Next Article in Journal
Establishing a New Platform to Investigate the Efficacy of Oncolytic Virotherapy in a Human Ex Vivo Peritoneal Carcinomatosis Model
Next Article in Special Issue
Experimental Infection of Domestic Pigs (Sus scrofa) with Rift Valley Fever Virus
Previous Article in Journal
A Renaissance for Oncolytic Adenoviruses?
Previous Article in Special Issue
Risk Factors and Spatiotemporal Analysis of Classical Swine Fever in Ecuador
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transmissible Gastroenteritis Virus: An Update Review and Perspective

1
Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
2
Chongqing Research Institute, Jilin University, Chongqing 401120, China
3
Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2023, 15(2), 359; https://doi.org/10.3390/v15020359
Submission received: 8 December 2022 / Revised: 18 January 2023 / Accepted: 24 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Porcine Viruses 2023)

Abstract

:
Transmissible gastroenteritis virus (TGEV) is a member of the alphacoronavirus genus, which has caused huge threats and losses to pig husbandry with a 100% mortality in infected piglets. TGEV is observed to be recombining and evolving unstoppably in recent years, with some of these recombinant strains spreading across species, which makes the detection and prevention of TGEV more complex. This paper reviews and discusses the basic biological properties of TGEV, factors affecting virulence, viral receptors, and the latest research advances in TGEV infection-induced apoptosis and autophagy to improve understanding of the current status of TGEV and related research processes. We also highlight a possible risk of TGEV being zoonotic, which could be evidenced by the detection of CCoV-HuPn-2018 in humans.

1. Introduction

In the past two decades, the continuous appearance of severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2, swine acute diarrhea syndrome coronavirus (SADS-CoV), and porcine deltacoronavirus (PDCoV) warns us that the coronavirus is still one of the biggest threats to human and livestock health [1,2,3,4,5]. TGEV is a member of the coronavirus subfamily, and although many advances in its study have been made over the past few decades, there are still many unresolved issues of viral virulence and cross-species transmission. Now it cannot be ignored that highly virulent recombinant TGEV strains are still being observed due to frequent evolution and recombination events [6,7,8,9]. Moreover, the sequencing results of the swine enteric coronavirus, SeCoV, reported in European countries, and the CCoV-HuPn-2018 virus in Malaysia both show that these newly discovered coronaviruses are inextricably linked to TGEV [10,11,12,13,14,15], and that the neglect of further work on TGEV will cause delays in public health responses to possible transboundary outbreaks.
TGEV has made entirely new advances in the wave of research on various coronaviruses [16,17,18,19]. In this review, we give a comprehensive overview of the research status of TGEV, discuss the effect of TGEV mutation on virulence, and attempt to find the patterns of change. Apoptosis is important to the process of infection in animals with the virus, may be involved in the proliferation of TGEV infection, and is also associated with severe damage to the intestinal tissue of piglets [20]. Therefore, we reviewed apoptosis induced by TGEV. In addition, we summarize the studies on the proliferation of TGEV in the host involving autophagy, in which inhibitors of the autophagy pathway can be potential antiviral drugs for TGEV. These pieces of information are hoped to help us to predict and deal with the possible future challenges in pig husbandry or new outbreaks in humans and to respond in a timely fashion to the public health problems caused by the spillover of TGEV recombinant coronavirus in the future.

2. TGEV Structure and Epidemiology

2.1. Structure of TGEV

TGEV is an alphacoronavirus with a positive-strand RNA genome of ~28.5 kb [21,22]. The entire viral genome structure includes nine open reading frames (ORF), and the arrangement is 5′ UTR-ORF1a-ORF1b-S-ORF3a-ORF3b-E-M-N-ORF7-3′UTR (Figure 1) [21,23]. ORF1a and ORF1b are involved in genome replication [22,23], occupying 2/3 of the whole TGEV genome [24]. The ORF1 function domain encodes the necessary structural protein for viral genome replication, including polymerase, helicase, and metal-binding motifs [23,25]. Additionally, ORF1 also encodes pp1a/1ab proteins after uncoating, and then 16 non-structural proteins (Nsps) are hydrolyzed by papain-like protease and 3C-like protease [26,27]. Nsp1 is one of the earliest proteins expressed by the virus in the host and is involved in multiple steps that interfere with the host immune response, inhibiting host protein production but not inducing host mRNA cleavage [27,28]. Nsp2 is the main protein involved in activating the NF-κB pathway to induce inflammation during TGEV infection [29]. Nsp3 has two papain-like protease activity domains and is involved in cleavage of Nsp2 and Nsp3 sites and may be involved in cleavage of Nsp3 and Nsp4 sites [30,31]. The main function of Nsp4 is to participate in the formation of double-membrane vesicles (DMVs) during viral infection with Nsp3 [32,33,34]. Nsp5 has protease activity and may be associated with interferon antagonism [35,36,37]. Nsp6 may also be involved in the formation of DMVs [38]. Nsp7–16 combine with each other to form a variety of multi-protein complexes involved in viral transcription and replication, and is associated with serious disruptions of intestinal homeostasis [39,40,41]. Spike (S) protein is a large transmembrane glycoprotein involved in recognizing and binding to host receptor aminopeptidase-N (APN) [42,43]. S protein has hemagglutination activity toward erythrocytes by combining sialic acid on the surface of erythrocytes [44,45]. ORF3 encodes accessory proteins ORF3a and ORF3b. Deletions in ORF3a have been reported in different TGEV and PRCV strains, which are used for homology and phylogenetic alignments between strains [7,9,46,47]. Envelope (E) protein temporarily resides in the pre-Golgi intermediate compartment during coronavirus maturation, which is a critical component for virion envelope morphogenesis [48,49]. Membrane (M) protein is related to the Golgi complex in cells. It is localized to the host Golgi when the M protein expresses alone or in the endoplasmic reticulum–Golgi intermediate compartment in TGEV-infected cells [50]. The M protein initiates virion assembly in the Golgi via virus genomic RNA and nucleoprotein, forming two distinct types of virions within the infected cells: larger virion firstly appears in the perinuclear region; smaller virion aggregates in secretory vesicles and on the cell surface [51,52]. Envelope (E) protein and M protein play an important role in the early maturation stage of the virus, which are involved in the formation of virus-like particles (VLPs) [53,54]. Therefore, the E and M proteins are indispensable in the formation of virion [55,56]. The nucleocapsid (N) protein is involved in the induction of apoptosis due to its cleavage sites of caspase-6 and caspase-7 [57,58]. ORF7 locates at the end of the viral genome and has been used for phylogenetic analysis in some research [59,60].

2.2. Epidemiology of TGEV and PRCV

TGEV was first reported in the United States in 1946 [61], and since then, swine transmissible gastroenteritis (TGE) has been reported successively in America, Europe, and Asia [62,63,64]. Although the prevalence of TGEV in diarrhea pigs in China is very low in recent years [65], there are still several strains prevalent in southern and northern China [7,8,9]. TGEV is divided into two classical subtypes according to the difference of the S protein and ORF3 deletion site. One is the Purdue strain with the 1123–1128nt deletion of the S protein, and this deletion feature was developed by Haelterman’s research team at Purdue University for serial passage of the virus [6,23,66,67,68]. The other is the Miller subtype with the −75–−60nt and 195–223nt deletions of ORF3 (Figure 1) [6,7,9,68].
In addition to the two classical TGEV subtypes, there is another related non-enteric pathogenic TGEV recombinant virus, porcine respiratory coronavirus (PRCV) [64,69]. PRCV is a variant of TGEV with a deletion of 621–681nt in the S protein. Unlike TGEV, PRCV is a respiratory disease (Figure 1) [64,70,71]. PRCV was first reported in Belgium in 1984 [72] and then spread to the Americas [73,74], Japan [75], the United Kingdom [76], and Hungary [77].
The lethality rate of TGEV in piglets is almost as high as 100%, and it has caused huge losses to the breeding industry in Europe, Asia, America, and other places [61,62,63,64]. The main transmission routes of TGEV are fecal–oral transmission, respiratory transmission, and breastfeeding transmission [78,79,80]. The severity of illness after TGEV infection is inversely related to the age of the infected piglets, with symptoms of vomiting, diarrhea, and dehydration two weeks after infection [79,81]. Symptoms in adult pigs include transient elevated body temperature, vomiting, diarrhea, anorexia, and sows stop lactating, with short duration and few deaths [82].
The transmission route of TGEV recombinant virus PRCV is similar to TGEV, and the main infection routes are the fecal–oral route and respiratory route through aerosol [83,84,85]. PRCV reproduces in the lungs and causes symptoms such as coughing, sneezing, fever, bronchoalveolar pneumonia, and interstitial pneumonia after infection [69,83]. Compared with the high virulence and mortality of TGEV [86,87], PRCV is less virulent and causes little pig mortality [84]. In addition, PRCV can be detected in the small intestine, but the virus does not replicate in villous epithelial cells [69,83,88].
The detection rate of TGEV has declined in the decades following the outbreak due to the continuous development of means against TGEV, such as vaccines [89,90], and the detection rate in diarrhea pigs is now very low (approximately <3%) [65,82,91]. Previous studies have shown that piglets can cross-protect against TGEV after PRCV infection, enabling piglets to effectively establish protective immune memory and survive TGEV infection [92,93]. PRCV-infected sows can also induce milk-derived immunity to TGEV in pregnancy, and immune memory can establish even in sows infected with PRCV at a young age [92,93]. Therefore, it may be exactly because of the low lethality rate of PRCV that piglets can establish immune protection against TGEV, so that TGEV does not have the same widespread epidemic effect as PEDV [84,86,92,93].

3. Factors Affecting TGEV Pathogenicity

3.1. Virulence Changes Caused by Fragment Deletion

The S protein is involved in receptor recognition and membrane fusion, delivering the nucleocapsid into host cells, which plays an important role in the virulence of the virus itself, tissue tropism in the host or cell tropism in vitro, and the pathogenesis of coronaviruses [94,95,96,97]. Reverse genetics has identified that the S1 subunit of the S protein is an important site for binding receptors. TGEV first recognizes and binds the key receptor APN on the surface of the cell through the C-terminal domain (CTD) of the S1 subunit [42,97], and then performs the process of invasion and membrane fusion to release the viral genome into the cell [97,98]. In addition, Luis Enjuanes et al. also proved that the S protein of the TGEV virus plays a crucial role in virus virulence and tissue tropism by targeted recombination of the S protein [67,99].
We could observe a classical and attenuating mutation that is considered to be typical of the Purdue strain by summarizing some of the classic virulent and attenuated strains (Table 1) [66,67]. The deletion at the 1123–1128nt site of the S protein is present in classic Purdue strains such as Purdue P115 and PUR46-MAD, and the cloned strains PTV no. C43 and C45, the 43rd and 45th generations obtained by Carlos et al. in TGEV serial passage experiments [6,23,67,68]. Purdue P115 was a clone obtained by passage 115 times on ST cells after the original TGEV strain was first isolated [100]. Thus, Purdue P115 is adapted to a specific cell by high-intensity serial passaging, resulting in the deletion of a segment of the viral genome [101,102]. However, it has been reported that not all strains carrying this 6nt deletion are all attenuated strains. For example, the SHXB strain isolated in China is considered to be a lethal virulent strain after being verified by an animal challenge test [103,104]. The specific reason for the phenomenon of strains carrying the deletion sites that were previously considered to be classical attenuated strains but still being virulent strains is not clear, which may be caused by mutations in other undiscovered sites in the viral genome. The specific mechanism of this phenomenon still needs more in-depth and specific research to verify it.
Strains carrying ORF3 −75–−60nt and 195–223nt deletion are classic Miller strains, but the deletion of these sites has not been reported to be associated with virulence. There is an attenuated strain, Miller M60, that is missing 531nt in ORF3b in the Miller strain (Table 1), and its specific deletion site is ORF3b 405–935nt, which results in a deletion of 67aa [68]. However, there is no experimental result that directly proves that the loss of 531nt in ORF3b will lead to the attenuation of virus virulence.

3.2. Virulence Changes Caused by Point Mutations

The factors that affect the virulence of TGEV are far more than only caused by deletion. We also describe the effect of point mutations on the S gene on virus virulence (Table 2). PRCV has been previously identified to be a variant of the 621–681nt deletion of the S protein of TGEV [64,70,71]. Based on this result, researchers focused on this deletion fragment and demonstrated through targeted recombination that changes in S gene nt655 (PTV-ts-dmar, nt G to T) and nt665 (PTV no.R29, aa L to P) [67,99] lead to attenuation of TGEV and loss of intestinal tropism.
The nt1753 base of the S protein of the firstly isolated Purdue strain is thymine, and the strains carrying T at this site, such as Purdue, TS, Miller M6, and JS2012, are all virulent strains [7]. Other strains that carry 1753T to G (aa585 S to A) mutation are attenuated strains, including attenuated TGEV strains Miller M60, Purdue P115, attenuated H, and PRCV strain PRCV-ISU-1. Therefore, the mutation of amino acid 585 from serine to alanine may be one of the reasons for the reduced virulence of TGEV strains [68]. However, some strains that also carry this mutation have been shown to be virulent, such as the AHHF and SHXB strains [6,103]. The reason for this phenomenon contradicting the previous conclusion is not clear, so whether the S585A locus can indeed lead to the attenuation of TGEV virulence needs to be proved by reverse genetics. C. Krempl et al. reported that aa145 to 155 on TGEV S protein are the sites that affect virulence, sialic acid binding sites, and hemagglutinating activity [105]. When mutations in S protein aa145 P to L, aa147 C to R, or aa155 C to F occur, TGEV loses the sialic acid binding site, hemagglutinating activity, and reduces virulence.
Table 2. Effect of point mutations on viral virulence and tropism.
Table 2. Effect of point mutations on viral virulence and tropism.
StrainIsolateSiteVariationChanges in VirusReference
PurduePTV no.R29Spike665nt T to CLoss of gut tropism; Reduced virulence[67,99]
PurduePTV-ts-dmarSpike655nt G to TLoss of gut tropism; Reduced virulence[67,99]
Purdue, MillerMiller M60, Purdue P115, attenuated H, AHHF, SHXBSpike1753nt T to GReduced virulence[7,68]
PurduePurdue P115Spikeaa145 P to L; aa147 C to R; aa155 C to FLoss of sialic acid binding site; Reduced virulence; Loss of hemagglutinating activity[105]
PurduePUR46-MADORF 1a637nt G to AReduced virulence[106]
The evaluation of the virulence of the virus is based on the virulence of the virus as described by the author in the first report: if the individual challenge experimental piglets die or the pathological sections of the small intestine show severe lesions, this strain is a virulent strain.
Not limited to point mutations in the S protein, Carmen et al. showed that G637A (Gly108Asp) in ORF1 of TGEV affects papain-like protease-mediated cleavage in vitro, resulting in virus attenuation [106].

4. TGEV Receptors

4.1. Aminopeptidase-N

The TGEV receptor is porcine APN (pAPN) [42]. APN is a type 2 transmembrane glycoprotein, approximately 150kDa, and it is highly expressed in intestinal, kidney, and respiratory epithelial cells. APN is mainly associated with the enzymatic regulation of peptides, viral receptors, tumor development, cell motility, and chemotaxis [107]. APN has been identified as a receptor for alphacoronaviruses from several different species, including TGEV [42,108], PRCV [108,109], human coronavirus 229E (HCoV-229E) [110], feline coronavirus (FCoV) [111,112], and canine coronavirus (CCoV) [111,113].
These alphacoronaviruses mentioned above specifically recognize the APNs of their respective host cells through triglycan proteins on the virus surface [114]. Afterwards, the S protein is divided into the S1 subunit, which specifically recognizes the cellular receptors, and the S2 subunit, which is involved in the membrane fusion, after cleavage by host cell proteases [115,116]. The viral S1 subunits have different binding sites of host APNs. Delmas et al. found that pAPN 717aa–813aa is essential for TGEV infection [117]. Kolb et al. proved that human APN (hAPN) aa288–295 is an important site for HCoV-229E by replacing the amino acid sequence QSVNETAQ (pAPN aa283–290) in pAPN with DYVEKQAS (hAPN aa288–295) [118]. Hegyi et al. found that feline APN (fAPN) aa670–840 is an important receptor binding site for FCoV [119], and Tusell et al. further narrowed the site to two regions: aa732–746 and aa764–788 [113].
Interestingly, the relationship between these alphacoronaviruses and APNs is not a one-to-one correspondence between viruses and animals. This means these viruses can bind to APNs from other species. FAPN is capable of being infected by a virus from different terminal hosts: FCoV from cats, TGEV from pigs, HCoV-229E from humans, and CCoV from dogs [111,113]. This finding suggests that when multiple alphacoronaviruses in cats are co-infected, homologous recombination may occur between similar but different strains, and recombination may produce new strains [14,112,120]. The sequencing results of disease samples collected in recent years indicate that TGEV still exists in pig husbandry and is undergoing natural recombination [7,9,10,11,12,13]. This phenomenon reminds us that the recombination and evolution of TGEV still need to be monitored. In response to the occurrence of highly pathogenic mutation events in TGEV, such as the outbreak of the highly pathogenic G2 strain of PEDV that occurred in 2010 [121,122,123] or the recombination virus that causes zoonotic diseases [14], it is necessary to maintain a high degree of vigilance in pig husbandry or public health problems.
APN is also not limited to alphacoronavirus-related receptor binding. A recent study on deltacoronaviruses showed that PDCoV can bind to pAPN through the C-terminal domain (CTD) of the S protein and use pAPN for cell entry [124]. Interestingly, Zhu et al. found that only some residues in the CTD of PDCoV S1 are involved in binding to pAPN [125]. This result is consistent with the finding that CD163 and pAPN double knockout (DKO) pigs can only reduce PDCoV susceptibility, but not block PDCoV [126]. Therefore, deltacoronaviruses can also use APN as a receptor to infect host cells like alphacoronaviruses, but cannot use APNs as a direct receptor that binds to the viral S protein and enters cells like alphacoronaviruses.

4.2. Non-Protein Receptors—Sialic Acid

Besides the classical TGEV receptor pAPN, the non-protein host factor sialic acid also acts as a receptor for binding to TGEV. Sialic acid is an acidic compound with a nine-carbon skeleton that can be used by the four coronaviruses of the alpha-, beta-, gamma-, and deltacoronavirus to bind and enter host cells [127,128,129,130,131,132,133,134]. TGEV is one of these coronaviruses that can use sialic acid as a non-protein receptor [44,105,135].
Unlike TGEVs, which bind to extracellular receptors using S1-CTD, TGEVs bind to sialic acid through their S1-N terminal domain (NTD) [105]. This ability to combine with sialic acid may be obtained by the ancestral coronaviruses, to use carbohydrates as receptors, steal the host galectin as a viral lectin, and then integrate it at the NTD [135,136]. The domain of sialic acid binding may be located in aa145–155 of the S protein, and point mutation or fragment deletion within this amino acid residue will lead to the loss of sialic acid binding activity (Table 2) [105]. TGEV can bind two sialic acids, N-glycolylneuraminic acid (Neu5Gc) and N-acetylneuraminic acid (Neu5Ac), and Neu5Gc is the preferred receptor for TGEV [44]. Conjugates of TGEV and sialic acid effectively protect virions in the stomach and can infect intestinal epithelial cells by binding to mucins across the mucus barrier [45]. The ability to bind sialic acid and form conjugates is dispensable for PRCVs that have lost the NT domain [105]. Although TGEV, PRCV, and HCoV-229E are all alphacoronaviruses that use APN as a receptor, PRCV and HCoV-229E are not enteropathogenic. They may have lost their intestinal tropism while losing their intact NTD. Thus, the sialic acid binding ability may be a key factor in the intestinal phase of alphacoronaviruses [44,105,137,138].

5. Apoptosis Associated with TGEV

TGEV invades the small intestinal epithelium after infection, causing small intestinal villous atrophy and severe diarrhea, which are central events of piglet death caused by infection. This process is closely related to apoptosis. Apoptosis is a process of programmed cell death regulated by genes, which removes damaged or virus-infected cells and plays an important role in viral infectious diseases. Apoptosis pathways are mainly divided into the mitochondrial pathway (intrinsic pathway) and the death receptor pathway (extrinsic pathway) [139,140]. We summarize the mechanism of apoptosis induced by TGEV in Figure 2.
The intrinsic apoptotic pathway is started by changes in mitochondrial membrane permeability, releasing cytochrome c (Cyt c) to activate caspase-9, and then caspase-9 activates caspase-3 to initiate the apoptotic program [141,142,143]. When caspase-3 is activated, the apoptosis program begins [144,145]. The extrinsic apoptotic pathway induces apoptosis through the specific activation of caspase-8 by death receptors, such as FasL, leading to the activation of caspase-3 [146,147]. TGEV infection promotes p53 phosphorylation and induces ROS accumulation by activated p53, leading to the translocation of Bax to mitochondria, the release of Cyt c from mitochondrial oxidative damage, and finally lead to apoptosis [141,148,149].
TGEV infection can activate not only intrinsic apoptotic pathways, but also extrinsic apoptotic pathways. After TGEV infection, caspase-8 indirectly activates caspase-9 by cleaving Bid, resulting in the release of Cyt c, or caspase-8 directly activating caspase- 3 to induce apoptosis [141]. In addition, the TGEV N protein can activate p53 and p21 during the S and G2/M phases, ultimately leading to apoptosis through the intrinsic apoptotic pathway [58]. A very interesting result related to the TGEV N protein is that caspase-6/7 has N protein cleavage activity [57], which also implies that N protein plays a crucial role in TGEV-induced apoptosis.

6. Autography Associated with TGEV

Macroautophagy, also known as autophagy, is a conserved degradation process in eukaryotes. Under stress conditions (such as organelle damage or pathogen invasion), autophagy-related genes (ATGs) mobilize the immune system to capture and eliminate pathogens [150,151,152]. During autophagy, the substrates to be degraded are encapsulated by autophagosomes formed by double-membrane vesicles (DMVs), then fuse with lysosomes, and finally hydrolyze by proteases in lysosomes [153,154]. Previous studies believed that autophagy, as a part of the immune system, has the function of delivering viruses into lysosomes for degradation to prevent infection, and can deliver viral antigens to the immune system [155,156,157,158]. Autophagy may have positive or negative effects on viral replication during viral infection: autophagy can be induced in TGEV infection, and the induction of autophagy can promote TGEV replication [159,160].
Several studies in recent years have demonstrated that the relationship between autophagy and viruses is not simply a positive or negative effect. With the technique called the genome-wide cellular knockout (GeCKO) library, researchers screened host factors for SARS-CoV-2 and other coronaviruses, and a brand new and poorly understood host factor in virus research, TMEM41B, came into view [16,17,18,19]. In previous studies, TMEM41B was found to be a multi-transmembrane protein localized in the endoplasmic reticulum, involved in VTT domain formation, and three independent studies have pointed out that it is involved in the early formation of autophagosomes [161,162,163]. Several recent experiments have shown that TMEM41B acts as a scramblase in the early stages of the autophagosome by flipping lipids in the lipid bilayer membrane structure [164,165], mobilizing lipids on the endoplasmic reticulum to promote membrane curvature [18,161,162,163,166], and forming the bilayer membrane of the autophagosome structure [32].
TMEM41B is critical in multiple viral infections including TGEV in multiple independent studies on multiple viruses and is an essential gene for DMV formation during coronavirus replication [16,17,18,167]. The DMV produced by coronavirus is approximately 200–400 nm in diameter, and the production of this bilayer is as closely related to the endoplasmic reticulum (ER) as the production of autophagosomes. TMEM41B does not seem to affect the binding and entry of extracellular receptors in the early stage of TGEV infection, but it mainly affects the stage of virus replication after entering cells [18,19]. This process is mainly related to the Nsp of the coronavirus. Several previous experiments have demonstrated the interaction between Nsp3 or Nsp4 and TMEM41B of several coronaviruses, including TGEV [18]. It has been proved that Nsps locate on both sides of DMV in DMV formation, which means Nsp3 and 4 are dissociated from each other [32,168]. Therefore, TGEV may take Nsp3 as the core, and through the interaction with Nsp4, hijack the ER in cells and remodel the virus adaptability, and Nsp6 may also be involved in this process [169,170,171,172,173]. After hijacking the ER, virions mobilize TMEM41B and VMP1 through the endoplasmic reticulum to recruit [32,174,175] and mobilize lipids to generate DMV structures necessary for their replication and transcription [18,19]. We summarized this progress in Figure 3. Overall, the discovery of a new host factor for TGEV, TMEM41B, expands our vision in a whole new direction. The new direction can not only promote the exploration of new mechanisms of virus infection but also allow us to have a deeper understanding of the functions of virus non-structural proteins in infection.

7. Conclusions

In the past 20 years, coronaviruses have emerged with alarming speed and high virulence strains, and human diseases such as SARS-CoV, MERS-CoV, and SARS-CoV2 have caused serious public health problems. TGEV is also back in our sights in the pig breeding industry and study of zoonotic disease along with the coronavirus pandemic and outbreak in recent years.
Although the detection rate of TGEV is now very low, it has been continuously detected in China [6,7,8,9], South Korea [176], and Japan [75] in recent years, and TGEV is still evolving and recombining. Naturally recombinant virulent strains JS2012 [7] and AHHF [9], with genotypes between the Purdue strain and Miller strain, were identified in southern China. Like virulent Purdue, AHHF does not carry a deletion of the Purdue signature S protein nt1123–1128 (Table 1), which may explain its high virulence. It also does not carry the deletions of Miller strains ORF3 −75–−60nt and 195–223nt (Table 1), so it was classified as a strain between Purdue and Miller clusters [9]. AHHF and SHXB carry mutations that were previously thought to cause attenuating strains, yet nevertheless show high virulence in challenge experiments. These strains were all detected and reported in China. What causes these strains to carry attenuating mutations but still show high virulence, as well as the reason why frequent recombination occurs and may produce new TGEV clusters (AHHF), needs more in-depth experiments to verify it. Not only limited to the recombination evolution of the TGEV virus itself, Italy [10], Germany [13], Spain [11], and Denmark [12] have also reported the swine enteric coronavirus (SeCoV) composed of natural recombinant strains of PEDV and TGEV. The S gene of SeCoV has more than 90% base identity with PEDV, and the rest of the viral genome has up to 97% base identity with TGEV virulent strains H16 and Miller M6 [10]. These important issues in pig farming caused by TGEV cannot be ignored. The evolution and mutation of these strains may lead to the impact of vaccines and other effective control methods.
TGEV is a cross-species infectious disease; we need to increase our focus on TGEV in this time of accelerated occurrence of zoonotic diseases. The canine–feline-like recombinant alphacoronavirus was recently reported for the first time in a case of pneumonia in Malaysia and was named CCoV-HuPn-2018 [14]. This virus has a CCoV S1 domain and a FCoV S2 domain, and sequence alignment analysis shows that it also has more than 90% similarity with the whole genome of TGEV Purdue (virulent). It is very interesting, and not to be overlooked, that TGEV, CCoV, and FCoV can all use fAPN to enter host cells [111,113]. This study not only reports the emergence of what may be an eighth human disease-causing coronavirus, but that it is also a very dangerous signal of possible future public health problems. In the past, we have rarely conducted in-depth research on the potential threats related to cats and dogs and their corresponding coronaviruses, and the research on pigs and their related coronaviruses has been limited to just solving the problem of economic losses caused by coronaviruses to the breeding industry. We did not pay attention to the hidden dangers that these coronaviruses may cause. The emergence of SeCoV and CCoV-HuPn-2018 reminds us that even though TGEV exists in nature with extremely low activity now, it is still undergoing continuous recombination and evolution. We should not only concentrate on the economic damage that TGEV has caused, but also on the public health problems it may cause in the future as a transboundary infectious disease.
In the recent future, exploring the host factors of TGEV that have not been covered based on GeCKO library technology provides insight into the infection mechanism of this classic alphacoronavirus. It will be important to conduct deeper information mining based on the results and discover and identify these gene functions, which have hardly been studied in the virus field before. Meanwhile, the research results that have been reported may continue to provide new clues for the biology of coronaviruses or other naturally recombined novel coronaviruses in the future. Moreover, these clues allow us to revisit and gain a deeper understanding of this virus that has been quietly lurking in nature for some time, which has implications for the possible future (and possibly contemporary) emergence of a zoonotic recombinant alphacoronavirus and its impact on human public health.

Author Contributions

Writing—original draft preparation, Y.C., Y.Z. and X.W.; illustrations— J.Z., L.M., J.L. and L.Y.; writing—review and editing, H.Y., D.P. and H.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Special Funds for Cultivation and Breeding of New Transgenic Organisms (No. 2016ZX08006003).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef]
  3. Raj, V.S.; Mou, H.; Smits, S.L.; Dekkers, D.H.; Müller, M.A.; Dijkman, R.; Muth, D.; Demmers, J.A.; Zaki, A.; Fouchier, R.A.; et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 2013, 495, 251–254. [Google Scholar] [CrossRef] [Green Version]
  4. Zhou, P.; Fan, H.; Lan, T.; Yang, X.L.; Shi, W.F.; Zhang, W.; Zhu, Y.; Zhang, Y.W.; Xie, Q.M.; Mani, S.; et al. Fatal swine acute diarrhoea syndrome caused by an HKU2-related coronavirus of bat origin. Nature 2018, 556, 255–258. [Google Scholar] [CrossRef] [Green Version]
  5. Wang, L.; Byrum, B.; Zhang, Y. Detection and genetic characterization of deltacoronavirus in pigs, Ohio, USA, 2014. Emerg. Infect Dis. 2014, 20, 1227–1230. [Google Scholar] [CrossRef]
  6. Yuan, D.; Yan, Z.; Li, M.; Wang, Y.; Su, M.; Sun, D. Isolation and Characterization of a Porcine Transmissible Gastroenteritis Coronavirus in Northeast China. Front Vet. Sci. 2021, 8, 611721. [Google Scholar] [CrossRef]
  7. Guo, R.; Fan, B.; Chang, X.; Zhou, J.; Zhao, Y.; Shi, D.; Yu, Z.; He, K.; Li, B. Characterization and evaluation of the pathogenicity of a natural recombinant transmissible gastroenteritis virus in China. Virology 2020, 545, 24–32. [Google Scholar] [CrossRef] [PubMed]
  8. Hu, X., Jr.; Li, N., Jr.; Tian, Z., Jr.; Yin, X., Jr.; Qu, L.; Qu, J. Molecular characterization and phylogenetic analysis of transmissible gastroenteritis virus HX strain isolated from China. BMC Vet. Res. 2015, 11, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Zhang, X.; Zhu, Y.; Zhu, X.; Shi, H.; Chen, J.; Shi, D.; Yuan, J.; Cao, L.; Liu, J.; Dong, H.; et al. Identification of a natural recombinant transmissible gastroenteritis virus between Purdue and Miller clusters in China. Emerg. Microbes Infect 2017, 6, e74. [Google Scholar] [CrossRef] [Green Version]
  10. Boniotti, M.B.; Papetti, A.; Lavazza, A.; Alborali, G.; Sozzi, E.; Chiapponi, C.; Faccini, S.; Bonilauri, P.; Cordioli, P.; Marthaler, D. Porcine Epidemic Diarrhea Virus and Discovery of a Recombinant Swine Enteric Coronavirus, Italy. Emerg. Infect Dis. 2016, 22, 83–87. [Google Scholar] [CrossRef] [PubMed]
  11. de Nova, P.J.G.; Cortey, M.; Díaz, I.; Puente, H.; Rubio, P.; Martín, M.; Carvajal, A. A retrospective study of porcine epidemic diarrhoea virus (PEDV) reveals the presence of swine enteric coronavirus (SeCoV) since 1993 and the recent introduction of a recombinant PEDV-SeCoV in Spain. Transbound Emerg. Dis. 2020, 67, 2911–2922. [Google Scholar] [CrossRef]
  12. Belsham, G.J.; Rasmussen, T.B.; Normann, P.; Vaclavek, P.; Strandbygaard, B.; Bøtner, A. Characterization of a Novel Chimeric Swine Enteric Coronavirus from Diseased Pigs in Central Eastern Europe in 2016. Transbound Emerg. Dis. 2016, 63, 595–601. [Google Scholar] [CrossRef] [PubMed]
  13. Akimkin, V.; Beer, M.; Blome, S.; Hanke, D.; Höper, D.; Jenckel, M.; Pohlmann, A. New Chimeric Porcine Coronavirus in Swine Feces, Germany, 2012. Emerg. Infect Dis. 2016, 22, 1314–1315. [Google Scholar] [CrossRef] [Green Version]
  14. Vlasova, A.N.; Diaz, A.; Damtie, D.; Xiu, L.; Toh, T.H.; Lee, J.S.; Saif, L.J.; Gray, G.C. Novel Canine Coronavirus Isolated from a Hospitalized Patient with Pneumonia in East Malaysia. Clin. Infect Dis. 2022, 74, 446–454. [Google Scholar] [CrossRef] [PubMed]
  15. Tortorici, M.A.; Walls, A.C.; Joshi, A.; Park, Y.J.; Eguia, R.T.; Miranda, M.C.; Kepl, E.; Dosey, A.; Stevens-Ayers, T.; Boeckh, M.J.; et al. Structure, receptor recognition, and antigenicity of the human coronavirus CCoV-HuPn-2018 spike glycoprotein. Cell 2022, 185, 2279–2291.e2217. [Google Scholar] [CrossRef]
  16. Baggen, J.; Persoons, L.; Vanstreels, E.; Jansen, S.; Van Looveren, D.; Boeckx, B.; Geudens, V.; De Man, J.; Jochmans, D.; Wauters, J.; et al. Genome-wide CRISPR screening identifies TMEM106B as a proviral host factor for SARS-CoV-2. Nat. Genet. 2021, 53, 435–444. [Google Scholar] [CrossRef]
  17. Schneider, W.M.; Luna, J.M.; Hoffmann, H.H.; Sánchez-Rivera, F.J.; Leal, A.A.; Ashbrook, A.W.; Le Pen, J.; Ricardo-Lax, I.; Michailidis, E.; Peace, A.; et al. Genome-Scale Identification of SARS-CoV-2 and Pan-coronavirus Host Factor Networks. Cell 2021, 184, 120–132.e114. [Google Scholar] [CrossRef] [PubMed]
  18. Sun, L.; Zhao, C.; Fu, Z.; Fu, Y.; Su, Z.; Li, Y.; Zhou, Y.; Tan, Y.; Li, J.; Xiang, Y.; et al. Genome-scale CRISPR screen identifies TMEM41B as a multi-function host factor required for coronavirus replication. PLoS Pathog. 2021, 17, e1010113. [Google Scholar] [CrossRef]
  19. Trimarco, J.D.; Heaton, B.E.; Chaparian, R.R.; Burke, K.N.; Binder, R.A.; Gray, G.C.; Smith, C.M.; Menachery, V.D.; Heaton, N.S. TMEM41B is a host factor required for the replication of diverse coronaviruses including SARS-CoV-2. PLoS Pathog. 2021, 17, e1009599. [Google Scholar] [CrossRef] [PubMed]
  20. Xu, Z.; Zhang, Y.; Cao, Y. The Roles of Apoptosis in Swine Response to Viral Infection and Pathogenesis of Swine Enteropathogenic Coronaviruses. Front Vet. Sci. 2020, 7, 572425. [Google Scholar] [CrossRef] [PubMed]
  21. Alonso, S.; Izeta, A.; Sola, I.; Enjuanes, L. Transcription regulatory sequences and mRNA expression levels in the coronavirus transmissible gastroenteritis virus. J. Virol. 2002, 76, 1293–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Eleouet, J.F.; Rasschaert, D.; Lambert, P.; Levy, L.; Vende, P.; Laude, H. Complete sequence (20 kilobases) of the polyprotein-encoding gene 1 of transmissible gastroenteritis virus. Virology 1995, 206, 817–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Almazán, F.; González, J.M.; Pénzes, Z.; Izeta, A.; Calvo, E.; Plana-Durán, J.; Enjuanes, L. Engineering the largest RNA virus genome as an infectious bacterial artificial chromosome. Proc. Natl. Acad. Sci. USA 2000, 97, 5516–5521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Brian, D.A.; Baric, R.S. Coronavirus genome structure and replication. Curr. Top. Microbiol. Immunol. 2005, 287, 1–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gorbalenya, A.E.; Koonin, E.V.; Donchenko, A.P.; Blinov, V.M. Coronavirus genome: Prediction of putative functional domains in the non-structural polyprotein by comparative amino acid sequence analysis. Nucleic Acids Res. 1989, 17, 4847–4861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Snijder, E.J.; Decroly, E.; Ziebuhr, J. The Nonstructural Proteins Directing Coronavirus RNA Synthesis and Processing. Adv. Virus Res. 2016, 96, 59–126. [Google Scholar] [CrossRef] [PubMed]
  27. Ziebuhr, J. The coronavirus replicase. Curr. Top. Microbiol. Immunol. 2005, 287, 57–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Huang, C.; Lokugamage, K.G.; Rozovics, J.M.; Narayanan, K.; Semler, B.L.; Makino, S. Alphacoronavirus transmissible gastroenteritis virus nsp1 protein suppresses protein translation in mammalian cells and in cell-free HeLa cell extracts but not in rabbit reticulocyte lysate. J. Virol. 2011, 85, 638–643. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, L.; Qiao, X.; Zhang, S.; Qin, Y.; Guo, T.; Hao, Z.; Sun, L.; Wang, X.; Wang, Y.; Jiang, Y.; et al. Porcine transmissible gastroenteritis virus nonstructural protein 2 contributes to inflammation via NF-κB activation. Virulence 2018, 9, 1685–1698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Wojdyla, J.A.; Manolaridis, I.; van Kasteren, P.B.; Kikkert, M.; Snijder, E.J.; Gorbalenya, A.E.; Tucker, P.A. Papain-like protease 1 from transmissible gastroenteritis virus: Crystal structure and enzymatic activity toward viral and cellular substrates. J. Virol. 2010, 84, 10063–10073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Putics, Á.; Gorbalenya, A.E.; Ziebuhr, J. Identification of protease and ADP-ribose 1′-monophosphatase activities associated with transmissible gastroenteritis virus non-structural protein 3. J. Gen. Virol. 2006, 87, 651–656. [Google Scholar] [CrossRef]
  32. Ji, M.; Li, M.; Sun, L.; Zhao, H.; Li, Y.; Zhou, L.; Yang, Z.; Zhao, X.; Qu, W.; Xue, H.; et al. VMP1 and TMEM41B are essential for DMV formation during β-coronavirus infection. J. Cell Biol. 2022, 221. [Google Scholar] [CrossRef]
  33. Sakai, Y.; Kawachi, K.; Terada, Y.; Omori, H.; Matsuura, Y.; Kamitani, W. Two-amino acids change in the nsp4 of SARS coronavirus abolishes viral replication. Virology 2017, 510, 165–174. [Google Scholar] [CrossRef]
  34. Clementz, M.A.; Kanjanahaluethai, A.; O’Brien, T.E.; Baker, S.C. Mutation in murine coronavirus replication protein nsp4 alters assembly of double membrane vesicles. Virology 2008, 375, 118–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Zhu, X.; Wang, D.; Zhou, J.; Pan, T.; Chen, J.; Yang, Y.; Lv, M.; Ye, X.; Peng, G.; Fang, L.; et al. Porcine Deltacoronavirus nsp5 Antagonizes Type I Interferon Signaling by Cleaving STAT2. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [Green Version]
  36. Chen, J.; Li, Z.; Guo, J.; Xu, S.; Zhou, J.; Chen, Q.; Tong, X.; Wang, D.; Peng, G.; Fang, L.; et al. SARS-CoV-2 nsp5 Exhibits Stronger Catalytic Activity and Interferon Antagonism than Its SARS-CoV Ortholog. J. Virol. 2022, 96, e0003722. [Google Scholar] [CrossRef]
  37. Moustaqil, M.; Ollivier, E.; Chiu, H.P.; Van Tol, S.; Rudolffi-Soto, P.; Stevens, C.; Bhumkar, A.; Hunter, D.J.B.; Freiberg, A.N.; Jacques, D.; et al. SARS-CoV-2 proteases PLpro and 3CLpro cleave IRF3 and critical modulators of inflammatory pathways (NLRP12 and TAB1): Implications for disease presentation across species. Emerg. Microbes Infect 2021, 10, 178–195. [Google Scholar] [CrossRef]
  38. Ricciardi, S.; Guarino, A.M.; Giaquinto, L.; Polishchuk, E.V.; Santoro, M.; Di Tullio, G.; Wilson, C.; Panariello, F.; Soares, V.C.; Dias, S.S.G.; et al. The role of NSP6 in the biogenesis of the SARS-CoV-2 replication organelle. Nature 2022, 606, 761–768. [Google Scholar] [CrossRef]
  39. Hartenian, E.; Nandakumar, D.; Lari, A.; Ly, M.; Tucker, J.M.; Glaunsinger, B.A. The molecular virology of coronaviruses. J. Biol. Chem. 2020, 295, 12910–12934. [Google Scholar] [CrossRef]
  40. Smith, E.C.; Sexton, N.R.; Denison, M.R. Thinking Outside the Triangle: Replication Fidelity of the Largest RNA Viruses. Annu. Rev. Virol. 2014, 1, 111–132. [Google Scholar] [CrossRef]
  41. Wu, A.; Yu, B.; Zhang, K.; Xu, Z.; Wu, D.; He, J.; Luo, J.; Luo, Y.; Yu, J.; Zheng, P.; et al. Transmissible gastroenteritis virus targets Paneth cells to inhibit the self-renewal and differentiation of Lgr5 intestinal stem cells via Notch signaling. Cell Death Dis. 2020, 11, 40. [Google Scholar] [CrossRef] [Green Version]
  42. Delmas, B.; Gelfi, J.; L’Haridon, R.; Vogel, L.K.; Sjöström, H.; Norén, O.; Laude, H. Aminopeptidase N is a major receptor for the entero-pathogenic coronavirus TGEV. Nature 1992, 357, 417–420. [Google Scholar] [CrossRef] [Green Version]
  43. Hansen, G.H.; Delmas, B.; Besnardeau, L.; Vogel, L.K.; Laude, H.; Sjöström, H.; Norén, O. The coronavirus transmissible gastroenteritis virus causes infection after receptor-mediated endocytosis and acid-dependent fusion with an intracellular compartment. J. Virol. 1998, 72, 527–534. [Google Scholar] [CrossRef] [Green Version]
  44. Schultze, B.; Krempl, C.; Ballesteros, M.L.; Shaw, L.; Schauer, R.; Enjuanes, L.; Herrler, G. Transmissible gastroenteritis coronavirus, but not the related porcine respiratory coronavirus, has a sialic acid (N-glycolylneuraminic acid) binding activity. J. Virol. 1996, 70, 5634–5637. [Google Scholar] [CrossRef] [Green Version]
  45. Schwegmann-Wessels, C.; Zimmer, G.; Schröder, B.; Breves, G.; Herrler, G. Binding of transmissible gastroenteritis coronavirus to brush border membrane sialoglycoproteins. J. Virol. 2003, 77, 11846–11848. [Google Scholar] [CrossRef] [Green Version]
  46. Zhang, X.; Zhu, Y.; Zhu, X.; Chen, J.; Shi, H.; Shi, D.; Dong, H.; Feng, L. ORF3a deletion in field strains of porcine-transmissible gastroenteritis virus in China: A hint of association with porcine respiratory coronavirus. Transbound Emerg. Dis. 2017, 64, 698–702. [Google Scholar] [CrossRef] [Green Version]
  47. Kim, L.; Hayes, J.; Lewis, P.; Parwani, A.V.; Chang, K.O.; Saif, L.J. Molecular characterization and pathogenesis of transmissible gastroenteritis coronavirus (TGEV) and porcine respiratory coronavirus (PRCV) field isolates co-circulating in a swine herd. Arch. Virol. 2000, 145, 1133–1147. [Google Scholar] [CrossRef] [Green Version]
  48. Raamsman, M.J.; Locker, J.K.; de Hooge, A.; de Vries, A.A.; Griffiths, G.; Vennema, H.; Rottier, P.J. Characterization of the coronavirus mouse hepatitis virus strain A59 small membrane protein E. J. Virol. 2000, 74, 2333–2342. [Google Scholar] [CrossRef] [Green Version]
  49. Arbely, E.; Khattari, Z.; Brotons, G.; Akkawi, M.; Salditt, T.; Arkin, I.T. A highly unusual palindromic transmembrane helical hairpin formed by SARS coronavirus E protein. J. Mol. Biol. 2004, 341, 769–779. [Google Scholar] [CrossRef]
  50. Weiss, S.R.; Leibowitz, J.L. Coronavirus pathogenesis. Adv. Virus Res. 2011, 81, 85–164. [Google Scholar] [CrossRef]
  51. Risco, C.; Muntión, M.; Enjuanes, L.; Carrascosa, J.L. Two types of virus-related particles are found during transmissible gastroenteritis virus morphogenesis. J. Virol. 1998, 72, 4022–4031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Salanueva, I.J.; Carrascosa, J.L.; Risco, C. Structural maturation of the transmissible gastroenteritis coronavirus. J. Virol. 1999, 73, 7952–7964. [Google Scholar] [CrossRef] [Green Version]
  53. Zou, H.; Zarlenga, D.S.; Sestak, K.; Suo, S.; Ren, X. Transmissible gastroenteritis virus: Identification of M protein-binding peptide ligands with antiviral and diagnostic potential. Antiviral Res. 2013, 99, 383–390. [Google Scholar] [CrossRef] [PubMed]
  54. Baudoux, P.; Carrat, C.; Besnardeau, L.; Charley, B.; Laude, H. Coronavirus pseudoparticles formed with recombinant M and E proteins induce alpha interferon synthesis by leukocytes. J. Virol. 1998, 72, 8636–8643. [Google Scholar] [CrossRef] [Green Version]
  55. Fischer, F.; Stegen, C.F.; Masters, P.S.; Samsonoff, W.A. Analysis of constructed E gene mutants of mouse hepatitis virus confirms a pivotal role for E protein in coronavirus assembly. J. Virol. 1998, 72, 7885–7894. [Google Scholar] [CrossRef] [Green Version]
  56. Vennema, H.; Godeke, G.J.; Rossen, J.W.; Voorhout, W.F.; Horzinek, M.C.; Opstelten, D.J.; Rottier, P.J. Nucleocapsid-independent assembly of coronavirus-like particles by co-expression of viral envelope protein genes. EMBO J. 1996, 15, 2020–2028. [Google Scholar] [CrossRef] [PubMed]
  57. Eléouët, J.F.; Slee, E.A.; Saurini, F.; Castagné, N.; Poncet, D.; Garrido, C.; Solary, E.; Martin, S.J. The viral nucleocapsid protein of transmissible gastroenteritis coronavirus (TGEV) is cleaved by caspase-6 and -7 during TGEV-induced apoptosis. J. Virol. 2000, 74, 3975–3983. [Google Scholar] [CrossRef] [Green Version]
  58. Ding, L.; Huang, Y.; Du, Q.; Dong, F.; Zhao, X.; Zhang, W.; Xu, X.; Tong, D. TGEV nucleocapsid protein induces cell cycle arrest and apoptosis through activation of p53 signaling. Biochem. Biophys. Res. Commun. 2014, 445, 497–503. [Google Scholar] [CrossRef] [PubMed]
  59. Yin, J.C.; Ren, X.F.; Li, Y.J. Molecular cloning and phylogenetic analysis of ORF7 region of chinese isolate TH-98 from transmissible gastroenteritis virus. Virus Genes 2005, 30, 395–401. [Google Scholar] [CrossRef]
  60. Taniguchi, T.; Taniguchi, R.; Takahashi, A.; Hayashidani, H.; Hanaki, K.; Shirai, J.; Honda, E. Sequence comparison of the ORF 7 region of transmissible gastroenteritis viruses isolated in Japan. J. Vet. Med. Sci. 2004, 66, 717–719. [Google Scholar] [CrossRef] [Green Version]
  61. Doyle, L.P.; Hutchings, L.M. A transmissible gastroenteritis in pigs. J. Am. Vet. Med. Assoc. 1946, 108, 257–259. [Google Scholar] [PubMed]
  62. Pritchard, G.C. Transmissible gastroenteritis in endemically infected breeding herds of pigs in East Anglia, 1981–85. Vet. Rec. 1987, 120, 226–230. [Google Scholar] [CrossRef] [PubMed]
  63. McGoldrick, A.; Lowings, J.P.; Paton, D.J. Characterisation of a recent virulent transmissible gastroenteritis virus from Britain with a deleted ORF 3a. Arch. Virol. 1999, 144, 763–770. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, L.; Chang, K.O.; Sestak, K.; Parwani, A.; Saif, L.J. Development of a reverse transcription-nested polymerase chain reaction assay for differential diagnosis of transmissible gastroenteritis virus and porcine respiratory coronavirus from feces and nasal swabs of infected pigs. J. Vet. Diagn. Investig. 2000, 12, 385–388. [Google Scholar] [CrossRef] [Green Version]
  65. Zhang, F.; Luo, S.; Gu, J.; Li, Z.; Li, K.; Yuan, W.; Ye, Y.; Li, H.; Ding, Z.; Song, D.; et al. Prevalence and phylogenetic analysis of porcine diarrhea associated viruses in southern China from 2012 to 2018. BMC Vet. Res. 2019, 15, 470. [Google Scholar] [CrossRef] [Green Version]
  66. Sánchez, C.M.; Gebauer, F.; Suñé, C.; Mendez, A.; Dopazo, J.; Enjuanes, L. Genetic evolution and tropism of transmissible gastroenteritis coronaviruses. Virology 1992, 190, 92–105. [Google Scholar] [CrossRef] [PubMed]
  67. Sánchez, C.M.; Izeta, A.; Sánchez-Morgado, J.M.; Alonso, S.; Sola, I.; Balasch, M.; Plana-Durán, J.; Enjuanes, L. Targeted recombination demonstrates that the spike gene of transmissible gastroenteritis coronavirus is a determinant of its enteric tropism and virulence. J. Virol. 1999, 73, 7607–7618. [Google Scholar] [CrossRef] [Green Version]
  68. Zhang, X.; Hasoksuz, M.; Spiro, D.; Halpin, R.; Wang, S.; Stollar, S.; Janies, D.; Hadya, N.; Tang, Y.; Ghedin, E.; et al. Complete genomic sequences, a key residue in the spike protein and deletions in nonstructural protein 3b of US strains of the virulent and attenuated coronaviruses, transmissible gastroenteritis virus and porcine respiratory coronavirus. Virology 2007, 358, 424–435. [Google Scholar] [CrossRef] [Green Version]
  69. Saif, L.J.; Jung, K. Comparative Pathogenesis of Bovine and Porcine Respiratory Coronaviruses in the Animal Host Species and SARS-CoV-2 in Humans. J. Clin. Microbiol. 2020, 58. [Google Scholar] [CrossRef]
  70. Turlewicz-Podbielska, H.; Pomorska-Mól, M. Porcine Coronaviruses: Overview of the State of the Art. Virol. Sin. 2021, 36, 833–851. [Google Scholar] [CrossRef]
  71. Chen, F.; Knutson, T.P.; Rossow, S.; Saif, L.J.; Marthaler, D.G. Decline of transmissible gastroenteritis virus and its complex evolutionary relationship with porcine respiratory coronavirus in the United States. Sci. Rep. 2019, 9, 3953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Pensaert, M.; Callebaut, P.; Vergote, J. Isolation of a porcine respiratory, non-enteric coronavirus related to transmissible gastroenteritis. Vet. Q. 1986, 8, 257–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wesley, R.D.; Woods, R.D.; McKean, J.D.; Senn, M.K.; Elazhary, Y. Prevalence of coronavirus antibodies in Iowa swine. Can. J. Vet. Res. 1997, 61, 305–308. [Google Scholar]
  74. Wang, L.; Zhang, Y. Genomic Characterization of a New PRCV Variant, United States, 2014. Transbound Emerg. Dis. 2017, 64, 672–674. [Google Scholar] [CrossRef] [PubMed]
  75. Usami, Y.; Fukai, K.; Ichikawa, Y.; Okuda, Y.; Shibata, I.; Motoyama, C.; Imai, K.; Kirisawa, R. Virological and serological studies of porcine respiratory coronavirus infection on a Japanese farm. J. Vet. Med. Sci. 2008, 70, 929–936. [Google Scholar] [CrossRef] [Green Version]
  76. Britton, P.; Mawditt, K.L.; Page, K.W. The cloning and sequencing of the virion protein genes from a British isolate of porcine respiratory coronavirus: Comparison with transmissible gastroenteritis virus genes. Virus Res. 1991, 21, 181–198. [Google Scholar] [CrossRef]
  77. Valkó, A.; Bálint, Á.; Bozsa, Á.; Cságola, A. Prevalence of antibodies against transmissible gastroenteritis virus (TGEV) in Hungary. Vet. Anim. Sci. 2019, 7, 100042. [Google Scholar] [CrossRef]
  78. Kemeny, L.J.; Woods, R.D. Quantitative transmissible gastroenteritis virus shedding patterns in lactating sows. Am. J. Vet. Res. 1977, 38, 307–310. [Google Scholar]
  79. Garwes, D.J. Transmissible gastroenteritis. Vet. Rec. 1988, 122, 462–463. [Google Scholar] [CrossRef]
  80. Casanova, L.; Rutala, W.A.; Weber, D.J.; Sobsey, M.D. Survival of surrogate coronaviruses in water. Water Res. 2009, 43, 1893–1898. [Google Scholar] [CrossRef]
  81. Enjuanes, L.; Smerdou, C.; Castilla, J.; Antón, I.M.; Torres, J.M.; Sola, I.; Golvano, J.; Sánchez, J.M.; Pintado, B. Development of protection against coronavirus induced diseases. A review. Adv. Exp. Med. Biol. 1995, 380, 197–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Saif, L.J. Comparative pathogenesis of enteric viral infections of swine. Adv. Exp. Med. Biol. 1999, 473, 47–59. [Google Scholar] [CrossRef] [PubMed]
  83. Jung, K.; Alekseev, K.P.; Zhang, X.; Cheon, D.S.; Vlasova, A.N.; Saif, L.J. Altered pathogenesis of porcine respiratory coronavirus in pigs due to immunosuppressive effects of dexamethasone: Implications for corticosteroid use in treatment of severe acute respiratory syndrome coronavirus. J. Virol. 2007, 81, 13681–13693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Opriessnig, T.; Giménez-Lirola, L.G.; Halbur, P.G. Polymicrobial respiratory disease in pigs. Anim. Health Res. Rev. 2011, 12, 133–148. [Google Scholar] [CrossRef] [PubMed]
  85. Cox, E.; Pensaert, M.B.; Callebaut, P. Intestinal protection against challenge with transmissible gastroenteritis virus of pigs immune after infection with the porcine respiratory coronavirus. Vaccine 1993, 11, 267–272. [Google Scholar] [CrossRef] [PubMed]
  86. Wesley, R.D.; Woods, R.D.; Cheung, A.K. Genetic analysis of porcine respiratory coronavirus, an attenuated variant of transmissible gastroenteritis virus. J. Virol. 1991, 65, 3369–3373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. van Reeth, K.; Nauwynck, H. Proinflammatory cytokines and viral respiratory disease in pigs. Vet. Res. 2000, 31, 187–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Cox, E.; Hooyberghs, J.; Pensaert, M.B. Sites of replication of a porcine respiratory coronavirus related to transmissible gastroenteritis virus. Res. Vet. Sci. 1990, 48, 165–169. [Google Scholar] [CrossRef] [PubMed]
  89. Pensaert, M.; Cox, E.; van Deun, K.; Callebaut, P. A sero-epizootiological study of porcine respiratory coronavirus in Belgian swine. Vet. Q. 1993, 15, 16–20. [Google Scholar] [CrossRef] [Green Version]
  90. Pritchard, G.C.; Paton, D.J.; Wibberley, G.; Ibata, G. Transmissible gastroenteritis and porcine epidemic diarrhoea in Britain. Vet. Rec. 1999, 144, 616–618. [Google Scholar] [CrossRef] [PubMed]
  91. Kaden, V.; Lange, E.; Hänel, A.; Hlinak, A.; Mewes, L.; Hergarten, G.; Irsch, B.; Dedek, J.; Bruer, W. Retrospective serological survey on selected viral pathogens in wild boar populations in Germany. Eur. J. Wildl. Res. 2009, 55, 153. [Google Scholar] [CrossRef]
  92. Wesley, R.D.; Lager, K.M. Increased litter survival rates, reduced clinical illness and better lactogenic immunity against TGEV in gilts that were primed as neonates with porcine respiratory coronavirus (PRCV). Vet. Microbiol. 2003, 95, 175–186. [Google Scholar] [CrossRef] [PubMed]
  93. Wesley, R.D.; Woods, R.D. Immunization of pregnant gilts with PRCV induces lactogenic immunity for protection of nursing piglets from challenge with TGEV. Vet. Microbiol. 1993, 38, 31–40. [Google Scholar] [CrossRef] [PubMed]
  94. Hulswit, R.J.; de Haan, C.A.; Bosch, B.J. Coronavirus Spike Protein and Tropism Changes. Adv. Virus Res. 2016, 96, 29–57. [Google Scholar] [CrossRef]
  95. Perlman, S.; Netland, J. Coronaviruses post-SARS: Update on replication and pathogenesis. Nat. Rev. Microbiol. 2009, 7, 439–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Enjuanes, L.; Almazán, F.; Sola, I.; Zuñiga, S. Biochemical aspects of coronavirus replication and virus-host interaction. Annu. Rev. Microbiol. 2006, 60, 211–230. [Google Scholar] [CrossRef] [Green Version]
  97. Belouzard, S.; Millet, J.K.; Licitra, B.N.; Whittaker, G.R. Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 2012, 4, 1011–1033. [Google Scholar] [CrossRef] [Green Version]
  98. Bosch, B.J.; van der Zee, R.; de Haan, C.A.; Rottier, P.J. The coronavirus spike protein is a class I virus fusion protein: Structural and functional characterization of the fusion core complex. J. Virol. 2003, 77, 8801–8811. [Google Scholar] [CrossRef] [Green Version]
  99. Ballesteros, M.L.; Sánchez, C.M.; Enjuanes, L. Two amino acid changes at the N-terminus of transmissible gastroenteritis coronavirus spike protein result in the loss of enteric tropism. Virology 1997, 227, 378–388. [Google Scholar] [CrossRef] [Green Version]
  100. Penzes, Z.; Gonzalez, J.M.; Calvo, E.; Izeta, A.; Smerdou, C.; Méndez, A.; Sanchez, C.M.; Sola, I.; Almazan, F.; Enjuanes, L. Complete genome sequence of transmissible gastroenteritis coronavirus PUR46-MAD clone and evolution of the purdue virus cluster. Virus Genes 2001, 23, 105–118. [Google Scholar] [CrossRef]
  101. Wang, T.; Wang, L.; Han, Y.; Pan, L.; Yang, J.; Sun, M.; Zhou, P.; Sun, Y.; Bi, Y.; Qiu, H.J. Adaptation of African swine fever virus to HEK293T cells. Transbound Emerg. Dis. 2021, 68, 2853–2866. [Google Scholar] [CrossRef]
  102. Krug, P.W.; Holinka, L.G.; O’Donnell, V.; Reese, B.; Sanford, B.; Fernandez-Sainz, I.; Gladue, D.P.; Arzt, J.; Rodriguez, L.; Risatti, G.R.; et al. The progressive adaptation of a georgian isolate of African swine fever virus to vero cells leads to a gradual attenuation of virulence in swine corresponding to major modifications of the viral genome. J. Virol. 2015, 89, 2324–2332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zhao, S.; Gao, Q.; Lin, J.; Yan, M.; Yu, Q.; Yang, Q. Differential response of porcine immature monocyte-derived dendritic cells to virulent and inactivated transmissible gastroenteritis virus. Res. Vet. Sci. 2014, 97, 623–630. [Google Scholar] [CrossRef] [PubMed]
  104. Zhao, S.; Gao, Q.; Qin, T.; Yin, Y.; Lin, J.; Yu, Q.; Yang, Q. Effects of virulent and attenuated transmissible gastroenteritis virus on the ability of porcine dendritic cells to sample and present antigen. Vet. Microbiol. 2014, 171, 74–86. [Google Scholar] [CrossRef] [PubMed]
  105. Krempl, C.; Schultze, B.; Laude, H.; Herrler, G. Point mutations in the S protein connect the sialic acid binding activity with the enteropathogenicity of transmissible gastroenteritis coronavirus. J. Virol. 1997, 71, 3285–3287. [Google Scholar] [CrossRef] [Green Version]
  106. Galán, C.; Enjuanes, L.; Almazán, F. A point mutation within the replicase gene differentially affects coronavirus genome versus minigenome replication. J. Virol. 2005, 79, 15016–15026. [Google Scholar] [CrossRef] [Green Version]
  107. Mina-Osorio, P. The moonlighting enzyme CD13: Old and new functions to target. Trends Mol. Med. 2008, 14, 361–371. [Google Scholar] [CrossRef]
  108. Reguera, J.; Santiago, C.; Mudgal, G.; Ordoño, D.; Enjuanes, L.; Casasnovas, J.M. Structural bases of coronavirus attachment to host aminopeptidase N and its inhibition by neutralizing antibodies. PLoS Pathog. 2012, 8, e1002859. [Google Scholar] [CrossRef] [Green Version]
  109. Delmas, B.; Gelfi, J.; Sjöström, H.; Noren, O.; Laude, H. Further characterization of aminopeptidase-N as a receptor for coronaviruses. Adv. Exp. Med. Biol. 1993, 342, 293–298. [Google Scholar] [CrossRef] [Green Version]
  110. Yeager, C.L.; Ashmun, R.A.; Williams, R.K.; Cardellichio, C.B.; Shapiro, L.H.; Look, A.T.; Holmes, K.V. Human aminopeptidase N is a receptor for human coronavirus 229E. Nature 1992, 357, 420–422. [Google Scholar] [CrossRef] [Green Version]
  111. Tresnan, D.B.; Levis, R.; Holmes, K.V. Feline aminopeptidase N serves as a receptor for feline, canine, porcine, and human coronaviruses in serogroup I. J. Virol. 1996, 70, 8669–8674. [Google Scholar] [CrossRef] [Green Version]
  112. Tresnan, D.B.; Holmes, K.V. Feline aminopeptidase N is a receptor for all group I coronaviruses. Adv. Exp. Med. Biol. 1998, 440, 69–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tusell, S.M.; Schittone, S.A.; Holmes, K.V. Mutational analysis of aminopeptidase N, a receptor for several group 1 coronaviruses, identifies key determinants of viral host range. J. Virol. 2007, 81, 1261–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Delmas, B.; Laude, H. Assembly of coronavirus spike protein into trimers and its role in epitope expression. J. Virol. 1990, 64, 5367–5375. [Google Scholar] [CrossRef] [Green Version]
  115. Li, F. Evidence for a common evolutionary origin of coronavirus spike protein receptor-binding subunits. J. Virol. 2012, 86, 2856–2858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Millet, J.K.; Whittaker, G.R. Host cell proteases: Critical determinants of coronavirus tropism and pathogenesis. Virus Res. 2015, 202, 120–134. [Google Scholar] [CrossRef]
  117. Delmas, B.; Gelfi, J.; Kut, E.; Sjöström, H.; Noren, O.; Laude, H. Determinants essential for the transmissible gastroenteritis virus-receptor interaction reside within a domain of aminopeptidase-N that is distinct from the enzymatic site. J. Virol. 1994, 68, 5216–5224. [Google Scholar] [CrossRef] [Green Version]
  118. Kolb, A.F.; Hegyi, A.; Siddell, S.G. Identification of residues critical for the human coronavirus 229E receptor function of human aminopeptidase N. J. Gen. Virol. 1997, 78 Pt 11, 2795–2802. [Google Scholar] [CrossRef]
  119. Hegyi, A.; Kolb, A.F. Characterization of determinants involved in the feline infectious peritonitis virus receptor function of feline aminopeptidase N. J. Gen. Virol. 1998, 79 Pt 6, 1387–1391. [Google Scholar] [CrossRef] [Green Version]
  120. Wardeh, M.; Baylis, M.; Blagrove, M.S.C. Predicting mammalian hosts in which novel coronaviruses can be generated. Nat. Commun. 2021, 12, 780. [Google Scholar] [CrossRef]
  121. Cima, G. PED virus reinfecting U.S. herds. Virus estimated to have killed 7 million-plus pigs. J. Am. Vet. Med. Assoc. 2014, 245, 166–167. [Google Scholar]
  122. Vlasova, A.N.; Marthaler, D.; Wang, Q.; Culhane, M.R.; Rossow, K.D.; Rovira, A.; Collins, J.; Saif, L.J. Distinct characteristics and complex evolution of PEDV strains, North America, May 2013–February 2014. Emerg. Infect Dis. 2014, 20, 1620–1628. [Google Scholar] [CrossRef] [Green Version]
  123. Guo, J.; Fang, L.; Ye, X.; Chen, J.; Xu, S.; Zhu, X.; Miao, Y.; Wang, D.; Xiao, S. Evolutionary and genotypic analyses of global porcine epidemic diarrhea virus strains. Transbound Emerg. Dis. 2019, 66, 111–118. [Google Scholar] [CrossRef] [Green Version]
  124. Wang, B.; Liu, Y.; Ji, C.M.; Yang, Y.L.; Liang, Q.Z.; Zhao, P.; Xu, L.D.; Lei, X.M.; Luo, W.T.; Qin, P.; et al. Porcine Deltacoronavirus Engages the Transmissible Gastroenteritis Virus Functional Receptor Porcine Aminopeptidase N for Infectious Cellular Entry. J. Virol. 2018, 92, e00318-18. [Google Scholar] [CrossRef] [Green Version]
  125. Zhu, X.; Liu, S.; Wang, X.; Luo, Z.; Shi, Y.; Wang, D.; Peng, G.; Chen, H.; Fang, L.; Xiao, S. Contribution of porcine aminopeptidase N to porcine deltacoronavirus infection. Emerg. Microbes Infect 2018, 7, 65. [Google Scholar] [CrossRef] [Green Version]
  126. Xu, K.; Zhou, Y.; Mu, Y.; Liu, Z.; Hou, S.; Xiong, Y.; Fang, L.; Ge, C.; Wei, Y.; Zhang, X.; et al. CD163 and pAPN double-knockout pigs are resistant to PRRSV and TGEV and exhibit decreased susceptibility to PDCoV while maintaining normal production performance. elife 2020, 9, e57132. [Google Scholar] [CrossRef] [PubMed]
  127. Neu, U.; Bauer, J.; Stehle, T. Viruses and sialic acids: Rules of engagement. Curr. Opin. Struct. Biol. 2011, 21, 610–618. [Google Scholar] [CrossRef]
  128. Qing, E.; Hantak, M.; Perlman, S.; Gallagher, T. Distinct Roles for Sialoside and Protein Receptors in Coronavirus Infection. mBio 2020, 11, e02764-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Hulswit, R.J.G.; Lang, Y.; Bakkers, M.J.G.; Li, W.; Li, Z.; Schouten, A.; Ophorst, B.; van Kuppeveld, F.J.M.; Boons, G.J.; Bosch, B.J.; et al. Human coronaviruses OC43 and HKU1 bind to 9-O-acetylated sialic acids via a conserved receptor-binding site in spike protein domain A. Proc. Natl. Acad. Sci. USA 2019, 116, 2681–2690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Li, W.; Hulswit, R.J.G.; Widjaja, I.; Raj, V.S.; McBride, R.; Peng, W.; Widagdo, W.; Tortorici, M.A.; van Dieren, B.; Lang, Y.; et al. Identification of sialic acid-binding function for the Middle East respiratory syndrome coronavirus spike glycoprotein. Proc. Natl. Acad. Sci. USA 2017, 114, E8508–E8517. [Google Scholar] [CrossRef] [Green Version]
  131. Winter, C.; Schwegmann-Weßels, C.; Cavanagh, D.; Neumann, U.; Herrler, G. Sialic acid is a receptor determinant for infection of cells by avian Infectious bronchitis virus. J. Gen. Virol. 2006, 87, 1209–1216. [Google Scholar] [CrossRef] [PubMed]
  132. Promkuntod, N.; van Eijndhoven, R.E.; de Vrieze, G.; Gröne, A.; Verheije, M.H. Mapping of the receptor-binding domain and amino acids critical for attachment in the spike protein of avian coronavirus infectious bronchitis virus. Virology 2014, 448, 26–32. [Google Scholar] [CrossRef] [Green Version]
  133. Slocum, B.; Devine, T. Pelvic osteotomy in the dog as treatment for hip dysplasia. Semin. Vet. Med. Surg. Small Anim. 1987, 2, 107–116. [Google Scholar]
  134. Yuan, Y.; Zu, S.; Zhang, Y.; Zhao, F.; Jin, X.; Hu, H. Porcine Deltacoronavirus Utilizes Sialic Acid as an Attachment Receptor and Trypsin Can Influence the Binding Activity. Viruses 2021, 13, 2442. [Google Scholar] [CrossRef] [PubMed]
  135. Li, F. Receptor recognition mechanisms of coronaviruses: A decade of structural studies. J. Virol. 2015, 89, 1954–1964. [Google Scholar] [CrossRef] [Green Version]
  136. Peng, G.; Sun, D.; Rajashankar, K.R.; Qian, Z.; Holmes, K.V.; Li, F. Crystal structure of mouse coronavirus receptor-binding domain complexed with its murine receptor. Proc. Natl. Acad. Sci. USA 2011, 108, 10696–10701. [Google Scholar] [CrossRef] [Green Version]
  137. Liu, C.; Tang, J.; Ma, Y.; Liang, X.; Yang, Y.; Peng, G.; Qi, Q.; Jiang, S.; Li, J.; Du, L.; et al. Receptor usage and cell entry of porcine epidemic diarrhea coronavirus. J. Virol. 2015, 89, 6121–6125. [Google Scholar] [CrossRef] [Green Version]
  138. Li, C.; Li, W.; Lucio de Esesarte, E.; Guo, H.; van den Elzen, P.; Aarts, E.; van den Born, E.; Rottier, P.J.M.; Bosch, B.J. Cell Attachment Domains of the Porcine Epidemic Diarrhea Virus Spike Protein Are Key Targets of Neutralizing Antibodies. J. Virol. 2017, 91, e00273-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Schmitz, I.; Kirchhoff, S.; Krammer, P.H. Regulation of death receptor-mediated apoptosis pathways. Int. J. Biochem. Cell Biol. 2000, 32, 1123–1136. [Google Scholar] [CrossRef]
  140. Gibson, C.J.; Davids, M.S. BCL-2 Antagonism to Target the Intrinsic Mitochondrial Pathway of Apoptosis. Clin. Cancer Res. 2015, 21, 5021–5029. [Google Scholar] [CrossRef] [Green Version]
  141. Ding, L.; Xu, X.; Huang, Y.; Li, Z.; Zhang, K.; Chen, G.; Yu, G.; Wang, Z.; Li, W.; Tong, D. Transmissible gastroenteritis virus infection induces apoptosis through FasL- and mitochondria-mediated pathways. Vet. Microbiol. 2012, 158, 12–22. [Google Scholar] [CrossRef] [PubMed]
  142. Li, C.Y.; Lee, J.S.; Ko, Y.G.; Kim, J.I.; Seo, J.S. Heat shock protein 70 inhibits apoptosis downstream of cytochrome c release and upstream of caspase-3 activation. J. Biol. Chem. 2000, 275, 25665–25671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jiang, S.; Cai, J.; Wallace, D.C.; Jones, D.P. Cytochrome c-mediated apoptosis in cells lacking mitochondrial DNA. Signaling pathway involving release and caspase 3 activation is conserved. J. Biol. Chem. 1999, 274, 29905–29911. [Google Scholar] [CrossRef] [Green Version]
  144. Kischkel, F.C.; Hellbardt, S.; Behrmann, I.; Germer, M.; Pawlita, M.; Krammer, P.H.; Peter, M.E. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995, 14, 5579–5588. [Google Scholar] [CrossRef] [PubMed]
  145. Sprick, M.R.; Weigand, M.A.; Rieser, E.; Rauch, C.T.; Juo, P.; Blenis, J.; Krammer, P.H.; Walczak, H. FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2. Immunity 2000, 12, 599–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Bortner, C.D.; Cidlowski, J.A. Cellular mechanisms for the repression of apoptosis. Annu. Rev. Pharmacol. Toxicol. 2002, 42, 259–281. [Google Scholar] [CrossRef]
  147. Kroemer, G.; Galluzzi, L.; Vandenabeele, P.; Abrams, J.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; El-Deiry, W.S.; Golstein, P.; Green, D.R.; et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009, 16, 3–11. [Google Scholar] [CrossRef]
  148. Ding, L.; Zhao, X.; Huang, Y.; Du, Q.; Dong, F.; Zhang, H.; Song, X.; Zhang, W.; Tong, D. Regulation of ROS in transmissible gastroenteritis virus-activated apoptotic signaling. Biochem. Biophys. Res. Commun. 2013, 442, 33–37. [Google Scholar] [CrossRef]
  149. Huang, Y.; Ding, L.; Li, Z.; Dai, M.; Zhao, X.; Li, W.; Du, Q.; Xu, X.; Tong, D. Transmissible gastroenteritis virus infection induces cell apoptosis via activation of p53 signalling. J. Gen. Virol. 2013, 94, 1807–1817. [Google Scholar] [CrossRef]
  150. Levine, B.; Mizushima, N.; Virgin, H.W. Autophagy in immunity and inflammation. Nature 2011, 469, 323–335. [Google Scholar] [CrossRef]
  151. Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef] [Green Version]
  152. Miller, K.; McGrath, M.E.; Hu, Z.; Ariannejad, S.; Weston, S.; Frieman, M.; Jackson, W.T. Coronavirus interactions with the cellular autophagy machinery. Autophagy 2020, 16, 2131–2139. [Google Scholar] [CrossRef]
  153. Lamb, C.A.; Yoshimori, T.; Tooze, S.A. The autophagosome: Origins unknown, biogenesis complex. Nat. Rev. Mol. Cell Biol. 2013, 14, 759–774. [Google Scholar] [CrossRef] [PubMed]
  154. Morishita, H.; Mizushima, N. Diverse Cellular Roles of Autophagy. Annu. Rev. Cell Dev. Biol. 2019, 35, 453–475. [Google Scholar] [CrossRef] [PubMed]
  155. English, L.; Chemali, M.; Duron, J.; Rondeau, C.; Laplante, A.; Gingras, D.; Alexander, D.; Leib, D.; Norbury, C.; Lippé, R.; et al. Autophagy enhances the presentation of endogenous viral antigens on MHC class I molecules during HSV-1 infection. Nat. Immunol. 2009, 10, 480–487. [Google Scholar] [CrossRef] [Green Version]
  156. Shibutani, S.T.; Saitoh, T.; Nowag, H.; Münz, C.; Yoshimori, T. Autophagy and autophagy-related proteins in the immune system. Nat. Immunol. 2015, 16, 1014–1024. [Google Scholar] [CrossRef]
  157. Romao, S.; Gannage, M.; Münz, C. Checking the garbage bin for problems in the house, or how autophagy assists in antigen presentation to the immune system. Semin. Cancer Biol. 2013, 23, 391–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Paludan, C.; Schmid, D.; Landthaler, M.; Vockerodt, M.; Kube, D.; Tuschl, T.; Münz, C. Endogenous MHC class II processing of a viral nuclear antigen after autophagy. Science 2005, 307, 593–596. [Google Scholar] [CrossRef]
  159. Xing, Y.; Liqi, Z.; Jian, L.; Qinghua, Y.; Qian, Y. Doxycycline Induces Mitophagy and Suppresses Production of Interferon-β in IPEC-J2 Cells. Front Cell Infect Microbiol. 2017, 7, 21. [Google Scholar] [CrossRef] [Green Version]
  160. Guo, L.; Yu, H.; Gu, W.; Luo, X.; Li, R.; Zhang, J.; Xu, Y.; Yang, L.; Shen, N.; Feng, L.; et al. Autophagy Negatively Regulates Transmissible Gastroenteritis Virus Replication. Sci. Rep. 2016, 6, 23864. [Google Scholar] [CrossRef] [Green Version]
  161. Morita, K.; Hama, Y.; Izume, T.; Tamura, N.; Ueno, T.; Yamashita, Y.; Sakamaki, Y.; Mimura, K.; Morishita, H.; Shihoya, W.; et al. Genome-wide CRISPR screen identifies TMEM41B as a gene required for autophagosome formation. J. Cell Biol. 2018, 217, 3817–3828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Shoemaker, C.J.; Huang, T.Q.; Weir, N.R.; Polyakov, N.J.; Schultz, S.W.; Denic, V. CRISPR screening using an expanded toolkit of autophagy reporters identifies TMEM41B as a novel autophagy factor. PLoS Biol. 2019, 17, e2007044. [Google Scholar] [CrossRef] [Green Version]
  163. Moretti, F.; Bergman, P.; Dodgson, S.; Marcellin, D.; Claerr, I.; Goodwin, J.M.; DeJesus, R.; Kang, Z.; Antczak, C.; Begue, D.; et al. TMEM41B is a novel regulator of autophagy and lipid mobilization. EMBO Rep. 2018, 19, e45889. [Google Scholar] [CrossRef]
  164. Ghanbarpour, A.; Valverde, D.P.; Melia, T.J.; Reinisch, K.M. A model for a partnership of lipid transfer proteins and scramblases in membrane expansion and organelle biogenesis. Proc. Natl. Acad. Sci. USA 2021, 118, e2101562118. [Google Scholar] [CrossRef]
  165. Zhang, T.; Li, Y.E.; Yuan, Y.; Du, X.; Wang, Y.; Dong, X.; Yang, H.; Qi, S. TMEM41B and VMP1 are phospholipid scramblases. Autophagy 2021, 17, 2048–2050. [Google Scholar] [CrossRef]
  166. Morita, K.; Hama, Y.; Mizushima, N. TMEM41B functions with VMP1 in autophagosome formation. Autophagy 2019, 15, 922–923. [Google Scholar] [CrossRef] [PubMed]
  167. Hoffmann, H.H.; Schneider, W.M.; Rozen-Gagnon, K.; Miles, L.A.; Schuster, F.; Razooky, B.; Jacobson, E.; Wu, X.; Yi, S.; Rudin, C.M.; et al. TMEM41B Is a Pan-flavivirus Host Factor. Cell 2021, 184, 133–148.e120. [Google Scholar] [CrossRef] [PubMed]
  168. Oudshoorn, D.; van der Hoeven, B.; Limpens, R.W.; Beugeling, C.; Snijder, E.J.; Bárcena, M.; Kikkert, M. Antiviral Innate Immune Response Interferes with the Formation of Replication-Associated Membrane Structures Induced by a Positive-Strand RNA Virus. mBio 2016, 7, e01991-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Knoops, K.; Bárcena, M.; Limpens, R.W.; Koster, A.J.; Mommaas, A.M.; Snijder, E.J. Ultrastructural characterization of arterivirus replication structures: Reshaping the endoplasmic reticulum to accommodate viral RNA synthesis. J. Virol. 2012, 86, 2474–2487. [Google Scholar] [CrossRef] [Green Version]
  170. Zhang, W.; Chen, K.; Zhang, X.; Guo, C.; Chen, Y.; Liu, X. An integrated analysis of membrane remodeling during porcine reproductive and respiratory syndrome virus replication and assembly. PLoS ONE 2018, 13, e0200919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Oudshoorn, D.; Rijs, K.; Limpens, R.; Groen, K.; Koster, A.J.; Snijder, E.J.; Kikkert, M.; Bárcena, M. Expression and Cleavage of Middle East Respiratory Syndrome Coronavirus nsp3-4 Polyprotein Induce the Formation of Double-Membrane Vesicles That Mimic Those Associated with Coronaviral RNA Replication. mBio 2017, 8, e01658-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Wolff, G.; Limpens, R.; Zevenhoven-Dobbe, J.C.; Laugks, U.; Zheng, S.; de Jong, A.W.M.; Koning, R.I.; Agard, D.A.; Grünewald, K.; Koster, A.J.; et al. A molecular pore spans the double membrane of the coronavirus replication organelle. Science 2020, 369, 1395–1398. [Google Scholar] [CrossRef] [PubMed]
  173. Angelini, M.M.; Akhlaghpour, M.; Neuman, B.W.; Buchmeier, M.J. Severe acute respiratory syndrome coronavirus nonstructural proteins 3, 4, and 6 induce double-membrane vesicles. mBio 2013, 4, e00524-13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kratzel, A.; Kelly, J.N.; V’Kovski, P.; Portmann, J.; Brüggemann, Y.; Todt, D.; Ebert, N.; Shrestha, N.; Plattet, P.; Staab-Weijnitz, C.A.; et al. A genome-wide CRISPR screen identifies interactors of the autophagy pathway as conserved coronavirus targets. PLoS Biol. 2021, 19, e3001490. [Google Scholar] [CrossRef] [PubMed]
  175. Huang, D.; Xu, B.; Liu, L.; Wu, L.; Zhu, Y.; Ghanbarpour, A.; Wang, Y.; Chen, F.J.; Lyu, J.; Hu, Y.; et al. TMEM41B acts as an ER scramblase required for lipoprotein biogenesis and lipid homeostasis. Cell Metab. 2021, 33, 1655–1670.e1658. [Google Scholar] [CrossRef]
  176. Park, J.H.; Han, J.H.; Kwon, H.M. Sequence analysis of the ORF 7 region of transmissible gastroenteritis viruses isolated in Korea. Virus Genes 2008, 36, 71–78. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of the TGEV genome structure. Non-structural proteins, including ORF1a and 1b, ORF3a/3b and ORF7; structural proteins, including spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins. S protein is used to divide different TGEV subtypes: (a) nt1123–1128 deletion of the S protein is considered to be Purdue strain; (b) ORF3 nt−75–−60 and nt195–223 deletions (between S protein and ORF 3) are considered to be Miller subtypes; (c) and TGEV with a deletion of nt621–681 in S protein is variant strain PRCV and infects the porcine respiratory tract.
Figure 1. Schematic diagram of the TGEV genome structure. Non-structural proteins, including ORF1a and 1b, ORF3a/3b and ORF7; structural proteins, including spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins. S protein is used to divide different TGEV subtypes: (a) nt1123–1128 deletion of the S protein is considered to be Purdue strain; (b) ORF3 nt−75–−60 and nt195–223 deletions (between S protein and ORF 3) are considered to be Miller subtypes; (c) and TGEV with a deletion of nt621–681 in S protein is variant strain PRCV and infects the porcine respiratory tract.
Viruses 15 00359 g001
Figure 2. Diagram of the roles of apoptosis in the TGEV infection.
Figure 2. Diagram of the roles of apoptosis in the TGEV infection.
Viruses 15 00359 g002
Figure 3. Schematic diagram of the role of autophagy in TGEV in infected host cells. TGEV infects host cell through the S protein recognition receptor pAPN, and this process can also be achieved by the non-protein receptor sialic acid. Then, it is entered and released through the endosomal pathway. Nsp3 and Nsp4 are hydrolyzed and bound to the endoplasmic reticulum, hijacking the host endoplasmic reticulum and recruiting TMEM41B and VMP1 to form the membrane vesicles necessary for viral replication, a process that may involve Nsp6.
Figure 3. Schematic diagram of the role of autophagy in TGEV in infected host cells. TGEV infects host cell through the S protein recognition receptor pAPN, and this process can also be achieved by the non-protein receptor sialic acid. Then, it is entered and released through the endosomal pathway. Nsp3 and Nsp4 are hydrolyzed and bound to the endoplasmic reticulum, hijacking the host endoplasmic reticulum and recruiting TMEM41B and VMP1 to form the membrane vesicles necessary for viral replication, a process that may involve Nsp6.
Viruses 15 00359 g003
Table 1. Representative strains, classic variation, and pathogenicity of each TGEV genotype.
Table 1. Representative strains, classic variation, and pathogenicity of each TGEV genotype.
StrainIsolateVariationPathogenicityGenbank IDReference
PurdueVirulent PurdueNAVirulentDQ811789.2[6,68]
PurduePurdue P115S protein nt1123–1128 deletionAttenuatedDQ811788.1[6,68]
PurduePUR46-MADS protein nt1123–1128 deletionAttenuatedNC_038861.1[6,23]
PurdueSHXBS protein nt1123–1128 deletionVirulentKP202848.1[103,104]
Between Purdue and MillerAHHFS protein nt2386–2388 deletionVirulentKX499468.1[6,9]
MillerH16ORF3 −75–−60nt and 195–223nt deletion, S protein nt2386–2388 deletionVirulentFJ755618.2[6,9]
MillerMiller M60ORF3 −75–−60nt and 195–223nt deletion, ORF3b 405–935nt deletionAttenuatedDQ811786.2[6,68]
MillerMiller M6ORF3 −75–−60nt and 195–223nt deletionVirulentDQ811785.1[6,68]
MillerJS2012ORF3 −75–−60nt and 195–223nt deletionVirulentKT696544.1[6,7]
Millerattenuated HORF3 −75–−60nt and 195–223nt deletion, S nt2386–2388 deletionAttenuatedEU074218.2[9]
The evaluation of the virulence of the virus is based on the virulence of the virus as described by the author in the first report: if the individual challenge experimental piglets die or the pathological sections of the small intestine show severe lesions, this strain is a virulent strain.
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

Chen, Y.; Zhang, Y.; Wang, X.; Zhou, J.; Ma, L.; Li, J.; Yang, L.; Ouyang, H.; Yuan, H.; Pang, D. Transmissible Gastroenteritis Virus: An Update Review and Perspective. Viruses 2023, 15, 359. https://doi.org/10.3390/v15020359

AMA Style

Chen Y, Zhang Y, Wang X, Zhou J, Ma L, Li J, Yang L, Ouyang H, Yuan H, Pang D. Transmissible Gastroenteritis Virus: An Update Review and Perspective. Viruses. 2023; 15(2):359. https://doi.org/10.3390/v15020359

Chicago/Turabian Style

Chen, Yiwu, Yuanzhu Zhang, Xi Wang, Jian Zhou, Lerong Ma, Jianing Li, Lin Yang, Hongsheng Ouyang, Hongming Yuan, and Daxin Pang. 2023. "Transmissible Gastroenteritis Virus: An Update Review and Perspective" Viruses 15, no. 2: 359. https://doi.org/10.3390/v15020359

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