Skip to main content

Membrane fusion, potential threats, and natural antiviral drugs of pseudorabies virus

Abstract

Pseudorabies virus (PrV) can infect several animals and causes severe economic losses in the swine industry. Recently, human encephalitis or endophthalmitis caused by PrV infection has been frequently reported in China. Thus, PrV can infect animals and is becoming a potential threat to human health. Although vaccines and drugs are the main strategies to prevent and treat PrV outbreaks, there is no specific drug, and the emergence of new PrV variants has reduced the effectiveness of classical vaccines. Therefore, it is challenging to eradicate PrV. In the present review, the membrane fusion process of PrV entering target cells, which is conducive to revealing new therapeutic and vaccine strategies for PrV, is presented and discussed. The current and potential PrV pathways of infection in humans are analyzed, and it is hypothesized that PrV may become a zoonotic agent. The efficacy of chemically synthesized drugs for treating PrV infections in animals and humans is unsatisfactory. In contrast, multiple extracts of traditional Chinese medicine (TCM) have shown anti-PRV activity, exerting its effects in different phases of the PrV life-cycle and suggesting that TCM compounds may have great potential against PrV. Overall, this review provides insights into developing effective anti-PrV drugs and emphasizes that human PrV infection should receive more attention.

1 Introduction

Pseudorabies virus (PrV) is a linear double-stranded DNA virus belonging to the Varicellovirus genus in the Alphaherpesvirinae subfamily of Herpesvirales. Other representative members of Alphaherpesvirinae are the varicella-zoster virus (VZV), Varicellovirus Bovine alphaherpesvirus 1 (BoHV-1), Herpes simplex virus 1 and 2 (HSV-1 and HSV-2, respectively), Marek’s disease virus (MDV), and infectious laryngotracheitis virus (ILTV). However, HSV-1 and HSV-2 belong to the Simplexvirus genus and MDV and ILTV to Mardivirus and Iltovirus, respectively [1]. The PrV genome is approximately 140 kb, with a GC content averaging 74%, and is predicted to encode more than 70 different proteins. The viral genome structure includes long- and short unique regions and internal- and terminal repeats [2, 3]. Genes on the PrV genome encode structural and virulence proteins, enzymes, and proteins related to viral invasion, replication, and release [4].

As the causative agent of Aujeszky’s disease (AD) or pseudorabies (PR), PrV has a broad host range, including domestic animals [5], wild animals [6, 7], and experimental animals such as rabbits and mice [8]. Although PrV infection is rare in humans, it is a potential threat to public health. PrV causes acute encephalitis, endophthalmitis, and retinal vasculitis in humans [9,10,11]. Pigs are the natural PrV hosts, and this viral infection is primarily reflected in respiratory symptoms in older pigs, fetus death, abortion in pregnant sows, or both. In young piglets and more susceptible species, PrV infection is often fatal as it leads to central nervous system disorders [12, 13]. PrV can also establish a lifelong latent infection in the peripheral nervous system. In pigs stimulated by external stimuli or with weakened immunity, latent PrV was reactivated from peripheral neurons, replicated, and produced new progeny viruses that led to the re-emergence of pseudorabies [14]. This ability of PrV brings challenges to eradicating pseudorabies.

This disease causes considerable economic losses in the swine industry every year [15], and vaccination using classical attenuated live PrV or inactivated PrV vaccines has been one of the most important methods to prevent and control pseudorabies [13]. Many European countries, New Zealand, and the United States rely on strict national eradication programs based on mandatory vaccination, culling positive pigs, and establishing PrV-free pig herds. AD has been eradicated from domestic pig populations [16], but PrV is still found in wild boars and other wild animals [17, 18]. Thus, PrV is still a risk for outdoor pig farms in these countries. In China, measures are being taken to eradicate AD. The traditional Bartha-K61 vaccine is the most widely used in China. Before 2011, this vaccine was able to control pseudorabies outbreaks, but since late 2011, Bartha-K61 has been unable to provide complete protection against PrV strains [19]. The emergence of mutant strains is undoubtedly a considerable obstacle to preventing PrV. Thus, developing PrV vaccines based on the epidemic strains is crucial, which can prevent or eradicate pseudorabies. Furthermore, novel anti-PrV drugs are required to prevent PrV-induced latent infection and decrease the virus pathogenicity. In addition to vaccines and drugs, one potential strategy to prevent pseudorabies is to develop antibodies or blockers targeting the biomolecules involved in PrV infection, such as receptors and ligands.

2 PrV invasion

The mature PrV virion comprises four structural elements: the linear DNA genome, a protective icosahedral capsid, the tegument, and the envelope. The envelope is derived from the membrane structure of the host cell infused with virus-encoded proteins and participates in virus entry, egress, cell-to-cell spread, and immune response. These proteins include gB, gC, gD, and gE and more than 11 proteins are modified by N- or O-linked sugars and non-glycosylated proteins UL20, UL43, and US9 [4, 20]. The tegument, located between the capsid and the envelope, contains outer and inner tegument proteins [21, 22]. The outer tegument protein preferentially associates with viral membranes, whereas the inner tegument protein adjacent to the capsid forms a capsid-tegument complex after virus entry into the cell. This capsid-tegument is closely related to PrV transport along microtubules into the nucleus. Compared with other enveloped viruses, PrV binding and access into target cells require the concerted actions of at least five glycoproteins (gC, gD, gL, gH, and gB). A detailed understanding of PrV-cellular membrane fusion at the molecular level is essential for developing new therapeutic strategies, especially receptor and ligand-targeting drugs to block virus invasion and transmission. The invasion mechanism of HSV alphaherpesviruses has been extensively described and often used as baseline research on other alphaherpesviruses. However, in addition to the conserved characteristics, alphaherpesviruses have unique features. Thus, in the following section, we explore the invasion mode of PrV.

2.1 PrV adhesion and gD

Virus entry into cells results from the coordinated interaction between viral proteins and cellular receptors. The initial PrV attachment to the cell is weak and does not trigger the virus entry. At this stage, PrV gC binds to the heparan sulfate (HS) receptor on the cell membrane [23], and then gD binds firmly to the cell receptor nectins (mainlly nectin-1) [24]. For most alphaherpesviruses, gD is an essential, non-conserved receptor binding protein that affects the tropism of the alphaherpesviruses to cells, although VZV does not encode gD [25]. The PrV gD structure includes an immunoglobulin variable (IgV)-like core wrapped by N- and C-terminal extensions [26]. The gD homologs of HSV, BoHV-1, and PrV share only 22–33% amino acid identity, indicating that they have different entry receptor and binding affinities. Entry receptors, nectin-1/2, herpesvirus entry mediator (HVEM), HS 3-O-sulfonated derivatives for HSV, nectin-1/2 for PrV, and nectin-1 for BoHV-1, have been identified [27]. Human nectin-1 shares 96% amino acid identity with porcine nectin-1. Although PrV gD exhibits similar affinity for both human and porcine nectin-1 [26, 28, 29], the PrV gD binding affinity for human nectin-1 is ten-fold higher than that of HSV-1 gD [26, 27], which may explain human PrV infection. Despite the low amino acid homology of PrV and HSV gDs, most of their secondary structure elements are similar. However, some features such as the PrV gD C-terminal extension contains an extra α2’ helix, the steric position of the α1’ helix in PrV, and HSV gD structures are different. Furthermore, compared with the HSV gD N-terminal loop (N-loop), the shorter PrV gD N-loop cannot form the hairpin structure necessary for HVEM binding. This inability to form the hairpin structure is consistent with the fact that PrV cannot utilize HVEM as a cellular receptor [26].

The PrV gD/swine nectin-1 complex structure has been recently identified [26]. First, PrV gD recognizes and engages the swine nectin-1 CC’C’’FG sheet through str2 and its flanking loops in the N-terminal extension, the α1’ helix in the IgV-core, and helices α2’, α3’, α3, and the α3’/α3 loop in its C-terminal extension. The N-loop is then reoriented upon binding as two aromatic residues in this structure, F11 and W22, are projected outwards, forming a hydrophobic interaction with nectin-1 amino acids K61, Q64, I80, N82, M85-S88, and F129. F11 undergoes further conformational changes after receptor binding. The major forces for stabilizing the N-loop orientation in the complex structure may result from the interactions of F11 with nectin-1 K61 and F129. Before binding to the receptor, gD monomers form dimers through disulfide bonds, likely stabilizing the C-terminus conformation [30]. The membrane-proximal loop of the gD ectodomain (proposed as the pro-fusion domain, PFD) and the conserved Pro291/Trp294 in the gD C-terminus are vital in gD-mediated alphaherpesvirus entry [30,31,32]. The PFD length affects the PrV gD affinity for nectin-1, with a short PFD leading to higher affinity [26]. The HSV PFD pre-locates in a position that blocks the binding of gD with its receptor. However, the controlled PFD displacement exposes the locked receptor-binding site allowing gD to recognize and bind to its receptor [30]. Nectin-1 homologues and heterodimer formation are the basis for nectin-1 adhesion [33]. Because the affinity of PrV gD and human nectin-1 is higher than the nectin-1 self-affinity [26, 34], PrV gD might destroy nectin-1 dimer formation by competing with nectin-1 and changing receptor presentation on target cells. Furthermore, gD may have evolved from an essential to a non-essential infection protein. While HSV-1 requires gD for cell entry and direct cell-to-cell spread [35], PrV gD is associated with cell penetration but is dispensable in direct cell-to-cell spread [36]. PrV gD is reportedly not a core fusion protein; gD-negative PrV mutants (PrV-gD Pass) remained infective with compensatory mutations in gB and gH, both in vivo and in vitro [36, 37]. Nevertheless, the host range of PrV-gD Pass changed as this mutant virus was unable to infect pigs but was fatal for mice [37].

2.2 The gH/L complex–a virus and cellular membrane fusion regulator

The heterodimeric complex formed by the integral membrane gH plus the anchorless gL and the homotrimeric gB jointly participate in the core PrV envelope fusion mechanism with the cellular cytoplasmic membrane. Moreover, without the gH/L complex, gB cannot function as a fusion protein [38]. Although gH/L acts as a regulator to transmit the fusion signal to gB [39], the fusion speed and rate are governed by gH/L, especially gH, but not gB [40]. As a type I transmembrane protein, gH has a small cytosolic tail and a large N-terminal ectodomain. Within the same virus subfamily, gH shows substantial variability in length and sequence, especially in its N-terminal. Additionally, compared with other herpesviruses, the PrV gH ectodomain is shorter and more compact [41]. According to the gH/L crystal structures of the Epstein-Barr virus (EBV, gammaherpesvirus) [42], HSV-2 [43], VZV [44], and PrV gH core fragment structure (residues 107 to 639) [41], gH/L (or gH alone) has no typical fusion protein features. Instead, they present a common folding structure and low sequence conservation, serving as a regulator in gB fusion activity [45]. The ectodomain comprises a predicted signal peptide (SP) and ectodomain parts I, II, III, and IV. The PrV gH domain I is formed by the N-domain in specific association with gL [46] and is similar in size to EBV domain I but half the size of HSV domain I [41].

In recent HSV studies, the physical gD to gH/L binding occurred within the gH/L ectodomain. Furthermore, the gD interaction sites on gH/L were the gH N-terminus and the gL C-terminus, more specifically, close to gL residue 77 [47]. gD also carried composite-independent binding sites for gH/L and gB, both partly located in the C-terminal pro-fusion domain [48]. The putative gH/L binding sites on HSV gD have been identified as independent to those used for HSV receptor binding, but no interaction was found between gB and gD or gH/L [39]. Monoclonal antibodies targeting gD-gH/L binding sites are also being developed to block the virus fusion [49]. However, more studies on PrV are needed to reveal whether its gD-gH/L binding site is similar to that of HSV. Domain II contains a β-sheet (named “fence” as it separates the domain from the rest of the molecule) and a syntaxin-like bundle (SLB) of three α-helices, which resembles functionally relevant elements in eukaryotic fusion proteins in the syntaxin family. The SLB structure and flexibility are relevant to the maturation and PrV gH membrane fusion activity [50, 51]. Domain III consists of eight consecutive α-helices (α6–α13). Conserved proline438 assists in bending at the end of α-helix 11, and when proline438 switches to serine, the formation of the cysteine404-cysteine439 disulfide bond is impaired, affecting cell-to-cell spread and fusion activity [52]. The flap and N-linked carbohydrate moiety, which covers an underlying patch of hydrophobic amino acids in the membrane-proximal domain IV, have attracted much research. A modest conformational change in moving the flap might be required to expose the hydrophobic patch for membrane interaction during the fusion [53]. Interestingly, the IV domains of PrV and HSV-1 gH exhibit functional conservation [46]. A chimeric gH, comprising PrV I, II, and III domains and HSV-1 IV domain showed considerable fusion activity in the presence of PrV gB, gD, and gL; Along with PrV gB, gD, and HSV-1 gL, but not PrV gL, another chimeric gH (consisting of PrV II, III, and IV domains and HSV-1 I domain) retained limited functionality, and the PrV II and III domains might be functional units that cannot be replaced [46].

The gH transmembrane domain (TMD) and cytoplasmic domain (CTD) functions differ in PrV and HSV’s fusion processes. In PrV, gH CTD can decrease membrane fusion activity but is dispensable for gH function during virus infection. In contrast, gH TMD is essential for triggering fusion and highly relevant for the PrV gH function. PrV gH lacking the TMD, membrane-anchored via a lipid linker or comprising the PrV gD TMD were unfunctional [54]. However, a transient assay has shown that soluble forms of HSV gH/L were sufficient to induce gB-mediated cell-to-cell fusion [55].

The anchorless gL plays a chaperonin protein-like function and is noncovalently linked to the gH ectodomain. gH maturation and processing require the presence of gL in herpesviruses such as HSV-1 or VZV [56,57,58]. However, in PrV, gL is dispensable for correct folding, transport, or gH virion localization but necessary for infectivity. In the absence of gL (PrV-gLβ mutant), PrV gH can be detected at the cell surface, but the mutant is capable of limited cell-to-cell spread. Therefore, gL cannot solely play a chaperonin-like function in PrV [59, 60]. Furthermore, using the limited spread of the PrV-gLβ mutant, a gDH fusion polypeptide (the gD N-terminal 271 residue was fused to the gH C-terminal 590 residue) containing a receptor binding domain of PrV gD has been isolated in serial cell passaging, which can compensate for the essential gL function in the entry and PrV cell-to-cell spread [61]. Additionally, in PrV-gLPassB4.1, a mutant lacking gL replicated effectively because a combination of point mutations in gH (namely gHB4.1, mutation points: L70P and W103R in the N-terminus) and gB (namely gBB4.1, mutation points: G672R and deletion of lysine at position 883) compensated for the gL function [62]. A defective mutant incapable of cell entry and spread, PrV-gH32/98K (deleted codons 32 to 97 in the gH N-terminus) was unable to bind gL, resulting in impaired gL processing and rapid secretion of mature gL. The replication and entry competence of pPrV-gH32/98K can be restored by gBB4.1 rather than the wild-type gB, but entry is considerably delayed [60]. In summary, PrV gL and the gH gL-interacting domain are not strictly required for membrane fusion because gH and gB mutations can compensate for their absence, suggesting that gB and gH are the core proteins in PrV fusion.

2.3 gB-the fusion protein

The critical core fusion protein of PrV, gB, is the most conserved envelope glycoprotein of herpesviruses and belongs to class III fusion proteins. Unlike most class III fusion proteins, which display a short and unstructured cytoplasmic tail activated by low pH and are critical for fusion, the alphaherpesviruses’ gBs have an extensive and structured CTD (90 amino acids in PrV gB) [63, 64]. Thus, PrV gB is not an autonomous fusogen but requires activation by its partner gH/L to drive pH-independent membrane fusion [65]. Signals from gH/L trigger a metastable prefusion state gB on the viral membrane, and gB undergoes a conformational change exposing the initially buried hydrophobic regions for interactions with the target membrane, resulting in the fusion of cell and viral membranes. The prefusion conformation of PrV gB remains elusive, while the structure of the post-fusion conformation gB ectodomain has been shown to display trimeric, elongated, rod-like molecules, with each monomer comprising five domains (I to V). PrV gB ectodomain shares 50.1, 25.6, and 25.3% amino acid sequence identity with HSV-1, human cytomegalovirus (betaherpesvirus), and EBV gB, respectively [66]. The CTD, TMD, and adjoining gB membrane proximal region (MPR) play critical roles in regulating extracellular domain fusion activity. The carboxy-terminally truncated derivative of PrV gB (stably expressed in cell lines), PrV gB-008 (gB protein lacking the putative internalization signals and the C-terminal α-helical domain), showed enhanced fusogenic activity, whereas gB-007 (gB protein lacking both α-helical domains) was unable to incorporate into gB-negative PrV (PrV-gB(-)) for full infectivity. However, gB-007 restored the direct viral cell-to-cell spread of PrV-gB(-). Lacking the entire CTD, the protein lost functionality [67], indicating that CTD is closely related to gB functionality. In another study, after serial passages in cell culture, PrV-007Pass gB with a compensatory mutation in the ectodomain was obtained, and it was efficiently incorporated into the viral envelope, restoring the infection ability of PrV-gB(-) [68]. The gB CTD acts as an inhibitory clamp to restrict the gB ectodomain rearrangement stabilizing the gB prefusion structure. Without CTD, the gB ectodomain remains in a post-fusion state and cannot perform its function, affecting gH/L and gB signaling. Residue V831 within the HSV gH cytoplasmic tail reportedly wedges into the pocket above the interface between the gB CTD adjacent monomer to push the monomer apart, releasing the inhibitory clamp [69]. The above activation mechanism may also exist in PrV. A chimeric gH (domains I, II, and III of PrV gH plus the IV domain, TMD, and CTD of HSV-1 gH) could trigger PrV gB effectively [46]. HSV gH efficiency might differ from PrV gH for PrV gB activation due to its insertion into the pocket. The gB CTD plays an essential role in maintaining gB prefusion, and a regulatory element in gB domain V controlling the fusogenic gB conformational change has been identified. The PrV-gBCTD2 mutant (gB lacking 60 C-terminal amino acids) showed defective entry but was capable of cell-to-cell transmission. A PrV-gBCTD2Pass mutant restoring viral infectivity was obtained using this exceptional phenotype after serial passaging in vitro. Analysis of this variant (PrV-gBCTDPass) revealed that a single mutation in a conserved domain V residue (N735S) could compensate for the absence of CTD and support the gH/L-independent viral entry. Similar to CTD, the mutated domain can stabilize the gB ectodomain in a fusion-active conformation. Additionally, a short conserved α-helical structure in domain V (residues 728 to 742) was defined as the molecular switch for gB pre-to-post fusion. As in PrV N735, a conserved amino acid mutation in ILTV and BoHV-1 (BoHV-1 N742S and ILTV Q684S/N685S, respectively) but not in HSV-1, can also compensate for the absence of CTD and lead to gB autonomous fusogenicity, independent of the gH/L complex. This finely tuned functional tandem between fusion regulatory domains within gB may be common in alphaherpesviruses PrV, ILTV, and BoHV-1 but not in HSV-1, which may have different regulatory mechanisms [63].

The role of the regulatory TMD and MPR ectodomains has been studied in HSV and PrV. The gB TMD participates in membrane anchoring and the late fusion stage. Therefore, when HSV-1 gB TMD is replaced by a lipid anchor, the fusion can only proceed halfway [70]. The PrV gB MPR has 50 residues and acts as a shield between the gB fusion loops (FLs) and the target cell membrane before fusion occurs [66, 71]. Upon receiving the fusion signal, the gB structure undergoes rearrangement, and the appropriate movement of the MPR exposes the FLs and initiates membrane fusion. The membrane-interacting regions of the fusion protein contain abundant hydrophobic and aromatic residues for insertion into the outer leaflet of the cell membrane [72]. In PrV, the FLs (FL1 and FL2) in domain I are exposed at the base of the gB molecule. First, Phe275 at the tip of FL2 is deeply inserted into the hydrocarbon core of the phospholipid bilayer. Then, Trp187, Tyr192, and Tyr276 on the side chains of FL1 and FL2 are inserted shallowly into the membrane, forming a fusion edge that catalyzes the fusion process between PrV and the cell membrane [66]. Developing protective monoclonal antibodies (mAbs) against PrV gB, 1H1 mAb acting on the gB domain I, showed broad neutralizing activity without the complement participation, presumably by inhibiting PrV fusion to the cell membrane [73]. The other 14 mAbs identified in this study required complement participation and acted on domain IV (crown region). These results indicate multiple B-cell epitopes in the gB crown and base regions.

In the current model of alphaherpesviruses entry, PrV invades cells through three basic steps: initial adsorption, binding of the entry receptor, and membrane fusion (Figure 1). However, in the well-studied HSV, multiple receptors that can interact with gH/L or gB have also been found, such as the gH alphavbeta3-, alphavbeta6-, and alphavbeta8-integrins receptors [74, 75], the paired immunoglobulin-like type 2 receptor-α, myelin-associated glycoprotein, and non-muscle myosin heavy chains IIA and IIB of gB [76,77,78,79]. HSV-1 and BoHV-1 entry is reportedly inhibited by the soluble form of their gBs, which implies that the receptors for gB may be directly involved in cell invasion [80, 81]. Compared with HSV, there is less information on PrV gH/L and gB receptors. In addition to the membrane fusion process mediated by gB, another fusion pathway with receptor-mediated or auxiliary involvement may be involved in PrV fusion, but further studies must be done.

Figure 1
figure 1

PrV entry mechanism through membrane fusion. A PrV virion simulation diagram. PrV virions are composed of four structural elements: a lipidic envelope containing non-glycosylated trans-membrane proteins UL20, UL43, US9, and glycosylated membrane proteins gB, gC, gD, gE, gG, gH, gI, gK, gL, gM, and gN; a dense protein tegument layer, generally classified as outer and inner tegument proteins; an icosahedral protein capsid; and a linear double-stranded DNA genome. B The essential viral glycoproteins gD, gH/L, gB, and host receptor nectin-1 for the PrV entry mechanism. C The viral-cell membrane fusion process. Attachment: PrV binds with HS via its gC; this step enhances entry but is not required for fusion. Receptor binding and attachment stabilization: gD binds to its nectin-1 receptor to stabilize PrV binding; the C-terminus of the gD ectodomain exposes the receptor binding site, completing gD binding to nectin-1. This transmits a signal to activate gH/L, which may come directly from the C-terminus moving along the gD ectodomain or from other interaction sites exposed by the C-terminus moving and physical interaction between gD and gH/L. Triggering and gB rearrangement: gH/L transmits this signal via the physical binding of gH/L and gB ectodomain and the gH CTD interaction with the gB CTD to trigger core fusion protein gB. Upon triggering, the prefusion gB conformation undergoes rearrangement, emerging as a more stable post-fusion gB conformation. Fusion loop insertion: the fusion loops of post-fusion gB insert into the target cell membrane. Fusion: as gB shrinks, the fusion loops gradually drive the cell membrane closer to the gB TMD completing the viral-cell membrane fusion. Finally, the capsid associated with inner tegument proteins enter the target cell.

3 Current status of PrV infection in humans

Although pigs are the natural PrV hosts, the virus can infect other animals, including cattle, sheep, mink, mice, rabbits, cats, and dogs. PrV can reportedly infect humans, and contact hosts history of the human suspected case included cats, dogs, and cattle infected with PrV; In contrast, a previous study confirmed that humans are not susceptible to PrV or at least to the strains used in the tests [82]. At the end of 2011, a PrV outbreak occurred in pig farms immunized with the Bartha-K61 vaccine in many regions of China. So far, several PrV variants with enhanced virulence have been isolated throughout the country. Since 2017, more than 28 human cases of PrV infection have been reported in China [83,84,85], and the PrV strain hSD-1/2019 was first isolated from human cerebrospinal fluid (CSF) [9]. Whether PrV can infect humans has been controversial because previous reports were not based on virus isolation. The diagnoses of the above cases were mainly based on the PrV nucleotide detection with metagenomic next-generation sequencing (mNGS). PCR and serological tests further validated some. We selected 16 of these 28 cases, confirmed by mNGS and had relatively complete and detailed case presentations (Table 1).

Table 1 Case reports of PrV infection in humans

The patients included swine herders, veterinarians, pig butchers, and people with occupations mostly related to pig production and sales. The clinical symptoms were not restricted to fever, headache, and cough in the prodromal stage, and seizures, coma, respiratory failure, and other symptoms in the dominant period [83]. Antiviral therapy, including acyclovir, ganciclovir, and foscarnet, is essential for patients, and intravenous human immunoglobulins, glucocorticoids, anti-epileptics, and antibiotics were administered according to clinical symptoms [9, 86]. The outcome of PrV-caused encephalitis or endophthalmitis is poor, and there is no specific drug to prevent the progress of human encephalitis or endophthalmitis caused by PrV. However, most patients have residual symptoms, and the number of deaths is small [9, 87]. The contact history of patients reveals that human infection with PrV is related to occupational exposure. In addition, PrV transmission from animals to humans may occur via companion animals, and suspected cases of PrV transmission from dogs or cats to humans have been reported [82]. In addition to the growing number of people keeping companion animals, cats and dogs are at high risk of PrV infection by eating PrV-infected raw meat or preying on wild animals [88, 89].

A study detected anti-gB and anti-gE positive samples in dog serum collected at pet hospitals and anti-gB positive samples in the dog serum from pig farms. Commercial vaccines for PrV may be a hidden risk for dogs and owners [90]. 60-day-old beagle dogs infected with the commercial vaccines Bartha-K61 or HB98 strains, which are widely used in the Chinese swine industry, could shed the virus via buccal/nasopharyngeal/anal swabs, and the Bartha-K61 strain could be transmitted among dogs. Moreover, PrV has been isolated in hunting dogs in many European countries where PrV has been eradicated from domestic pigs. Genetic analysis of PrV strains showed that in addition to genotype I, frequently reported in Western Europe, there is also genotype II of Asian origin, suggesting that wild animals are potential sources of infection [91]. The sequence alignment analysis of hSD-1/2019 indicated a close phylogenetic relationship with HN1201 and HLJ8 strains and the BJ/YT strain isolated from a Yorkshire terrier dog [9]. Combining the above studies and considering the closeness between humans and dogs, more studies are needed to confirm whether there is a relation between human PrV infection and exposure to dogs.

A retrospective PrV seroepidemiological investigation was carried out on 1335 serum samples from patients with encephalitis from nine provinces in China. The enzyme-linked immunosorbent assay results showed that the PrV-positive rates were 12.16, 14.25, and 6.52% in 2012, 2013, and 2017, respectively [92]. This result suggests that PrV may be the pathogenic agent in some undefined human encephalitis. PrV and HSV belong to the Alphaherpesvirinae subfamily and share some biological characteristics and infection mechanisms. Humans are the natural host of HSV, and silent infections are extremely common in the population. Moreover, the binding affinity of PrV gD to the human nectin-1 receptor is ten times higher than that of HSV gD [26, 27]. These characteristics suggest that PrV is highly likely as a potential zoonotic pathogen.

4 Natural plant extracts as potential anti-PrV drugs

Despite the global vaccination programs, many countries still report PrV infections in domestic pigs and wild animals, and latent infections may be present. Moreover, there is no approved effective drug to treat diseases caused by PrV in swine, humans, and other animals. Acyclovir is one of the most effective treatment options against HSV-1 and VZV. However, after acyclovir treatment, human viral encephalitis caused by PrV showed limited or slight improvement [9]. Currently, PR prevention and control are mainly based on vaccines in the pig industry. Since 2011, the classic Bartha-K61 vaccine is not entirely effective against some PrV variants, which hinders the eradication of PR in China. Gene-edited vaccines based on PrV epidemic strains have been widely studied [97] and include single- [98], double- [99], triple- [100], four- [101], and five gene-deleted [102] vaccines with good development prospects. Furthermore, some of these vaccines have been licensed [103]. Compared with many studies on PrV vaccines, research on specific drugs, mAbs, and small molecule PrV inhibitors must be supplemented. PrV risks should be taken seriously, especially regarding mutant strains. Antiviral strategies are required for swine and humans to prevent and treat PrV-induced diseases and decrease the virus’ pathogenicity. The following section mainly describes the plant extracts used against PrV in traditional Chinese medicine (TCM).

4.1 Flavonoids

In the history of human civilization, TCM has been widely used in China for thousands of years. Natural drugs have less resistance and side effects. Their antiviral effects include direct action on the virus and target cells and enhanced immunity to protect the body from virus damage. Natural products have a wide structural diversity and are essential sources of new therapeutic drugs. After years of research, TCM has also progressed in resisting PrV (Table 2). Epidemiological evidence suggests that dietary flavonoids are beneficial to human health [104, 105].

Table 2 Potential traditional Chinese medicine ingredient against pseudorabies virus

Brain-derived neurotrophic factor (BDNF) is highly involved in neuronal plasticity, protection, and mitochondrial transport and distribution regulation [106]. Increasing BDNF levels protect neurons from insults. This neuroprotective effect of BDNF is achieved primarily by binding to transmembrane tropomyosin-related kinase B (TrkB) on the mitochondrial membrane [107, 108]. PrV infection induced oxidative damage and decreased BDNF levels in rat hippocampal neurons, while 10 µM curcumin improved the viability of PrV-infected (a titer of 3.06 × 106 TCID50) hippocampal neurons [109]. Curcumin, a natural phenolic compound found in Curcuma longa L., has anti-inflammatory, neuroprotective, and antiviral activity [110, 111]. Curcumin can cross the blood–brain barrier and is a promising agent for treating neurodegenerative diseases [112]. The mechanisms underlying the curcumin-mediated neuronal protection against PrV infection is reflected in that curcumin can inhibit the decrease of BDNF and reduce oxidative stress, apoptosis and mitochondrial damage through the BDNF-TrKB pathway [109]. Disease caused by HSV and PrV is characterized by recurrence, and some patients with HSV or PrV infections will need lifelong treatment. Therefore, drug toxicity and residues must be considered in developing new anti-PrV drugs. Curcumin was classified as a “Generally Recognized as Safe” compound and confirmed with excellent tolerance in doses up to 12 g/day in humans [113]. Nevertheless, curcumin has weak bioavailability [114]. Efficient drug delivery systems were developed to solve this shortcoming, such as curcumin nanocrystals [115], nanoliposome-encapsulated curcumin, and curcumin proniosomes [116, 117], and oral supplements with improved curcumin absorption and bioavailability are currently accessible on the market [118, 119].

Kaempferol is a flavonoid found in medicinal plants such as Ginkgo biloba, Tilia spp., Equisetum spp., Moringa oleifera and edible plants, including tea, broccoli, cabbage, and leeks [120]. Kaempferol shows anti-inflammatory [121], antioxidant [122], antiviral [123], and other pharmacological activities [120]. Moreover, kaempferol can inhibit PrV replication in a dose-dependent manner in vitro and exhibits higher anti-PrV activity than acyclovir, an approved therapy drug for HSV-1/2. Acyclovir displays dramatic anti-HSV activity by targeting thymidine kinase and competing with deoxyguanosine triphosphate for the viral DNA polymerase [124]. PrV inhibition by kaempferol reportedly occurs at the early stage of virus replication. In Chinese Kun Ming (KM) mice, kaempferol showed anti-PrV activity through an improved survival rate (22.22%) at 6 days-post-infection, which was higher than that of when acyclovir was administered (16.67%). Kaempferol could inhibit virus replication in several organs, especially in the brain, where viral gene copies were reduced by over 700-fold. At the same time, the pathogenic changes induced by PrV infection in the brain, lung, kidney, heart, and spleen were also alleviated [125]. Viral gene transcription is a critical step in viral replication. Immediate-early gene-, early gene-, and late gene- are the stages of PrV gene expression. PrV-only immediate-early gene IE180 is a transcriptional activator of early gene transcription [126, 127]. Research confirmed that kaempferol considerably inhibited IE180 transcription, leading to decreased transcriptional levels of the early genes EP0 and thymidine kinase. In addition, the latency-associated transcript expression was markedly reduced by kaempferol but not by acyclovir, suggesting that kaempferol could inhibit PrV latency [125].

Quercetin, another flavonoid found in vegetables, fruits, and grains and used in TCM [128], was effective against the PrV strains HNX, Ea, Bartha, and Fa in vitro. In a mouse model, quercetin plantar injection reduced mortality and PrV replication in the brain tissue, ultimately protecting the mice from the lethal challenge. Furthermore, in silico assays suggested that quercetin interacted with the PrV gD-protein and occupied most of the gD subject binding regions, inhibiting PrV adsorption by blocking nectin-1 binding to gD [129]. Intracellular reactive oxygen species (ROS) levels are generally low but can increase rapidly after virus infection, leading to oxidative stress and cell damage [130]. Quercetin could substantially reduce ROS secretion and alleviate the oxidative stress caused by PrV infection in 3D4/2 cells. According to the long non-coding RNA-micro RNA-mRNA network, this process may be related to thioredoxin-interacting protein and nitric oxide synthase 2 decrease [131].

(-)-epigallocatechin-3-gallate (EGCG), a bioactive polyphenol found in tea, has antiviral, antibacterial, antioxidant, anti-inflammatory, and antitumor activities. Recently, 50 mM EGCG could reportedly completely block viral infection at different infection multiplicities in vitro. Additionally, 40 mg/kg EGCG administered pre- and post-challenge in BALB/c mice could provide 100% protection against PrV XJ5 [132]. Additionally, dihydromyricetin extracted from Ampelopsis grossedentata [133], isobavachalcone, a natural chalcone compound extracted from Psoralea corylifolia [134], and an ethyl acetate fraction extracted from Polygonum hydropiper in which rutin was the most abundant compound [135], also showed antiviral activity.

4.2 Polysaccharides

Polysaccharides are essential components and participate in maintaining biological functions. Plant polysaccharides have a wide range of sources, pharmacological effects, and a high safety, all contributing to their broad application in disease prevention and treatment. Platycodon grandiflorus has both edible and medicinal value, and the polysaccharides extracted from this herbal medicine (PGPSt) have an anti-PrV effect [136]. Viruses have evolved to exploit or avoid autophagy to survive by co-existing with their hosts for millions of years. Although PrV lacks the ICP34.5 gene, which can inhibit autophagy in HSV-1, recent studies revealed that autophagy inhibition is not conducive to PrV replication in mouse Neuro-2a cells in vitro [137, 138]. However, PGPSt can reduce PrV replication by inhibiting autophagosomal accumulation induced by PrV infection in vitro. Additionally, 200 µg/mL PGPSt decreased the viral gC mRNA level to 50%, and the viral proteins in PGPSt-treated PK-15 cells were considerably reduced in 12 h [136].

Hippophae rhamnoides polysaccharide (HRP) can enhance immunity and has antioxidant and antitumor activity [139, 140]. HRP has been suggested as a potential anti-PrV drug, which inhibits PrV adsorption and entry and reduces PrV infection-induced oxidative stress. A total of 2000 µg/mL HRP had no apparent toxicity to PK-15 cells, while 1000 µg/mL HRP almost completely inhibited viral protein gB expression. Moreover, its antiviral activity was similar to that of 10 mM lithium chloride (LiCl), a chemical drug with anti-PrV activity. However, HRP is safer than LiCl [141].

Panax notoginseng has various pharmacological effects, including immune modulation, neuroprotection, and antitumor activity [142]. The anti-PrV effect of P. notoginseng polysaccharides is mainly associated with virus adsorption and replication interferences, especially adsorption. The expression of PrV gB protein decreased substantially (98.7% inhibition rate) using 600 µg/mL P. notoginseng polysaccharides, but it cannot inhibit the PrV entry into PK-15 cells. Therefore, the anti-PRV mechanism of P. notoginseng polysaccharides needs further research [143].

Glycyrrhiza polysaccharide (GCP), a common Chinese medicine licorice extract, is widely used worldwide. Reportedly, 600 µg/mL GCP could completely block PrV infection. GCP’s primary antiviral effect was to inhibit PrV attachment and internalization rather than replication and release, indicating that GCP plays an inhibitory role in the early stages of PrV infection [144]. Plantago asiatica polysaccharide (PLP), extracted from the whole P. asiatica plant exerts an antiviral effect by reducing viral adsorption, entry, and intracellular ROS levels rather than affecting PrV replication and inactivation [145]. The intracellular ROS levels in PrV-infected PK-15 cells were markedly decreased and practically returned to normal after PLP treatment [145]. In addition, Radix isatidis polysaccharide and huaier (Trametes robiniophila) polysaccharide have a direct inactivation effect on PrV [146, 147]. In summary, different polysaccharides play antiviral roles in different phases of the PrV life-cycle. Thus, polysaccharide-combination therapy to increase the efficacy or reduce the toxic side effects of drugs may become a new route for PrV treatment.

In addition to flavonoids and polysaccharides, resveratrol (RES), a non-flavonoid polyphenol compound in plants and fruit, shows antiviral activity on PrV. RES has been found to inhibit PrV replication in PK-15 cells [148]. In vivo, RES protects against PrV-induced reproductive failure by recovering serum progesterone levels in mice [149]. Moreover, RES can decrease the mortality of PrV-infected piglets, inhibit viral reproduction, and improve cytokine levels [150]. The RES antiviral mechanism against PrV includes inhibiting IE180 transcriptional activation activity by targeting the Thr601, Ser603, and Pro606 sites of IE180 protein [151].

TCM extracts can also be used as vaccine adjuvants to enhance animals’ immune responses. The PrV live vaccine is mostly commercialized as a lyophilized powder diluted in saline before injection. Ginseng saponins isolated from the stem and leaves of P. ginseng can improve the mice’s immune response to the attenuated PrV (aPrV) vaccine as a diluent component. The aPrV vaccine containing ginseng saponins and sodium selenite (GSe) induced a considerably higher immune response in mice, and the animals showed much higher resistance to the virulent PrV challenge [152]. The function of GSe as an adjuvant was also confirmed in neonatal mice carrying the maternal antibodies (MatAbs). Despite the presence of MatAbs, the specific gB antibody and cytokine responses in neonatal mice immunized with aPrV and saline-GSe were markedly increased, and the protection of vaccinated neonates was enhanced [153]. However, researching natural drugs against PrV is mostly in vitro and the experimental animal stage. Because only a few confirmed cases have been reported, the data on natural drugs against PRV-induced human encephalitis is rare.

5 Conclusions and perspectives

PR causes severe financial losses to the pig industry and considerably affects other animals with an economic interest, such as mink [154]. Furthermore, the presence of PrV in wildlife is an extensive challenge for countries that have eradicated PrV or in areas working towards eradication. In addition, PrV infection in humans requires more attention. On the one hand, as emphasized by Heldwein, “Determining how the triggering signal reaches the fusogen gB represents the next frontier in structural biology of herpesvirus entry” [45]. On the other hand, fully and correctly understanding the invasion process helps develop targeted drugs, antibodies, or blocking agents and for using genetic engineering technology to establish PrV-resistant pigs. However, several details of the PrV entry pattern remain unclear, such as the gH/L action site on gD, the entire PrV gH/L structure, the existence of a gB binding site on gD, and the existence of receptors for gH/L and gB that can mediate PrV invasion. Drugs or antibodies targeting the PrV fusion process may improve therapeutic strategies, but relevant research is scarce. The PrV-gD specific mAb 10B6 binding to gD pro-fusion domain 316QPAEPFP322, a conserved epitope, exhibited effectively neutralizing activities both in vitro and in vivo by blocking gD binding to nectin-1 [155].

Regarding gene editing, a recent study confirmed that the nectin-1/2 knockout in PK-15 cells using the CRISPR/Cas9-mediated gene targeting method conferred resistance to PrV [156]. In vivo, homozygous mutant mice with a single point amino acid mutation (F129A) in nectin-1 had milder symptoms and lower mortality than a heterozygous mutant and wild-type mice artificially infected with a highly pathogenic PrV strain. However, the homozygous mutant mice had deficient eye development [157]. According to the above results, using nectin-1 to establish animals resistant to PrV is a practical approach, although more studies are needed to determine the appropriate mutagenesis mode.

The first isolation of PrV strain hSD-1/2019 from human CSF proves that people can be infected with PRV. Due to the small number of human PrV infection reported cases, there is still a lack of clinical consensus, available treatment guidelines, and prevention experience in human PrV infection. However, a diagnostic workflow of PrV-induced human encephalitis was proposed [93]. The emergence of PrV variants is the primary reason for the classical vaccine immunization failure in pigs and contributes to diagnostic obstacles and treatment of human encephalitis. We speculate that the biological characteristics (such as virulence) of PrV variants are closely related to patients’ incubation period and disease degree. Unfortunately, there are no specific drugs for PR or PrV-induced human encephalitis. Therefore, developing antiviral drugs to prevent and treat PrV is crucial. TCM has multiple medicinal values, which also showed antiviral activity against PrV. Although research on TCM in anti-PRV primarily focuses on a single extract, TCM’s potential is undoubtedly high because TCM compounds are likely to inhibit PrV at different stages of its life-cycle. Moreover, the pathogens are less likely to develop drug resistance than single-component drugs.

In conclusion, studies of the PrV fusion mechanism and how PrV infects humans are the focus of future research. At the same time, developing natural antiviral active ingredients and effective drug targets will also be the main direction of anti-PrV drug research. Therefore, we have a long way to go to eradicate PrV.

References

  1. Davison AJ, Eberle R, Ehlers B, Hayward GS, McGeoch DJ, Minson AC, Pellett PE, Roizman B, Studdert MJ, Thiry E (2009) The order Herpesvirales. Arch Virol 154:171–177

    Article  CAS  PubMed  Google Scholar 

  2. Klupp BG, Hengartner CJ, Mettenleiter TC, Enquist LW (2004) Complete, annotated sequence of the pseudorabies virus genome. J Virol 78:424–440

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mulder WA, Pol JM, Gruys E, Jacobs L, De Jong MC, Peeters BP, Kimman TG (1997) Pseudorabies virus infections in pigs. Role of viral proteins in virulence, pathogenesis and transmission. Vet Res 28:1–17

    CAS  PubMed  Google Scholar 

  4. Pomeranz LE, Reynolds AE, Hengartner CJ (2005) Molecular biology of pseudorabies virus: impact on neurovirology and veterinary medicine. Microbiol Mol Biol Rev 69:462–500

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Di Marco LPV, Moreno A, Castelli A, Ippolito D, Aliberti A, Amato B, Vitale M, Fiasconaro M, Pruiti CF (2021) Retrieving historical cases of Aujeszky’s disease in Sicily (Italy): report of a natural outbreak affecting sheep, goats, dogs, cats and foxes and considerations on critical issues and perspectives in light of the recent EU regulation 429/2016. Pathogens 10:1301

    Article  Google Scholar 

  6. Verin R, Varuzza P, Mazzei M, Poli A (2014) Serologic, molecular, and pathologic survey of pseudorabies virus infection in hunted wild boars (Sus scrofa) in Italy. J Wildlife Dis 50:559–565

    Article  Google Scholar 

  7. Glass CM, McLean RG, Katz JB, Maehr DS, Cropp CB, Kirk LJ, McKeirnan AJ, Evermann JF (1994) Isolation of pseudorabies (Aujeszky’s disease) virus from a Florida panther. J Wildlife Dis 30:180–184

    Article  CAS  Google Scholar 

  8. Hong W, Xiao S, Zhou R, Fang L, He Q, Wu B, Zhou F, Chen H (2002) Protection induced by intramuscular immunization with DNA vaccines of pseudorabies in mice, rabbits and piglets. Vaccine 20:1205–1214

    Article  CAS  PubMed  Google Scholar 

  9. Liu Q, Wang X, Xie C, Ding S, Yang H, Guo S, Li J, Qin L, Ban F, Wang D, Wang C, Feng L, Ma H, Wu B, Zhang L, Dong C, Xing L, Zhang J, Chen H, Yan R, Wang X, Li W (2021) A novel human acute encephalitis caused by pseudorabies virus variant strain. Clin Infect Dis 73:e3690–e3700

    Article  CAS  PubMed  Google Scholar 

  10. Ying M, Hu X, Wang M, Cheng X, Zhao B, Tao Y (2021) Vitritis and retinal vasculitis caused by pseudorabies virus. J Int Med Res 49:675847378

    Article  Google Scholar 

  11. Ai J, Weng S, Cheng Q, Cui P, Li Y, Wu H, Zhu Y, Xu B, Zhang W (2018) Human endophthalmitis caused by pseudorabies virus infection, China, 2017. Emerg Infect Dis 24:1087–1090

    Article  PubMed  PubMed Central  Google Scholar 

  12. Verpoest S, Cay B, Favoreel H, De Regge N (2017) Age-dependent differences in pseudorabies virus neuropathogenesis and associated cytokine expression. J Virol 91:e02058-16

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Freuling CM, Muller TF, Mettenleiter TC (2017) Vaccines against pseudorabies virus (PrV). Vet Microbiol 206:3–9

    Article  CAS  PubMed  Google Scholar 

  14. Koyuncu OO, MacGibeny MA, Enquist LW (2018) Latent versus productive infection: the alpha herpesvirus switch. Future Virol 13:431–443

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu Y, Chen Q, Rao X, Diao X, Yang L, Fang X, Hogeveen H (2019) An economic assessment of pseudorabies (Aujeszky’ disease) elimination on hog farms in China. Prev Vet Med 163:24–30

    Article  PubMed  Google Scholar 

  16. Muller T, Batza HJ, Schluter H, Conraths FJ, Mettenleiter TC (2003) Eradication of Aujeszky’s disease in Germany. J Vet Med B Infect Dis Vet Public Health 50:207–213

    Article  CAS  PubMed  Google Scholar 

  17. Liu A, Xue T, Zhao X, Zou J, Pu H, Hu X, Tian Z (2022) Pseudorabies virus associations in wild animals: review of potential reservoirs for cross-host transmission. Viruses 14:2254

    Article  PubMed  PubMed Central  Google Scholar 

  18. Gaskamp JA, Gee KL, Campbell TA, Silvy NJ, Webb SL (2016) Pseudorabies virus and Brucella abortus from an expanding wild pig (Sus scrofa) population in Southern Oklahoma, USA. J Wildlife Dis 52:383–386

    Article  CAS  Google Scholar 

  19. Sun Y, Liang W, Liu Q, Zhao T, Zhu H, Hua L, Peng Z, Tang X, Stratton CW, Zhou D, Tian Y, Chen H, Wu B (2018) Epidemiological and genetic characteristics of swine pseudorabies virus in mainland China between 2012 and 2017. PeerJ 6:e5785

    Article  PubMed  PubMed Central  Google Scholar 

  20. Owen DJ, Crump CM, Graham SC (2015) Tegument assembly and secondary envelopment of Alphaherpesviruses. Viruses 7:5084–5114

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Radtke K, Kieneke D, Wolfstein A, Michael K, Steffen W, Scholz T, Karger A, Sodeik B (2010) Plus- and minus-end directed microtubule motors bind simultaneously to herpes simplex virus capsids using different inner tegument structures. PLoS Pathog 6:e1000991

    Article  PubMed  PubMed Central  Google Scholar 

  22. Schipke J, Pohlmann A, Diestel R, Binz A, Rudolph K, Nagel CH, Bauerfeind R, Sodeik B (2012) The C terminus of the large tegument protein pUL36 contains multiple capsid binding sites that function differently during assembly and cell entry of herpes simplex virus. J Virol 86:3682–3700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mettenleiter TC, Zsak L, Zuckermann F, Sugg N, Kern H, Ben-Porat T (1990) Interaction of glycoprotein gIII with a cellular heparinlike substance mediates adsorption of pseudorabies virus. J Virol 64:278–286

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Karger A, Mettenleiter TC (1993) Glycoproteins gIII and gp50 play dominant roles in the biphasic attachment of pseudorabies virus. Virology 194:654–664

    Article  CAS  PubMed  Google Scholar 

  25. Oliver SL, Yang E, Arvin AM (2016) Varicella-Zoster virus glycoproteins: entry, replication, and pathogenesis. Curr Clin Microbiol 3:204–215

    Article  Google Scholar 

  26. Li A, Lu G, Qi J, Wu L, Tian K, Luo T, Shi Y, Yan J, Gao GF (2017) Structural basis of nectin-1 recognition by pseudorabies virus glycoprotein D. PLoS Pathog 13:e1006314

    Article  PubMed  PubMed Central  Google Scholar 

  27. Connolly SA, Whitbeck JJ, Rux AH, Krummenacher C, van Drunen LDHS, Cohen GH, Eisenberg RJ (2001) Glycoprotein D homologs in herpes simplex virus type 1, pseudorabies virus, and bovine herpes virus type 1 bind directly to human HveC(nectin-1) with different affinities. Virology 280:7–18

    Article  CAS  PubMed  Google Scholar 

  28. Milne RS, Connolly SA, Krummenacher C, Eisenberg RJ, Cohen GH (2001) Porcine HveC, a member of the highly conserved HveC/nectin 1 family, is a functional alphaherpesvirus receptor. Virology 281:315–328

    Article  CAS  PubMed  Google Scholar 

  29. Cairns TM, Connolly SA (2021) Entry of alphaherpesviruses. Curr Issues Mol Biol 41:63–124

    Article  PubMed  Google Scholar 

  30. Krummenacher C, Supekar VM, Whitbeck JC, Lazear E, Connolly SA, Eisenberg RJ, Cohen GH, Wiley DC, Carfi A (2005) Structure of unliganded HSV gD reveals a mechanism for receptor-mediated activation of virus entry. Embo J 24:4144–4153

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zago A, Jogger CR, Spear PG (2004) Use of herpes simplex virus and pseudorabies virus chimeric glycoprotein D molecules to identify regions critical for membrane fusion. Proc Natl Acad Sci U S A 101:17498–17503

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cocchi F, Fusco D, Menotti L, Gianni T, Eisenberg RJ, Cohen GH, Campadelli-Fiume G (2004) The soluble ectodomain of herpes simplex virus gD contains a membrane-proximal pro-fusion domain and suffices to mediate virus entry. Proc Natl Acad Sci U S A 101:7445–7450

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Takai Y, Ikeda W, Ogita H, Rikitake Y (2008) The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu Rev Cell Dev Bi 24:309–342

    Article  CAS  Google Scholar 

  34. Harrison OJ, Vendome J, Brasch J, Jin X, Hong S, Katsamba PS, Ahlsen G, Troyanovsky RB, Troyanovsky SM, Honig B, Shapiro L (2012) Nectin ectodomain structures reveal a canonical adhesive interface. Nat Struct Mol Biol 19:906–915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ligas MW, Johnson DC (1988) A herpes simplex virus mutant in which glycoprotein D sequences are replaced by beta-galactosidase sequences binds to but is unable to penetrate into cells. J Virol 62:1486–1494

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Schmidt J, Klupp BG, Karger A, Mettenleiter TC (1997) Adaptability in herpesviruses: glycoprotein D-independent infectivity of pseudorabies virus. J Virol 71:17–24

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Schmidt J, Gerdts V, Beyer J, Klupp BG, Mettenleiter TC (2001) Glycoprotein D-independent infectivity of pseudorabies virus results in an alteration of in vivo host range and correlates with mutations in glycoproteins B and H. J Virol 75:10054–10064

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Eisenberg RJ, Atanasiu D, Cairns TM, Gallagher JR, Krummenacher C, Cohen GH (2012) Herpes virus fusion and entry: a story with many characters. Viruses 4:800–832

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cairns TM, Ditto NT, Atanasiu D, Lou H, Brooks BD, Saw WT, Eisenberg RJ, Cohen GH (2019) Surface plasmon resonance reveals direct binding of herpes simplex virus glycoproteins gH/gL to gD and locates a gH/gL binding site on gD. J Virol 93:e00289-19

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Atanasiu D, Saw WT, Eisenberg RJ, Cohen GH (2016) Regulation of herpes simplex virus glycoprotein-induced cascade of events governing cell-cell fusion. J Virol 90:10535–10544

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Backovic M, DuBois RM, Cockburn JJ, Sharff AJ, Vaney MC, Granzow H, Klupp BG, Bricogne G, Mettenleiter TC, Rey FA (2010) Structure of a core fragment of glycoprotein H from pseudorabies virus in complex with antibody. Proc Natl Acad Sci U S A 107:22635–22640

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Matsuura H, Kirschner AN, Longnecker R, Jardetzky TS (2010) Crystal structure of the Epstein-Barr virus (EBV) glycoprotein H/glycoprotein L (gH/gL) complex. Proc Natl Acad Sci U S A 107:22641–22646

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chowdary TK, Cairns TM, Atanasiu D, Cohen GH, Eisenberg RJ, Heldwein EE (2010) Crystal structure of the conserved herpesvirus fusion regulator complex gH-gL. Nat Struct Mol Biol 17:882–888

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xing Y, Oliver SL, Nguyen T, Ciferri C, Nandi A, Hickman J, Giovani C, Yang E, Palladino G, Grose C, Uematsu Y, Lilja AE, Arvin AM, Carfi A (2015) A site of varicella-zoster virus vulnerability identified by structural studies of neutralizing antibodies bound to the glycoprotein complex gHgL. Proc Natl Acad Sci U S A 112:6056–6061

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Heldwein EE (2016) gH/gL supercomplexes at early stages of herpesvirus entry. Curr Opin Virol 18:1–8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bohm SW, Backovic M, Klupp BG, Rey FA, Mettenleiter TC, Fuchs W (2016) Functional Characterization of glycoprotein H chimeras composed of conserved domains of the pseudorabies virus and herpes simplex virus 1 homologs. J Virol 90:421–432

    Article  PubMed  Google Scholar 

  47. Cairns TM, Atanasiu D, Saw WT, Lou H, Whitbeck JC, Ditto NT, Bruun B, Browne H, Bennett L, Wu C, Krummenacher C, Brooks BD, Eisenberg RJ, Cohen GH (2020) Localization of the interaction site of herpes simplex virus glycoprotein D (gD) on the membrane fusion regulator, gH/gL. J Virol 94:e00983-20

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gianni T, Amasio M, Campadelli-Fiume G (2009) Herpes simplex virus gD forms distinct complexes with fusion executors gB and gH/gL in part through the C-terminal profusion domain. J Biol Chem 284:17370–17382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Atanasiu D, Saw WT, Cairns TM, Eisenberg RJ, Cohen GH (2021) Using split luciferase assay and anti-HSV glycoprotein monoclonal antibodies to predict a functional binding site between gD and gH/gL. J Virol 95:e00053-21

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bohm SW, Eckroth E, Backovic M, Klupp BG, Rey FA, Mettenleiter TC, Fuchs W (2015) Structure-based functional analyses of domains II and III of pseudorabies virus glycoprotein H. J Virol 89:1364–1376

    Article  PubMed  Google Scholar 

  51. Bohm SW, Backovic M, Klupp BG, Rey FA, Mettenleiter TC, Fuchs W (2015) A replication defect of pseudorabies virus induced by targeted alpha-helix distortion in the syntaxin-like bundle of glycoprotein H (V275P) is corrected by an adjacent compensatory mutation (V271A). J Gen Virol 96:2349–2354

    Article  PubMed  Google Scholar 

  52. Schroter C, Klupp BG, Fuchs W, Gerhard M, Backovic M, Rey FA, Mettenleiter TC (2014) The highly conserved proline at position 438 in pseudorabies virus gH is important for regulation of membrane fusion. J Virol 88:13064–13072

    Article  PubMed  PubMed Central  Google Scholar 

  53. Fuchs W, Backovic M, Klupp BG, Rey FA, Mettenleiter TC (2012) Structure-based mutational analysis of the highly conserved domain IV of glycoprotein H of pseudorabies virus. J Virol 86:8002–8013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Vallbracht M, Fuchs W, Klupp BG, Mettenleiter TC (2018) functional relevance of the transmembrane domain and cytoplasmic tail of the pseudorabies virus glycoprotein H for membrane fusion. J Virol 92:e00376-18

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Atanasiu D, Saw WT, Cohen GH, Eisenberg RJ (2010) Cascade of events governing cell-cell fusion induced by herpes simplex virus glycoproteins gD, gH/gL, and gB. J Virol 84:12292–12299

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Roop C, Hutchinson L, Johnson DC (1993) A mutant herpes simplex virus type 1 unable to express glycoprotein L cannot enter cells, and its particles lack glycoprotein H. J Virol 67:2285–2297

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hutchinson L, Browne H, Wargent V, Davis-Poynter N, Primorac S, Goldsmith K, Minson AC, Johnson DC (1992) A novel herpes simplex virus glycoprotein, gL, forms a complex with glycoprotein H (gH) and affects normal folding and surface expression of gH. J Virol 66:2240–2250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Duus KM, Grose C (1996) Multiple regulatory effects of varicella-zoster virus (VZV) gL on trafficking patterns and fusogenic properties of VZV gH. J Virol 70:8961–8971

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Klupp BG, Fuchs W, Weiland E, Mettenleiter TC (1997) Pseudorabies virus glycoprotein L is necessary for virus infectivity but dispensable for virion localization of glycoprotein H. J Virol 71:7687–7695

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Vallbracht M, Rehwaldt S, Klupp BG, Mettenleiter TC, Fuchs W (2017) Functional relevance of the N-terminal domain of pseudorabies virus envelope glycoprotein H and its interaction with glycoprotein L. J Virol 91:e00061-17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Klupp BG, Mettenleiter TC (1999) Glycoprotein gL-independent infectivity of pseudorabies virus is mediated by a gD-gH fusion protein. J Virol 73:3014–3022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Schroter C, Vallbracht M, Altenschmidt J, Kargoll S, Fuchs W, Klupp BG, Mettenleiter TC (2015) Mutations in pseudorabies virus glycoproteins gB, gD, and gH functionally compensate for the absence of gL. J Virol 90:2264–2272

    Article  PubMed  Google Scholar 

  63. Vallbracht M, Lotzsch H, Klupp BG, Fuchs W, Vollmer B, Grunewald K, Backovic M, Rey FA, Mettenleiter TC (2021) In vitro viral evolution identifies a critical residue in the alphaherpesvirus fusion glycoprotein B ectodomain that controls gH/gL-independent entry. mBio 12:e00557-21

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Cooper RS, Georgieva ER, Borbat PP, Freed JH, Heldwein EE (2018) Structural basis for membrane anchoring and fusion regulation of the herpes simplex virus fusogen gB. Nat Struct Mol Biol 25:416–424

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stampfer SD, Lou H, Cohen GH, Eisenberg RJ, Heldwein EE (2010) Structural basis of local, pH-dependent conformational changes in glycoprotein B from herpes simplex virus type 1. J Virol 84:12924–12933

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Vallbracht M, Brun D, Tassinari M, Vaney MC, Pehau-Arnaudet G, Guardado-Calvo P, Haouz A, Klupp BG, Mettenleiter TC, Rey FA, Backovic M (2018) Structure-function dissection of pseudorabies virus glycoprotein B fusion loops. J Virol 92:e01203-17

    Article  PubMed  Google Scholar 

  67. Nixdorf R, Klupp BG, Karger A, Mettenleiter TC (2000) Effects of truncation of the carboxy terminus of pseudorabies virus glycoprotein B on infectivity. J Virol 74:7137–7145

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Nixdorf R, Klupp BG, Mettenleiter TC (2001) Restoration of function of carboxy-terminally truncated pseudorabies virus glycoprotein B by point mutations in the ectodomain. J Virol 75:11526–11533

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Pataki Z, Sanders EK, Heldwein EE (2022) A surface pocket in the cytoplasmic domain of the herpes simplex virus fusogen gB controls membrane fusion. PLoS Pathog 18:e1010435

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Jones NA, Geraghty RJ (2004) Fusion activity of lipid-anchored envelope glycoproteins of herpes simplex virus type 1. Virology 324:213–228

    Article  CAS  PubMed  Google Scholar 

  71. Shelly SS, Cairns TM, Whitbeck JC, Lou H, Krummenacher C, Cohen GH, Eisenberg RJ (2012) The membrane-proximal region (MPR) of herpes simplex virus gB regulates association of the fusion loops with lipid membranes. mBio 3:e00429-12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. White JM, Delos SE, Brecher M, Schornberg K (2008) Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Crit Rev Biochem Mol 43:189–219

    Article  CAS  Google Scholar 

  73. Li X, Yang F, Hu X, Tan F, Qi J, Peng R, Wang M, Chai Y, Hao L, Deng J, Bai C, Wang J, Song H, Tan S, Lu G, Gao GF, Shi Y, Tian K (2017) Two classes of protective antibodies against Pseudorabies virus variant glycoprotein B: Implications for vaccine design. PLoS Pathog 13:e1006777

    Article  PubMed  PubMed Central  Google Scholar 

  74. Parry C, Bell S, Minson T, Browne H (2005) Herpes simplex virus type 1 glycoprotein H binds to alphavbeta3 integrins. J Gen Virol 86:7–10

    Article  CAS  PubMed  Google Scholar 

  75. Gianni T, Salvioli S, Chesnokova LS, Hutt-Fletcher LM, Campadelli-Fiume G (2013) alphavbeta6- and alphavbeta8-integrins serve as interchangeable receptors for HSV gH/gL to promote endocytosis and activation of membrane fusion. PLoS Pathog 9:e1003806

    Article  PubMed  PubMed Central  Google Scholar 

  76. Wang C, Liang Q, Sun D, He Y, Jiang J, Guo R, Malla T, Hamrah P, Liu X, Huang Z, Hu K (2022) Nectin-1 and non-muscle myosin heavy chain-IIB: major mediators of herpes simplex virus-1 entry into corneal nerves. Front Microbiol 13:830699

    Article  PubMed  PubMed Central  Google Scholar 

  77. Arii J, Goto H, Suenaga T, Oyama M, Kozuka-Hata H, Imai T, Minowa A, Akashi H, Arase H, Kawaoka Y, Kawaguchi Y (2010) Non-muscle myosin IIA is a functional entry receptor for herpes simplex virus-1. Nature 467:859–862

    Article  CAS  PubMed  Google Scholar 

  78. Suenaga T, Satoh T, Somboonthum P, Kawaguchi Y, Mori Y, Arase H (2010) Myelin-associated glycoprotein mediates membrane fusion and entry of neurotropic herpesviruses. Proc Natl Acad Sci U S A 107:866–871

    Article  CAS  PubMed  Google Scholar 

  79. Satoh T, Arii J, Suenaga T, Wang J, Kogure A, Uehori J, Arase N, Shiratori I, Tanaka S, Kawaguchi Y, Spear PG, Lanier LL, Arase H (2008) PILRalpha is a herpes simplex virus-1 entry coreceptor that associates with glycoprotein B. Cell 132:935–944

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bender FC, Whitbeck JC, Lou H, Cohen GH, Eisenberg RJ (2005) Herpes simplex virus glycoprotein B binds to cell surfaces independently of heparan sulfate and blocks virus entry. J Virol 79:11588–11597

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Li Y, van Drunen LDHS, Babiuk LA, Liang X (1995) Characterization of cell-binding properties of bovine herpesvirus 1 glycoproteins B, C, and D: identification of a dual cell-binding function of gB. J Virol 69:4758–4768

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Skinner GR, Ahmad A, Davies JA (2001) The infrequency of transmission of herpesviruses between humans and animals; postulation of an unrecognised protective host mechanism. Comp Immunol Microbiol Infect Dis 24:255–269

    Article  CAS  PubMed  Google Scholar 

  83. Liu Q, Kuang Y, Li Y, Guo H, Zhou C, Guo S, Tan C, Wu B, Chen H, Wang X (2022) The epidemiology and variation in pseudorabies virus: a continuing challenge to pigs and humans. Viruses 14:1463

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hou Y, Wang Y, Zhang Y, Yu H, Zhao Y, Yi A (2022) Human encephalitis caused by pseudorabies virus in China: a case report and systematic review. Vector Borne Zoonotic Dis 22:391–396

    Article  PubMed  Google Scholar 

  85. Wang Y, Nian H, Li Z, Wang W, Wang X, Cui Y (2019) Human encephalitis complicated with bilateral acute retinal necrosis associated with pseudorabies virus infection: a case report. Int J Infect Dis 89:51–54

    Article  PubMed  Google Scholar 

  86. Yang X, Guan H, Li C, Li Y, Wang S, Zhao X, Zhao Y, Liu Y (2019) Characteristics of human encephalitis caused by pseudorabies virus: a case series study. Int J Infect Dis 87:92–99

    Article  CAS  PubMed  Google Scholar 

  87. Fan S, Yuan H, Liu L, Li H, Wang S, Zhao W, Wu Y, Wang P, Hu Y, Han J, Lyu Y, Zhang W, Chen P, Wu H, Gong Y, Ma Z, Li Y, Yu J, Qiao X, Li G, Zhao Y, Wang D, Ren H, Peng B, Cui L, Wang J, Guan H (2020) Pseudorabies virus encephalitis in humans: a case series study. J Neurovirol 26:556–564

    Article  CAS  PubMed  Google Scholar 

  88. Ciarello FP, Moreno A, Miragliotta N, Antonino A, Fiasconaro M, Purpari G, Amato B, Ippolito D, Di Marco LPV (2022) Aujeszky’s disease in hunting dogs after the ingestion of wild boar raw meat in Sicily (Italy): clinical, diagnostic and phylogenetic features. BMC Vet Res 18:27

    Article  PubMed  PubMed Central  Google Scholar 

  89. Quiroga MI, Nieto JM, Sur J, Osorio F (1998) Diagnosis of Aujeszky’s disease virus infection in dogs by use of immunohistochemistry and in-situ hybridization. Zentralbl Veterinarmed A 45:75–81

    Article  CAS  PubMed  Google Scholar 

  90. Lin W, Shao Y, Tan C, Shen Y, Zhang X, Xiao J, Wu Y, He L, Shao G, Han M, Wang H, Ma J, Xie Q (2019) Commercial vaccine against pseudorabies virus: a hidden health risk for dogs. Vet Microbiol 233:102–112

    Article  CAS  PubMed  Google Scholar 

  91. Deblanc C, Oger A, Simon G, Le Potier MF (2019) Genetic diversity among pseudorabies viruses isolated from dogs in France from 2006 to 2018. Pathogens 8:266

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Li XD, Fu SH, Chen LY, Li F, Deng JH, Lu XC, Wang HY, Tian KG (2020) Detection of pseudorabies virus antibodies in human encephalitis cases. Biomed Environ Sci 33:444–447

    CAS  PubMed  Google Scholar 

  93. Zhou Y, Nie C, Wen H, Long Y, Zhou M, Xie Z, Hong D (2022) Human viral encephalitis associated with suid herpesvirus 1. Neurol Sci 43:2681–2692

    Article  PubMed  Google Scholar 

  94. Yan W, Hu Z, Zhang Y, Wu X, Zhang H (2021) Case Report: metagenomic next-generation sequencing for diagnosis of human encephalitis and endophthalmitis caused by pseudorabies virus. Front Med 8:753988

    Article  Google Scholar 

  95. Wang D, Tao X, Fei M, Chen J, Guo W, Li P, Wang J (2020) Human encephalitis caused by pseudorabies virus infection: a case report. J Neurovirol 26:442–448

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Yang H, Han H, Wang H, Cui Y, Liu H, Ding S (2019) A case of human viral encephalitis caused by pseudorabies virus infection in China. Front Neurol 10:534

    Article  PubMed  PubMed Central  Google Scholar 

  97. Tan L, Yao J, Yang Y, Luo W, Yuan X, Yang L, Wang A (2021) Current status and challenge of pseudorabies virus infection in China. Virol Sin 36:588–607

    Article  PubMed  PubMed Central  Google Scholar 

  98. Wang J, Guo R, Qiao Y, Xu M, Wang Z, Liu Y, Gu Y, Liu C, Hou J (2016) An inactivated gE-deleted pseudorabies vaccine provides complete clinical protection and reduces virus shedding against challenge by a Chinese pseudorabies variant. BMC Vet Res 12:277

    Article  PubMed  PubMed Central  Google Scholar 

  99. Wang J, Song Z, Ge A, Guo R, Qiao Y, Xu M, Wang Z, Liu Y, Zheng Y, Fan H, Hou J (2018) Safety and immunogenicity of an attenuated Chinese pseudorabies variant by dual deletion of TK&gE genes. BMC Vet Res 14:287

    Article  PubMed  PubMed Central  Google Scholar 

  100. Lin J, Li Z, Feng Z, Fang Z, Chen J, Chen W, Liang W, Chen Q (2020) Pseudorabies virus (PRV) strain with defects in gE, gC, and TK genes protects piglets against an emerging PRV variant. J Vet Med Sci 82:846–855

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sun L, Tang Y, Yan K, Zhang H (2022) Construction of a quadruple gene-deleted vaccine confers complete protective immunity against emerging PRV variant challenge in piglets. Virol J 19:19

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Yan S, Huang B, Bai X, Zhou Y, Guo L, Wang T, Shan Y, Wang Y, Tan F, Tian K (2021) Construction and immunogenicity of a recombinant pseudorabies virus variant with TK/gI/gE/11k/28k Deletion. Front Vet Sci 8:797611

    Article  PubMed  Google Scholar 

  103. Wang T, Xiao Y, Yang Q, Wang Y, Sun Z, Zhang C, Yan S, Wang J, Guo L, Yan H, Gao Z, Wang L, Li X, Tan F, Tian K (2015) Construction of a gE-deleted pseudorabies virus and its efficacy to the new-emerging variant PRV challenge in the form of killed vaccine. Biomed Res Int 2015:684945

    PubMed  PubMed Central  Google Scholar 

  104. Ferlay J, Autier P, Boniol M, Heanue M, Colombet M, Boyle P (2007) Estimates of the cancer incidence and mortality in Europe in 2006. Ann Oncol 18:581–592

    Article  CAS  PubMed  Google Scholar 

  105. Ramos S (2008) Cancer chemoprevention and chemotherapy: dietary polyphenols and signalling pathways. Mol Nutr Food Res 52:507–526

    Article  CAS  PubMed  Google Scholar 

  106. Chen SD, Wu CL, Hwang WC, Yang DI (2017) More insight into BDNF against neurodegeneration: anti-apoptosis, anti-oxidation, and suppression of autophagy. Int J Mol Sci 18:545

    Article  PubMed  PubMed Central  Google Scholar 

  107. Wiedemann FR, Siemen D, Mawrin C, Horn TF, Dietzmann K (2006) The neurotrophin receptor TrkB is colocalized to mitochondrial membranes. Int J Biochem Cell B 38:610–620

    Article  CAS  Google Scholar 

  108. Shimoke K, Utsumi T, Kishi S, Nishimura M, Sasaya H, Kudo M, Ikeuchi T (2004) Prevention of endoplasmic reticulum stress-induced cell death by brain-derived neurotrophic factor in cultured cerebral cortical neurons. Brain Res 1028:105–111

    Article  CAS  PubMed  Google Scholar 

  109. Yang B, Luo G, Zhang C, Feng L, Luo X, Gan L (2020) Curcumin protects rat hippocampal neurons against pseudorabies virus by regulating the BDNF/TrkB pathway. Sci Rep 10:22204

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kutluay SB, Doroghazi J, Roemer ME, Triezenberg SJ (2008) Curcumin inhibits herpes simplex virus immediate-early gene expression by a mechanism independent of p300/CBP histone acetyltransferase activity. Virology 373:239–247

    Article  CAS  PubMed  Google Scholar 

  111. Motterlini R, Foresti R, Bassi R, Green CJ (2000) Curcumin, an antioxidant and anti-inflammatory agent, induces heme oxygenase-1 and protects endothelial cells against oxidative stress. Free Radic Biol Med 28:1303–1312

    Article  CAS  PubMed  Google Scholar 

  112. Mythri RB, Bharath MM (2012) Curcumin: a potential neuroprotective agent in Parkinson’s disease. Curr Pharm Design 18:91–99

    Article  CAS  Google Scholar 

  113. Lao CD, Ruffin MT, Normolle D, Heath DD, Murray SI, Bailey JM, Boggs ME, Crowell J, Rock CL, Brenner DE (2006) Dose escalation of a curcuminoid formulation. BMC Complement Altern Med 6:10

    Article  PubMed  PubMed Central  Google Scholar 

  114. Liu W, Zhai Y, Heng X, Che FY, Chen W, Sun D, Zhai G (2016) Oral bioavailability of curcumin: problems and advancements. J Drug Target 24:694–702

    Article  CAS  PubMed  Google Scholar 

  115. Onoue S, Takahashi H, Kawabata Y, Seto Y, Hatanaka J, Timmermann B, Yamada S (2010) Formulation design and photochemical studies on nanocrystal solid dispersion of curcumin with improved oral bioavailability. J Pharm Sci 99:1871–1881

    Article  CAS  PubMed  Google Scholar 

  116. Thangapazham RL, Puri A, Tele S, Blumenthal R, Maheshwari RK (2008) Evaluation of a nanotechnology-based carrier for delivery of curcumin in prostate cancer cells. Int J Oncol 32:1119–1123

    CAS  PubMed  Google Scholar 

  117. Aboali FA, Habib DA, Elbedaiwy HM, Farid RM (2020) Curcumin-loaded proniosomal gel as a biofreindly alternative for treatment of ocular inflammation: In-vitro and in-vivo assessment. Int J Pharm 589:119835

    Article  CAS  PubMed  Google Scholar 

  118. McFarlin BK, Venable AS, Henning AL, Sampson JN, Pennel K, Vingren JL, Hill DW (2016) Reduced inflammatory and muscle damage biomarkers following oral supplementation with bioavailable curcumin. BBA Clin 5:72–78

    Article  PubMed  PubMed Central  Google Scholar 

  119. Kurita T, Makino Y (2013) Novel curcumin oral delivery systems. Anticancer Res 33:2807–2821

    CAS  PubMed  Google Scholar 

  120. Calderon-Montano JM, Burgos-Moron E, Perez-Guerrero C, Lopez-Lazaro M (2011) A review on the dietary flavonoid kaempferol. Mini Rev Med Chem 11:298–344

    Article  CAS  PubMed  Google Scholar 

  121. Park MJ, Lee EK, Heo HS, Kim MS, Sung B, Kim MK, Lee J, Kim ND, Anton S, Choi JS, Yu BP, Chung HY (2009) The anti-inflammatory effect of kaempferol in aged kidney tissues: the involvement of nuclear factor-kappaB via nuclear factor-inducing kinase/IkappaB kinase and mitogen-activated protein kinase pathways. J Med Food 12:351–358

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Santos J, Monte A, Lins T, Barberino RS, Menezes VG, Gouveia BB, Macedo T, Oliveira JJ, Donfack NJ, Matos M (2019) Kaempferol can be used as the single antioxidant in the in vitro culture medium, stimulating sheep secondary follicle development through the phosphatidylinositol 3-kinase signaling pathway. Theriogenology 136:86–94

    Article  CAS  PubMed  Google Scholar 

  123. Zhang R, Ai X, Duan Y, Xue M, He W, Wang C, Xu T, Xu M, Liu B, Li C, Wang Z, Zhang R, Wang G, Tian S, Liu H (2017) Kaempferol ameliorates H9N2 swine influenza virus-induced acute lung injury by inactivation of TLR4/MyD88-mediated NF-kappaB and MAPK signaling pathways. Biomed Pharmacother 89:660–672

    Article  CAS  PubMed  Google Scholar 

  124. Elion GB (1983) The biochemistry and mechanism of action of acyclovir. J Antimicrob Chemother 12:9–17

    Article  CAS  PubMed  Google Scholar 

  125. Li L, Wang R, Hu H, Chen X, Yin Z, Liang X, He C, Yin L, Ye G, Zou Y, Yue G, Tang H, Jia R, Song X (2021) The antiviral activity of kaempferol against pseudorabies virus in mice. BMC Vet Res 17:247

    Article  PubMed  PubMed Central  Google Scholar 

  126. Wu BW, Engel EA, Enquist LW (2014) Characterization of a replication-incompetent pseudorabies virus mutant lacking the sole immediate early gene IE180. mBio 5:e1850

    Article  Google Scholar 

  127. Ono E, Watanabe S, Nikami H, Tasaki T, Kida H (1998) Pseudorabies virus (PRV) early protein 0 activates PRV gene transcription in combination with the immediate-early protein IE180 and enhances the infectivity of PRV genomic DNA. Vet Microbiol 63:99–107

    Article  CAS  PubMed  Google Scholar 

  128. Manach C, Scalbert A, Morand C, Remesy C, Jimenez L (2004) Polyphenols: food sources and bioavailability. Am J Clin Nutr 79:727–747

    Article  CAS  PubMed  Google Scholar 

  129. Sun Y, Li C, Li Z, Shangguan A, Jiang J, Zeng W, Zhang S, He Q (2021) Quercetin as an antiviral agent inhibits the Pseudorabies virus in vitro and in vivo. Virus Res 305:198556

    Article  CAS  PubMed  Google Scholar 

  130. Lai IH, Chang CD, Shih WL (2019) Apoptosis induction by pseudorabies virus via oxidative stress and subsequent DNA damage signaling. Intervirology 62:116–123

    Article  CAS  PubMed  Google Scholar 

  131. Wang Q, Xie X, Chen Q, Yi S, Chen J, Xiao Q, Yu M, Wei Y, Hu T (2022) Effects of quercitrin on PRV-induced secretion of reactive oxygen species and prediction of lncRNA regulatory targets in 3D4/2 cells. Antioxidants 11:631

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Huan C, Xu W, Guo T, Pan H, Zou H, Jiang L, Li C, Gao S (2020) (-)-Epigallocatechin-3-gallate inhibits the life cycle of pseudorabies virus in vitro and protects mice against fatal infection. Front Cell Infect Microbiol 10:616895

    Article  PubMed  Google Scholar 

  133. Sun W, Liu S, Lu A, Yang F, Duan J (2021) In vitro anti-PRV activity of dihydromyricetin from Ampelopsis grossedentata. Nat Prod Res 36:4448–4451

    PubMed  Google Scholar 

  134. Wang Y, Liu TX, Wang TY, Tang YD, Wei P (2020) Isobavachalcone inhibits Pseudorabies virus by impairing virus-induced cell-to-cell fusion. Virol J 17:39

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Ren CZ, Hu WY, Li JC, Xie YH, Jia NN, Shi J, Wei YY, Hu TJ (2020) Ethyl acetate fraction of flavonoids from Polygonum hydropiper L. modulates pseudorabies virus-induced inflammation in RAW264.7 cells via the nuclear factor-kappa B and mitogen-activated protein kinase pathways. J Vet Med Sci 82:1781–1792

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Xing Y, Wang L, Xu G, Guo S, Zhang M, Cheng G, Liu Y, Liu J (2021) Platycodon grandiflorus polysaccharides inhibit Pseudorabies virus replication via downregulating virus-induced autophagy. Res Vet Sci 140:18–25

    Article  CAS  PubMed  Google Scholar 

  137. Xu C, Wang M, Song Z, Wang Z, Liu Q, Jiang P, Bai J, Li Y, Wang X (2018) Pseudorabies virus induces autophagy to enhance viral replication in mouse neuro-2a cells in vitro. Virus Res 248:44–52

    Article  CAS  PubMed  Google Scholar 

  138. Grose C, Buckingham EM, Jackson W, Carpenter JE (2015) The pros and cons of autophagic flux among herpesviruses. Autophagy 11:716–717

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang H, Bi H, Gao T, Zhao B, Ni W, Liu J (2018) A homogalacturonan from Hippophae rhamnoides L. Berries enhance immunomodulatory activity through TLR4/MyD88 pathway mediated activation of macrophages. Int J Biol Macromol 107:1039–1045

    Article  CAS  PubMed  Google Scholar 

  140. Wang H, Gao T, Du Y, Yang H, Wei L, Bi H, Ni W (2015) Anticancer and immunostimulating activities of a novel homogalacturonan from Hippophae rhamnoides L. berry. Carbohyd Polym 131:288–296

    Article  CAS  Google Scholar 

  141. Huan C, Xu Y, Zhang W, Pan H, Zhou Z, Yao J, Guo T, Ni B, Gao S (2022) Hippophae rhamnoides polysaccharides dampen pseudorabies virus infection through downregulating adsorption, entry and oxidative stress. Int J Biol Macromol 207:454–463

    Article  CAS  PubMed  Google Scholar 

  142. Wang T, Guo R, Zhou G, Zhou X, Kou Z, Sui F, Li C, Tang L, Wang Z (2016) Traditional uses, botany, phytochemistry, pharmacology and toxicology of Panax notoginseng (Burk.) F.H. Chen: a review. J Ethnopharmacol 188:234–258

    Article  CAS  PubMed  Google Scholar 

  143. Huan C, Zhou Z, Yao J, Ni B, Gao S (2022) The antiviral effect of panax notoginseng polysaccharides by inhibiting PRV adsorption and replication in vitro. Molecules 27:1254

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Huan C, Xu Y, Zhang W, Ni B, Gao S (2022) Glycyrrhiza polysaccharide inhibits pseudorabies virus infection by interfering with virus attachment and internalization. Viruses 14:1772

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Huan C, Zhang W, Xu Y, Ni B, Gao S (2022) Antiviral activity of Plantago asiatica polysaccharide against pseudorabies virus in vitro. Oxid Med Cell Longev 2022:3570475

    Article  PubMed  PubMed Central  Google Scholar 

  146. Tong C, Chen Z, Liu F, Qiao Y, Chen T, Wang X (2020) Antiviral activities of Radix isatidis polysaccharide against pseudorabies virus in swine testicle cells. BMC Complement Med Ther 20:48

    Article  PubMed  PubMed Central  Google Scholar 

  147. Huan C, Yao J, Xu W, Zhang W, Zhou Z, Pan H, Gao S (2022) Huaier polysaccharide interrupts PRV infection via reducing virus adsorption and entry. Viruses 14:745

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Zhao X, Cui Q, Fu Q, Song X, Jia R, Yang Y, Zou Y, Li L, He C, Liang X, Yin L, Lin J, Ye G, Shu G, Zhao L, Shi F, Lv C, Yin Z (2017) Antiviral properties of resveratrol against pseudorabies virus are associated with the inhibition of IκB kinase activation. Sci Rep 7:8782

    Article  PubMed  PubMed Central  Google Scholar 

  149. Su D, Wu S, Guo J, Wu X, Yang Q, Xiong X (2016) Protective effect of resveratrol against pseudorabies virus-induced reproductive failure in a mouse model. Food Sci Biotechnol 25:103–106

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhao X, Tong W, Song X, Jia R, Li L, Zou Y, He C, Liang X, Lv C, Jing B, Lin J, Yin L, Ye G, Yue G, Wang Y, Yin Z (2018) Antiviral effect of resveratrol in piglets infected with virulent pseudorabies virus. Viruses 10:457

    Article  PubMed  PubMed Central  Google Scholar 

  151. Chen X, Song X, Li L, Chen Y, Jia R, Zou Y, Wan H, Zhao L, Tang H, Lv C, Zhao X, Yin Z (2022) Resveratrol inhibits pseudorabies virus replication by targeting IE180 protein. Front Microbiol 13:891978

    Article  PubMed  PubMed Central  Google Scholar 

  152. Wang Y, Cui X, Yuan L, Maqbool B, Xu W, He S, Guan R, Hu S (2020) A solution with ginseng saponins and selenium as vaccine diluent to increase Th1/Th2 immune responses in mice. J Immunol Res 2020:2714257

    Article  PubMed  PubMed Central  Google Scholar 

  153. Wang Y, Yuan L, Cui X, Xu W, Fang S, Li Z, Lu M, Wu Y, Ma X, Chi X, Hu S (2020) Ginseng stem-leaf saponins in combination with selenium promote the immune response in neonatal mice with maternal antibody. Vaccines 8:755

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Wang GS, Du Y, Wu JQ, Tian FL, Yu XJ, Wang JB (2018) Vaccine resistant pseudorabies virus causes mink infection in China. BMC Vet Res 14:20

    Article  PubMed  PubMed Central  Google Scholar 

  155. Zhang T, Liu Y, Chen Y, Wang J, Feng H, Wei Q, Zhao S, Yang S, Liu D, Zhang G (2021) A monoclonal antibody neutralizes pesudorabies virus by blocking gD binding to the receptor nectin-1. Int J Biol Macromol 188:359–368

    Article  CAS  PubMed  Google Scholar 

  156. Huang Y, Li Z, Song C, Wu Z, Yang H (2020) Resistance to pseudorabies virus by knockout of nectin1/2 in pig cells. Arch Virol 165:2837–2846

    Article  CAS  PubMed  Google Scholar 

  157. Yang X, Yu C, Zhang Q, Hong L, Gu T, Zheng E, Xu Z, Li Z, Song C, Cai G, Wu Z, Yang H (2022) A nectin1 mutant mouse model is resistant to pseudorabies virus infection. Viruses 14:874

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Editage for English language editing services.

Funding

This study is supported by the Introduced talent project of Guizhou University (GDRJHZ [2016]73).

Author information

Authors and Affiliations

Authors

Contributions

NY, BW, WF, TF, TD and ZZ conceived the work; NY, WF and TF collected information, analysed and wrote the manuscript; NY, BW, TD and ZZ edited the manuscript; BW arranged the funding and administered the project. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bin Wang.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Handling editor: Marie Galloux

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ye, N., Feng, W., Fu, T. et al. Membrane fusion, potential threats, and natural antiviral drugs of pseudorabies virus. Vet Res 54, 39 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s13567-023-01171-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13567-023-01171-z

Keywords