al. The Zinc-Finger Protein ZCCHC3 Binds RNA and Facilitates Viral RNA Sensing and Activation of the RIG-I-like Receptors. Immunity 2018, 49, 438–448.e435. [Google Scholar] [CrossRef] [PubMed]
  93. Sun, Z.; Chen, Z.; Lawson, S.R.; Fang, Y. The cysteine protease domain of porcine reproductive and respiratory syndrome virus nonstructural protein 2 possesses deubiquitinating and interferon antagonism functions. J. Virol. 2010, 84, 7832–7846. [Google Scholar] [CrossRef]
  94. Liu, H.; Li, C.; He, W.; Chen, J.; Yang, G.; Chen, L.; Chang, H. Free ISG15 inhibits Pseudorabies virus infection by positively regulating type I IFN signaling. PLoS Pathog. 2022, 18, e1010921. [Google Scholar] [CrossRef] [PubMed]
  95. Frias-Staheli, N.; Giannakopoulos, N.V.; Kikkert, M.; Taylor, S.L.; Bridgen, A.; Paragas, J.; Richt, J.A.; Rowland, R.R.; Schmaljohn, C.S.; Lenschow, D.J.; et al. Ovarian tumor domain-containing viral proteases evade ubiquitin- and ISG15-dependent innate immune responses. Cell Host Microbe 2007, 2, 404–416. [Google Scholar] [CrossRef] [PubMed]
  96. Jin, J.; Meng, X.; Huo, Y.; Deng, H. Induced TRIM21 ISGylation by IFN-β enhances p62 ubiquitination to prevent its autophagosome targeting. Cell Death Dis. 2021, 12, 697. [Google Scholar] [CrossRef] [PubMed]
  97. Yi, H.; Wang, Q.; Lu, L.; Ye, R.; Xie, E.; Yu, Z.; Sun, Y.; Chen, Y.; Cai, M.; Qiu, Y.; et al. PSMB4 Degrades the Porcine Reproductive and Respiratory Syndrome Virus Nsp1α Protein via the Autolysosome Pathway and Induces the Production of Type I Interferon. J. Virol. 2023, 97, e0026423. [Google Scholar] [CrossRef]
  98. Oleksiewicz, M.B.; Bøtner, A.; Toft, P.; Normann, P.; Storgaard, T. Epitope mapping porcine reproductive and respiratory syndrome virus by phage display: The nsp2 fragment of the replicase polyprotein contains a cluster of B-cell epitopes. J. Virol. 2001, 75, 3277–3290. [Google Scholar] [CrossRef]
  99. Brown, E.; Lawson, S.; Welbon, C.; Gnanandarajah, J.; Li, J.; Murtaugh, M.P.; Nelson, E.A.; Molina, R.M.; Zimmerman, J.J.; Rowland, R.R.; et al. Antibody response to porcine reproductive and respiratory syndrome virus (PRRSV) nonstructural proteins and implications for diagnostic detection and differentiation of PRRSV types I and II. Clin. Vaccine Immunol. 2009, 16, 628–635. [Google Scholar] [CrossRef]
  100. De Lima, M.; Pattnaik, A.K.; Flores, E.F.; Osorio, F.A. Serologic marker candidates identified among B-cell linear epitopes of Nsp2 and structural proteins of a North American strain of porcine reproductive and respiratory syndrome virus. Virology 2006, 353, 410–421. [Google Scholar] [CrossRef]
  101. Johnson, C.R.; Yu, W.; Murtaugh, M.P. Cross-reactive antibody responses to nsp1 and nsp2 of Porcine reproductive and respiratory syndrome virus. J. Gen. Virol. 2007, 88 Pt 4, 1184–1195. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, F.X.; Song, N.; Chen, L.Z.; Cheng, S.P.; Wu, H.; Wen, Y.J. Non-structural protein 2 of the porcine reproductive and respiratory syndrome (PRRS) virus: A crucial protein in viral pathogenesis, immunity and diagnosis. Res. Vet. Sci. 2013, 95, 1–7. [Google Scholar] [CrossRef] [PubMed]
  103. Yan, Y.; Guo, X.; Ge, X.; Chen, Y.; Cha, Z.; Yang, H. Monoclonal antibody and porcine antisera recognized B-cell epitopes of Nsp2 protein of a Chinese strain of porcine reproductive and respiratory syndrome virus. Virus Res. 2007, 126, 207–215. [Google Scholar] [CrossRef] [PubMed]
  104. Kappes, M.A.; Miller, C.L.; Faaberg, K.S. Highly divergent strains of porcine reproductive and respiratory syndrome virus incorporate multiple isoforms of nonstructural protein 2 into virions. J. Virol. 2013, 87, 13456–13465. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, H.; Xu, Y.; Feng, W. Porcine Reproductive and Respiratory Syndrome Virus: Immune Escape and Application of Reverse Genetics in Attenuated Live Vaccine Development. Vaccines 2021, 9, 480. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, H.M.; Liu, Y.G.; Tang, Y.D.; Liu, T.X.; Zheng, L.L.; Wang, T.Y.; Liu, S.G.; Wang, G.; Cai, X.H. A natural recombinant PRRSV between HP-PRRSV JXA1-like and NADC30-like strains. Transbound. Emerg. Dis. 2018, 65, 1078–1086. [Google Scholar] [CrossRef]
  107. Lu, W.; Sun, B.; Mo, J.; Zeng, X.; Zhang, G.; Wang, L.; Zhou, Q.; Zhu, L.; Li, Z.; Xie, Q.; et al. Attenuation and immunogenicity of a live high pathogenic PRRSV vaccine candidate with a 32-amino acid deletion in the nsp2 protein. J. Immunol. Res. 2014, 2014, 810523. [Google Scholar] [CrossRef] [PubMed]
  108. Xu, Y.Z.; Zhou, Y.J.; Zhang, S.R.; Jiang, Y.F.; Tong, W.; Yu, H.; Tong, G.Z. Stable expression of foreign gene in nonessential region of nonstructural protein 2 (nsp2) of porcine reproductive and respiratory syndrome virus: Applications for marker vaccine design. Vet. Microbiol. 2012, 159, 1–10. [Google Scholar] [CrossRef]
  109. Du, L.; Wang, H.; Liu, F.; Wei, Z.; Weng, C.; Tang, J.; Feng, W.H. NSP2 Is Important for Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus to Trigger High Fever-Related COX-2-PGE2 Pathway in Pigs. Front. Immunol. 2021, 12, 657071. [Google Scholar] [CrossRef]
  110. Kim, D.Y.; Kaiser, T.J.; Horlen, K.; Keith, M.L.; Taylor, L.P.; Jolie, R.; Calvert, J.G.; Rowland, R.R. Insertion and deletion in a non-essential region of the nonstructural protein 2 (nsp2) of porcine reproductive and respiratory syndrome (PRRS) virus: Effects on virulence and immunogenicity. Virus Genes. 2009, 38, 118–128. [Google Scholar] [CrossRef]
  111. Li, D.Y.; Cui, X.Y.; Huang, X.Y.; Hu, Y.; Tian, X.X.; Wang, T.; Yang, Y.B.; Wang, Q.; Tian, Z.J.; Cai, X.H.; et al. Characterization of Two Immunodominant Antigenic Peptides in NSP2 of PRRSV-2 and Generation of a Marker PRRSV Strain Based on the Peptides. Front. Vet. Sci. 2022, 9, 902822. [Google Scholar] [CrossRef] [PubMed]
  112. Kong, C.; Li, D.; Hu, Y.; Gao, P.; Zhang, Y.; Zhou, L.; Ge, X.; Guo, X.; Han, J.; Yang, H. The Genetic Variation of Porcine Reproductive and Respiratory Syndrome Virus Replicase Protein nsp2 Modulates Viral Virulence and Persistence. J. Virol. 2023, 97, e0168922. [Google Scholar] [CrossRef] [PubMed]
  113. Klionsky, D.J.; Emr, S.D. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290, 1717–1721. [Google Scholar] [CrossRef] [PubMed]
  114. Hui, X.; Zhang, L.; Cao, L.; Huang, K.; Zhao, Y.; Zhang, Y.; Chen, X.; Lin, X.; Chen, M.; Jin, M. SARS-CoV-2 promote autophagy to suppress type I interferon response. Signal Transduct. Target. Ther. 2021, 6, 180. [Google Scholar] [CrossRef] [PubMed]
  115. Tan, X.; Cai, K.; Li, J.; Yuan, Z.; Chen, R.; Xiao, H.; Xu, C.; Hu, B.; Qin, Y.; Ding, B. Coronavirus subverts ER-phagy by hijacking FAM134B and ATL3 into p62 condensates to facilitate viral replication. Cell Rep. 2023, 42, 112286. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, R.; Zhu, Y.; Zhao, J.; Ren, C.; Li, P.; Chen, H.; Jin, M.; Zhou, H. Autophagy Promotes Replication of Influenza A Virus In Vitro. J. Virol. 2019, 93, e01984-18. [Google Scholar] [CrossRef]
  117. Diao, F.; Jiang, C.; Sun, Y.; Gao, Y.; Bai, J.; Nauwynck, H.; Wang, X.; Yang, Y.; Jiang, P.; Liu, X. Porcine reproductive and respiratory syndrome virus infection triggers autophagy via ER stress-induced calcium signaling to facilitate virus replication. PLoS Pathog. 2023, 19, e1011295. [Google Scholar] [CrossRef]
  118. Sun, M.X.; Huang, L.; Wang, R.; Yu, Y.L.; Li, C.; Li, P.P.; Hu, X.C.; Hao, H.P.; Ishag, H.A.; Mao, X. Porcine reproductive and respiratory syndrome virus induces autophagy to promote virus replication. Autophagy 2012, 8, 1434–1447. [Google Scholar] [CrossRef]
  119. Nwadike, C.; Williamson, L.E.; Gallagher, L.E.; Guan, J.L.; Chan, E.Y.W. AMPK Inhibits ULK1-Dependent Autophagosome Formation and Lysosomal Acidification via Distinct Mechanisms. Mol. Cell Biol. 2018, 38, e00023-18. [Google Scholar] [CrossRef]
  120. Ferreira-Marques, M.; Carvalho, A.; Cavadas, C.; Aveleira, C.A. PI3K/AKT/MTOR and ERK1/2-MAPK signaling pathways are involved in autophagy stimulation induced by caloric restriction or caloric restriction mimetics in cortical neurons. Aging 2021, 13, 7872–7882. [Google Scholar] [CrossRef]
  121. Mendoza, M.C.; Er, E.E.; Blenis, J. The Ras-ERK and PI3K-mTOR pathways: Cross-talk and compensation. Trends Biochem. Sci. 2011, 36, 320–328. [Google Scholar] [CrossRef] [PubMed]
  122. Lunz, V.; Romanin, C.; Frischauf, I. STIM1 activation of Orai1. Cell Calcium 2019, 77, 29–38. [Google Scholar] [CrossRef] [PubMed]
  123. Decuypere, J.P.; Kindt, D.; Luyten, T.; Welkenhuyzen, K.; Missiaen, L.; De Smedt, H.; Bultynck, G.; Parys, J.B. mTOR-Controlled Autophagy Requires Intracellular Ca2+ Signaling. PLoS ONE 2013, 8, e61020. [Google Scholar] [CrossRef] [PubMed]
  124. Amemiya, Y.; Maki, M.; Shibata, H.; Takahara, T. New Insights into the Regulation of mTOR Signaling via Ca2+-Binding Proteins. Int. J. Mol. Sci. 2023, 24, 3923. [Google Scholar] [CrossRef] [PubMed]
  125. Poggio, E.; Vallese, F.; Hartel, A.J.W.; Morgenstern, T.J.; Kanner, S.A.; Rauh, O.; Giamogante, F.; Barazzuol, L.; Shepard, K.L.; Colecraft, H.M.; et al. Perturbation of the host cell Ca2+ homeostasis and ER-mitochondria contact sites by the SARS-CoV-2 structural proteins E and M. Cell Death Dis. 2023, 14, 297. [Google Scholar] [CrossRef] [PubMed]
  126. Chen, X.; Cao, R.; Zhong, W. Host Calcium Channels and Pumps in Viral Infections. Cells 2019, 9, 94. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, Q.; Men, Y.; Wang, D.; Xu, D.; Liu, S.; Xiao, S.; Fang, L. Porcine reproductive and respiratory syndrome virus infection induces endoplasmic reticulum stress, facilitates virus replication, and contributes to autophagy and apoptosis. Sci. Rep. 2020, 10, 13131. [Google Scholar] [CrossRef]
  128. Wang, L.; Zhou, L.; Zhang, H.; Li, Y.; Ge, X.; Guo, X.; Yu, K.; Yang, H. Interactome profile of the host cellular proteins and the nonstructural protein 2 of porcine reproductive and respiratory syndrome virus. PLoS ONE 2014, 9, e99176. [Google Scholar] [CrossRef]
  129. Ong, D.S.; Mu, T.W.; Palmer, A.E.; Kelly, J.W. Endoplasmic reticulum Ca2+ increases enhance mutant glucocerebrosidase proteostasis. Nat. Chem. Biol. 2010, 6, 424–432. [Google Scholar] [CrossRef]
  130. Liu, Y.; Ma, Y.; Xu, J.; Zhang, G.; Zhao, X.; He, Z.; Wang, L.; Yin, N.; Peng, M. VMP1 prevents Ca2+ overload in endoplasmic reticulum and maintains naive T cell survival. J. Exp. Med. 2023, 220, e20221068. [Google Scholar] [CrossRef]
  131. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef]
  132. Gómez-Laguna, J.; Salguero, F.J.; Fernández de Marco, M.; Barranco, I.; Rodríguez-Gómez, I.M.; Quezada, M.; Carrasco, L. Type 2 Porcine Reproductive and Respiratory Syndrome Virus infection mediated apoptosis in B- and T-cell areas in lymphoid organs of experimentally infected pigs. Transbound. Emerg. Dis. 2013, 60, 273–278. [Google Scholar] [CrossRef] [PubMed]
  133. Karniychuk, U.U.; Saha, D.; Geldhof, M.; Vanhee, M.; Cornillie, P.; Van den Broeck, W.; Nauwynck, H.J. Porcine reproductive and respiratory syndrome virus (PRRSV) causes apoptosis during its replication in fetal implantation sites. Microb. Pathog. 2011, 51, 194–202. [Google Scholar] [CrossRef] [PubMed]
  134. Kim, T.S.; Benfield, D.A.; Rowland, R.R. Porcine reproductive and respiratory syndrome virus-induced cell death exhibits features consistent with a nontypical form of apoptosis. Virus Res. 2002, 85, 133–140. [Google Scholar] [CrossRef] [PubMed]
  135. Sur, J.H.; Doster, A.R.; Christian, J.S.; Galeota, J.A.; Wills, R.W.; Zimmerman, J.J.; Osorio, F.A. Porcine reproductive and respiratory syndrome virus replicates in testicular germ cells, alters spermatogenesis, and induces germ cell death by apoptosis. J. Virol. 1997, 71, 9170–9179. [Google Scholar] [CrossRef]
  136. Sur, J.H.; Doster, A.R.; Osorio, F.A. Apoptosis induced in vivo during acute infection by porcine reproductive and respiratory syndrome virus. Vet. Pathol. 1998, 35, 506–514. [Google Scholar] [CrossRef] [PubMed]
  137. Ma, Z.; Wang, Y.; Zhao, H.; Xu, A.T.; Wang, Y.; Tang, J.; Feng, W.H. Porcine reproductive and respiratory syndrome virus nonstructural protein 4 induces apoptosis dependent on its 3C-like serine protease activity. PLoS ONE 2013, 8, e69387. [Google Scholar] [CrossRef]
  138. Costers, S.; Lefebvre, D.J.; Delputte, P.L.; Nauwynck, H.J. Porcine reproductive and respiratory syndrome virus modulates apoptosis during replication in alveolar macrophages. Arch. Virol. 2008, 153, 1453–1465. [Google Scholar] [CrossRef]
  139. Dikic, I. CIN85/CMS family of adaptor molecules. FEBS Lett. 2002, 529, 110–115. [Google Scholar] [CrossRef]
  140. Yu, L.; Hébert, M.C.; Zhang, Y.E. TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. EMBO J. 2002, 21, 3749–3759. [Google Scholar] [CrossRef]
  141. Zhang, H.; Wang, X. A dual effect of porcine reproductive and respiratory syndrome virus replication on the phosphatidylinositol-3-kinase-dependent Akt pathway. Arch. Virol. 2010, 155, 571–575. [Google Scholar] [CrossRef]
  142. Xie, W.; Chen, C.; Han, Z.; Huang, J.; Liu, X.; Chen, H.; Zhang, T.; Chen, S.; Chen, C.; Lu, M.; et al. CD2AP inhibits metastasis in gastric cancer by promoting cellular adhesion and cytoskeleton assembly. Mol. Carcinog. 2020, 59, 339–352. [Google Scholar] [CrossRef]
  143. Srivatsan, S.; Swiecki, M.; Otero, K.; Cella, M.; Shaw, A.S. CD2-associated protein regulates plasmacytoid dendritic cell migration, but is dispensable for their development and cytokine production. J. Immunol. 2013, 191, 5933–5940. [Google Scholar] [CrossRef]
Figure 1. Map of the PRRSV genome. (A) The PRRSV genome is composed of ORF1a, ORF1b, ORF2a, ORF2b, ORF3–7 and ORF5a and produces at least 16 nonstructural and 8 structural proteins. NSP2 is cleaved from ORF1 translation products to form mature proteins and it is composed of HV-I, PLP2, HV-2, TM and C-terminal domains. (B) NSP2 undergoes a −1/2 ribosomal frameshift and generates NSP2N and NSP2TF variants in response to both pcbp2 and NSP1β.
Figure 1. Map of the PRRSV genome. (A) The PRRSV genome is composed of ORF1a, ORF1b, ORF2a, ORF2b, ORF3–7 and ORF5a and produces at least 16 nonstructural and 8 structural proteins. NSP2 is cleaved from ORF1 translation products to form mature proteins and it is composed of HV-I, PLP2, HV-2, TM and C-terminal domains. (B) NSP2 undergoes a −1/2 ribosomal frameshift and generates NSP2N and NSP2TF variants in response to both pcbp2 and NSP1β.
Viruses 15 02310 g001
Figure 2. NSP2 regulates innate immunity. NSP2 antagonizes the innate immunity of host cells by deubiquitinating RIG-1 and IKBα or reducing the protein level of ISG15 or ISG15. NSP2 antagonizes host innate immunity by deubiquitinating RIG-1 and IKBα or separating ISG15 from its substrate and reducing ISG15 protein levels.
Figure 2. NSP2 regulates innate immunity. NSP2 antagonizes the innate immunity of host cells by deubiquitinating RIG-1 and IKBα or reducing the protein level of ISG15 or ISG15. NSP2 antagonizes host innate immunity by deubiquitinating RIG-1 and IKBα or separating ISG15 from its substrate and reducing ISG15 protein levels.
Viruses 15 02310 g002
Figure 3. NSP2 regulates cell autophagy and apoptosis. NSP2 regulates endoplasmic reticulum stress and autophagy by binding to endoplasmic reticulum stress protein GRP78 and Ca2+ sensor protein STIM1. NSP2 binds to anti-apoptotic protein Bcl-2 and reduces its expression to induce apoptosis.
Figure 3. NSP2 regulates cell autophagy and apoptosis. NSP2 regulates endoplasmic reticulum stress and autophagy by binding to endoplasmic reticulum stress protein GRP78 and Ca2+ sensor protein STIM1. NSP2 binds to anti-apoptotic protein Bcl-2 and reduces its expression to induce apoptosis.
Viruses 15 02310 g003
Figure 4. NSP2 of PRRSV regulates host cell and viral replication. NSP2 regulates cytoskeleton, cell cycle, autophagy, apoptosis, cellular innate immunity, viral protein maturation and viral assembly by reducing the expression of key proteins or modifying key proteins by means of deubiquitination.
Figure 4. NSP2 of PRRSV regulates host cell and viral replication. NSP2 regulates cytoskeleton, cell cycle, autophagy, apoptosis, cellular innate immunity, viral protein maturation and viral assembly by reducing the expression of key proteins or modifying key proteins by means of deubiquitination.
Viruses 15 02310 g004
Table 1. Important epidemic PRRSV strain information.
Table 1. Important epidemic PRRSV strain information.
YearsCountryStrainStrain CharacteristicsReferences
1987AmericaATCC VR2332PRRSV-2 prototype strain[14]
1990EuropeLVPRRSV-1 prototype strain[15]
1992JapaneseGU992MFirst reported in Asia[16,17]
1990sAmericaVR2385A highly virulent PRRSV-2 strain identified in the mid-1990s[18]
1995ChinaCH-1aFirst reported in China, the PRRSV pathogen was isolated by Guo et al. (1996) and Yang et al. (1997)[4]
1998AmericaAtypical PRRSVCauses high fetal mortality and abortions in pig herds[19]
2001ChinaMN184The highly virulent MN184 strain has very obvious nucleotide differences (>14.5%) from other genotype 2 strains[20]
2006ChinaJXA130-amino-acid deletion in NSP2 and highly pathogenic PRRSV (HP-PRRSV)[5]
2015ChinaNADC30-likeThe current main circulating strains in China[6,9]
2018ChinaNADC34-likeCould become a potential pandemic strain in China[7,8]
Table 2. Functions of each protein of PRRSV.
Table 2. Functions of each protein of PRRSV.
ORFProteinFunctionReferences
ORF1aNSP1 (α)Loss of functional NSP1αpapain-like proteinase activity is associated with the inhibition of sg mRNA synthesis.[24]
NSP1 (β)Loss of papain-like proteinase activity of NSP1β results in the NSP1β/NSP2 adapter not being processed in vitro.[25,26]
NSP2a/NSP2b/NSP2c/NSP2d/NSP2e//NSP2f/NSP2TFNSP2 is a cofactor of nsp4 serine protease and participates in the assembly of polyproteins during virus replication.[27]
NSP3NSP3 remodels the cell membrane.[28,29]
NSP4NSP4 is a potential IFN antagonist that induces apoptosis.[30,31,32,33,34]
NSP5NSP5 is a transmembrane protein involved in cell membrane modification.[35]
nsp6It is involved in the virulence of the virus.[36]
NSP7αIt is one of the most conserved proteins in PRRSV, NSP7 and NSP7α/NSP7β and is critical for virus viability and recovery.[37]
NSP7β
NSP8It is involved in the virulence of the virus.[38]
ORF1abNSP9It is a key protein for the high virulence of HP-PRRSV and the RdRp of PRRSV, which plays a key role in the virus replication process.[39,40,41,42,43]
NSP10It is the helicase of PRRSV and is the key to unraveling dsRNA and RNA synthesis during PRRSV replication and ultimately completing virus replication.[44]
NSP11It is the B cell linear epitope, has RNA endoribonuclease activity and deubiquitination activity.[45,46,47]
NSP12It is necessary for the synthesis of viral subgenomic SG mRNA and promotes viral replication.[48,49,50]
ORF2aGP2aSpecial glycosylated proteins capable of inducing specific neutralizing antibodies against PRRSV.[51]
ORF2bEIt is a pore-forming protein that mediates the formation of ion channels for viral particle uncoating.[52,53,54]
ORF3GP3The main component of the viral envelope surface polymer promotes the early infection of highly pathogenic PRRSV in epithelial cells and can induce the production of neutralizing antibodies against PRRSV.[55,56]
ORF4GP4The key adhesion protein of the PRRSV virus, the GP4 protein of PRRSV-1, has a neutralizing antibody-inducing epitope, which can induce the body to produce homologous strain-specific strong neutralizing antibodies.[55,57,58]
ORF5GP5As the most variable transmembrane glycosylated protein among structural proteins, it has linear and conformational neutralizing antibody epitopes and is the most important protective anti-neutralizing antibody-inducing protein.[59]
ORF6MIt is the most conserved structural protein and contains a large number of antigenic epitopes. It has strong immunogenicity and can form heterodimers with GP5.[60,61,62,63,64,65]
ORF7NViral capsid protein promotes PRRSV RNA synthesis, contributes to PRRSV replication and increases PRRSV virulence.[66,67,68]
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

Liu, B.; Luo, L.; Shi, Z.; Ju, H.; Yu, L.; Li, G.; Cui, J. Research Progress of Porcine Reproductive and Respiratory Syndrome Virus NSP2 Protein. Viruses 2023, 15, 2310. https://doi.org/10.3390/v15122310

AMA Style

Liu B, Luo L, Shi Z, Ju H, Yu L, Li G, Cui J. Research Progress of Porcine Reproductive and Respiratory Syndrome Virus NSP2 Protein. Viruses. 2023; 15(12):2310. https://doi.org/10.3390/v15122310

Chicago/Turabian Style

Liu, Benjin, Lingzhi Luo, Ziqi Shi, Houbin Ju, Lingxue Yu, Guoxin Li, and Jin Cui. 2023. "Research Progress of Porcine Reproductive and Respiratory Syndrome Virus NSP2 Protein" Viruses 15, no. 12: 2310. https://doi.org/10.3390/v15122310

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