key: cord-0751424-9q1i20o6 authors: Jung, Kwonil; Saif, Linda J.; Wang, Qiuhong title: Porcine epidemic diarrhea virus (PEDV): An update on etiology, transmission, pathogenesis, and prevention and control date: 2020-06-02 journal: Virus Res DOI: 10.1016/j.virusres.2020.198045 sha: 90ed73b90d735bb33f08ab033b3e3dfa523549e6 doc_id: 751424 cord_uid: 9q1i20o6 Porcine epidemic diarrhea virus (PEDV), a member of the genus Alphacoronavirus in the family Coronaviridae, causes acute diarrhea and/or vomiting, dehydration and high mortality in neonatal piglets. Two different genogroups of PEDV, S INDEL [PEDV variant containing multiple deletions and insertions in the S1 subunit of the spike (S) protein, G1b] and non-S INDEL (G2b) strains were detected during the diarrheal disease outbreak in US swine in 2013-2014. Similar viruses are also circulating globally. Continuous improvement and update of biosecurity and vaccine strains and protocols are still needed to control and prevent PEDV infections worldwide. Although the non-S INDEL PEDV was highly virulent and the S INDEL PEDV caused milder disease, the latter has the capacity to cause illness in a high number of piglets on farms with low biosecurity and herd immunity. The main PEDV transmission route is fecal–oral, but airborne transmission via the fecal–nasal route may play a role in pig-to-pig and farm-to-farm spread. PEDV infection of neonatal pigs causes fecal virus shedding (alongside frequent detection of PEDV RNA in the nasal cavity), acute viremia, severe atrophic enteritis (mainly jejunum and ileum), and increased pro-inflammatory and innate immune responses. PEDV-specific IgA effector and memory B cells in orally primed sows play a critical role in sow lactogenic immunity and passive protection of piglets. This review focuses on the etiology, transmission, pathogenesis, and prevention and control of PEDV infection. Porcine epidemic diarrhea virus (PEDV), a member of the genus Alphacoronavirus in the family Coronaviridae of the order Nidovirales, causes acute diarrhea, vomiting, dehydration and high mortality in neonatal piglets. The disease was reported in the European and Asian pig industries over the last 30 years, with the virus first appearing in England (Wood, 1977) and J o u r n a l P r e -p r o o f One study reported that four sows exposed via feedback to a G1b S INDEL PEDV at approximately seven months pre-farrowing and then re-exposed to a G2b non-S INDEL PEDV at day 109 of gestation provided long-term (7 months), passive immune protection of piglets against challenge with the same G2b non-S INDEL PEDV (Goede et al., 2015) . There was 0% mortality rate of piglets (with 57% reduced incidence of diarrhea), compared with mean 33% mortality and 100% morbidity rates of piglets born to non-immunized sows. However, another study revealed that more than 80% of the piglets inoculated orally with a G1b S INDEL PEDV (Iowa106 strain) at 3-4 days of age had diarrhea after challenging with a G2b non-S INDEL PEDV at 24 days of age, whereas none of the piglets inoculated previously and then challenged with the same non-S INDEL PEDV showed diarrhea (Annamalai et al., 2017; Lin et al., 2015a) . Antigenic cross-reactivity or cross-neutralization between PEDV and feline infectious peritonitis virus (FIPV) was detected by enzyme linked immunosorbent assay (ELISA), immunoblotting and immune-precipitation (Zhou et al., 1988) , or VN tests (Zhao et al., 2019) . Despite no serological cross-neutralization detected between PEDV and TGEV (Hofmann and Wyler, 1989; Pensaert and de Bouck, 1978) , investigators found some antigenic cross-activity between PEDV and TGEV (Miller, but not Purdue strain) based on at least two conserved epitopes on the N-terminal region of their N proteins, as well as via their M proteins or whole virus particles (Gimenez-Lirola et al., 2017; Lin et al., 2015b; Xie et al., 2019) . Truncation of the N-terminal region of the N protein or avoidance of M-or purified whole virus-based serologic tests may eliminate PEDV and TGEV shared epitope(s) or reduce the possible cross-reactivity for serologic J o u r n a l P r e -p r o o f assays (Gimenez-Lirola et al., 2017; Xie et al., 2019) . Polyclonal hyperimmune antisera against PDCoV did not cross-react with PEDV . Among N, M and E proteins and whole PEDV particles, only the M protein of PEDV showed cross-reactivity in 1/12 convalescent sera from PDCoV-infected pigs (Gimenez-Lirola et al., 2017) . However, one study reported antigenic cross-reactivity between US PDCoV and PEDV strains, possibly due to at least one shared epitope in the N-terminal region of their N proteins . For SADS-CoV, monoclonal antibody against the N protein did not cross-react with PEDV, TGEV or PDCoV (Pan et al., 2017) . The fecal-oral route is the main means of direct transmission of PEDV via the feces and/or vomitus of infected pigs (Jung and Saif, 2015) . Indirect contact transmission of PEDV is also frequent within and between farms, particularly, with a low biosecurity, via other contaminated fomites including (Jung and Saif, 2015; Kim et al., 2017b) : transport trailers (Lowe et al., 2014) , farm workers' hands, boots and clothes (Kim et al., 2017b) , feed (Bowman et al., 2015a; Dee et al., 2014; Schumacher et al., 2017) , feed ingredients and additives, such as spray-dried porcine plasma (Pasick et al., 2014; Perri et al., 2018) , and feed totes used for transporting bulk feed or feed ingredients (Anon., 2015; Scott et al., 2016) . PEDV remained infectious on tote material for 35 days at room temperature (Scott et al., 2016) . PEDV cross-contamination also occurred during feed manufacturing (Schumacher et al., 2018) . J o u r n a l P r e -p r o o f 8 The fecal-nasal route is another route of pig-to-pig, or farm-to-farm (up to 10 miles away) airborne transmission of PEDV via aerosolized PEDV particles that are infectious in nursing pigs (Alonso et al., 2014; Alonso et al., 2015; Beam et al., 2015; Gallien et al., 2018a; . Airborne PEDV transmission occurs within farrowing herds where newborn piglets, highly susceptible to the virus, are raised (Alonso et al., 2015; Niederwerder et al., 2016) . The nasal cavity of naïve pigs housed at a distance from clinical pigs was frequently positive for PEDV RNA (Niederwerder et al., 2016) . Aerosolized PEDV does not infect only the intestine of pigs (Alonso et al., 2015) , but it also infects the epithelium lining the nasal cavity . reported that dendritic cells in the lamina propria of the nasal mucosa or lymphoid tissue carry and transfer PEDV to CD3 + T cells. PEDV-loaded T cells may reach the intestine via unknown specialized endothelial venules during blood circulation. The virus loaded onto T cells had the capacity to infect intestinal epithelial cells via cell-to-cell contact and transfer infection . Compared with neonatal piglets, however, higher doses of aerosolized PEDV may be required to infect weaned and older pigs (Niederwerder et al., 2016) . In the study, none of aerosol contact, 28-day-old pigs (0/5 pigs) was infected, although their nasal cavities (5/5 pigs) were positive for PEDV RNA. The severity of PEDV infection and the disease, and the transmissibility of PEDV, depends on the overall immunity and health status of the pig population and the levels of biosecurity on farms (Pensaert and Martelli, 2016) . Nevertheless, transmissibility of PEDV via direct or aerosol J o u r n a l P r e -p r o o f contact also varies dependent on the PEDV genogroup. Direct contact or aerosol transmission rates were significantly higher in pigs infected with non-S INDEL PEDV compared with S INDELinfected pigs (Gallien et al., 2018a) . Gallien et al. (2018) revealed that despite the presence of aerosolized S INDEL PEDV, none of aerosol contact pigs (0/10 pigs) was infected, whereas 10/10 aerosol contact pigs were infected by non-S INDEL PEDV (Gallien et al., 2018a) . Porcine small intestinal villous enterocytes express aminopeptidase N (APN), a 150-kDa glycosylated transmembrane protein, which was tentatively identified as the cellular receptor for PEDV (Li et al., 2007; Liu et al., 2015a; Nam and Lee, 2010) . However, porcine APN may not be the major cell surface receptor for PEDV (Ji et al., 2018; Shirato et al., 2016) . Concomitantly, APN knockout pigs were susceptible to infection with PEDV, but not with TGEV that uses APN as the cellular receptor (Whitworth et al., 2019; Zhang et al., 2019) . Cell membrane cholesterol or two cell surface molecules, such as sialic acids and occludin expressed on the apical surface of secretary (goblet) or absorptive enterocytes, respectively, were involved partially in binding and entry of PEDV into enterocytes (Deng et al., 2016; Jeon and Lee, 2017; Luo et al., 2017) . The strains of PEDV, such as cell culture-adapted vs. wild-type, may be a factor involved in the cellular entry via sialic acids (Deng et al., 2016; . Intracytoplasmic localization of PEDV antigen in goblet cells of infected pigs and porcine intestinal enteroids in vitro (Jung and Saif, 2017; raises questions on whether or how goblet cells are utilized for PEDV replication, or if they only transfer PEDV to absorptive enterocytes. J o u r n a l P r e -p r o o f We demonstrated the tissue tropism of PEDV to certain intestinal locations (Jung et al., 2018) . During acute PEDV infection [post-inoculation hour (PIH) 12-24] in nursing pigs, PEDV initially infected mainly the mid-jejunum and ileum and to a lesser extent, the proximal and distal jejunum and duodenum (Jung et al., 2018) (Fig. 1) . The pylorus was not the site of acute PEDV infection (Jung et al., 2018) , whereas the villous enterocytes of the large intestine were frequently infected; however, infected colonic enterocytes did not undergo necrosis (Jung et al., 2015a; Jung et al., 2014) . Unlike colonic enterocytes, PEDV-infected small intestinal villous enterocytes undergo acute necrosis and exfoliation from the lamina propria, leading to marked villous atrophy or fusion in the small intestine (Jung et al., 2018; Jung et al., 2014) . Like TGEV (Kim et al., 2000) and PDCoV (Jung et al., 2016) , PEDV may not induce apoptotic death of intestinal villous enterocytes in vivo (Jung and Saif, 2015) . However, PEDV-infected Vero cells in vitro underwent apoptosis (Kim and Lee, 2014) . During acute PEDV infection (PIH 12-24) of nursing pigs, early localization of PEDV antigen was evident in the villous-crypt interface of the small intestine, rather than the villous tips (Jung et al., 2018) (Fig. 1) . Iimmature enterocytes were the major site of initial PEDV infection, although the exact reason remains obscure (Jung et al., 2018) . The PEDV antigen-positive regions subsequently expanded to the upper and then the entire villous epithelium of the jejunum to ileum (<24 hours after oral inoculation) (Jung et al., 2018) . The villous-crypt interface is close to blood vessels in the submucosa. Although PEDV-related viremia might be a result of diffusion of replicated PEDV from the acutely infected intestine to blood (Jung et al., 2018) , further studies are J o u r n a l P r e -p r o o f needed to investigate whether PEDV or PEDV-loaded CD3+ T cells circulating in blood reach and infect the villous-crypt interface or other villous regions, and other types of cells of extra-intestinal origin (Jung et al., 2018; . PEDV antigens were also frequently detected in the intestinal crypt cells or antigen presenting cells, such as macrophages, in the lamina propria or Peyer's patches (Debouck et al., 1981; Jung et al., 2014; Lin et al., 2015a; Madson et al., 2016; Stevenson et al., 2013; Sueyoshi et al., 1995) . PEDV remains infectious in dendritic cells for <24 hours in vitro (Gao et al., 2015) , but dendritic cells may not be a site of PEDV replication . Instead, PEDV appeared to employ dendritic cells to cross the epithelial barrier of the nasal cavity . Lung tissues of oronasally infected pigs were negative for PEDV antigen (Debouck et al., 1981; Jung et al., 2014; Stevenson et al., 2013; Sueyoshi et al., 1995) . However, the upper respiratory tract may be the site of PEDV infection, because the epithelium lining the nasal cavity was positive for PEDV antigens . Although acute viremia was frequently noted in infected pigs during the acute or incubation stage of infection (Jung et al., 2018; Jung et al., 2014; , PEDV antigens were not detected in other major organs, such as liver and kidneys (Debouck et al., 1981; Jung et al., 2014; Stevenson et al., 2013; Sueyoshi et al., 1995) . The reproductive organs of experimentally infected boars, such as Cowpers's glands, were also negative for PEDV RNA, but their semen was transiently positive for PEDV RNA (Gallien et al., 2018b; Gallien et al., 2019) . Nevertheless, whether the PEDV RNA-positive semen contained infectious viral particles is unclear. J o u r n a l P r e -p r o o f During the incubation period (