{"title":"A veterinary virapalooza: a summary of the 2024 American Society for Virology (ASV) Veterinary/Zoonotic Virology Satellite Symposium and online H5N1 panel discussion.","authors":"Andrew Broadbent","doi":"10.1128/jvi.00499-25","DOIUrl":"https://doi.org/10.1128/jvi.00499-25","url":null,"abstract":"<p><p>The year 2024 saw veterinary/zoonotic virology take center stage once more as the American Society for Virology (ASV) hosted a satellite symposium on the subject in June and an online panel discussion in December. The symposium comprised six talks from experts on viruses of economic importance to agriculture and of public health importance. The viruses in question spanned foot and mouth disease virus (FMDV), African swine fever virus (ASFV), Marek's disease virus (MDV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and influenza A viruses (IAVs), and topics covered fundamental virology, applied virology, epidemiology, and surveillance. The goal was to emphasize that improving the control of animal viral diseases requires an integrated, holistic approach involving academia, government, and industry labs undertaking research on basic virology, vaccinology, epidemiology, and surveillance. Moreover, the symposium aimed to highlight career opportunities in the agricultural/veterinary sector for those with virology training. Six months following the symposium, the ASV held an online panel discussion on the ongoing H5N1 IAV situation in poultry, cattle, and people to provide more up-to-date information to its membership. A summary of the talks and discussions is presented here.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0049925"},"PeriodicalIF":4.0,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144063893","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Marios Koutsakos, Arnold Reynaldi, Malet Aban, Ian G Barr, David S Khoury, Miles P Davenport, Ali H Ellebedy, Philip A Mudd
{"title":"Binding antibody titers against the hemagglutinin and neuraminidase correlate with protection against medically attended influenza A and B disease.","authors":"Marios Koutsakos, Arnold Reynaldi, Malet Aban, Ian G Barr, David S Khoury, Miles P Davenport, Ali H Ellebedy, Philip A Mudd","doi":"10.1128/jvi.00391-25","DOIUrl":"https://doi.org/10.1128/jvi.00391-25","url":null,"abstract":"<p><p>Human challenge and cohort studies have identified various correlates of protection (CoP) against influenza A and B viruses (IAV/IBV). However, associations with viral load, investigation of mucosal CoPs, and CoPs against IBV are limited in the context of natural infections. Plasma and nasal swabs were collected (2017-2020) from 56 adults diagnosed with IAV (<i>n</i> = 25 H1N1, <i>n</i> = 19 H3N2) or IBV (<i>n</i> = 9 B/Victoria, <i>n</i> = 3 B/Yamagata) in the emergency department. Viral load was determined in nasal swabs. Antibodies (total Ig and IgA) specific for the hemagglutinin (HA) and neuraminidase (NA) of contemporary viruses from the subtype or lineage infecting each individual were measured by enzyme-linked immunosorbent assay (ELISA). Antibodies to a non-infecting influenza strain were measured and used as \"control cases\" to determine associations with protection from infection. Viral load decreased with time post-symptom onset. The Ct value at which 50% of the samples were positive in viral culture was 24.75 (95% confidence intervals, 23.7-27.01). Systemic HA and NA-specific Ig titers correlated with protection from medically-attended influenza disease. Neither systemic nor mucosal antibody measurements were associated with disease severity. We observed an inverse correlation between Ig anti-NA antibodies in nasal swabs and viral load by Ct value (regression coefficient = 3.25, CI = 0.3-6.2, <i>P</i> = 0.031), though this analysis was not corrected for multiple comparisons. Overall, high titers of HA and NA-specific antibodies measured by ELISA were associated with protection from the development of influenza A or B disease. Further work is needed to understand immune parameters associated with viral clearance and mucosal CoPs.IMPORTANCEThere is a great need to better understand correlates of protection (CoP) against influenza A and B viruses (IAV/IBV). In our study, we analyzed paired plasma and nasal swabs from patients presenting with influenza A or B disease as well as control patients. We measured hemagglutinin (HA) and neuraminidase (NA) specific antibodies in both sample types and also determined the amount of virus in nasal swabs. We found that higher systemic binding antibodies to the hemagglutinin and neuraminidase were associated with protection from medically attended disease. These findings expand our understanding of correlates of protection against influenza viruses and identify areas of future research to further understand protection from influenza.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0039125"},"PeriodicalIF":4.0,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144031955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Typhaine Filhol, Alice Mac Kain, Marie-Line Joffret, Nolwenn Jouvenet, Vincent Caval, Maël Bessaud
{"title":"The use of sialic acids as attachment factors is a common feature of <i>Enterovirus</i>-D species.","authors":"Typhaine Filhol, Alice Mac Kain, Marie-Line Joffret, Nolwenn Jouvenet, Vincent Caval, Maël Bessaud","doi":"10.1128/jvi.00429-25","DOIUrl":"https://doi.org/10.1128/jvi.00429-25","url":null,"abstract":"<p><p>Among the hundreds of enteroviruses (EVs) infecting humans, the members of the species EV-D (<i>Enterovirus deconjuncti</i>) display original traits. First, only five serotypes are known within this species, while other EV species have tens of serotypes each. Second, EV-Ds display a wide variety of tropisms: EV-D68s are respiratory viruses, EV-D70s have an ocular tropism, while EV-D94s, EV-D111s, and EV-D120s seem to be enteric viruses. Besides, while EV-D68s, EV-D70s, and EV-D94s have been detected in humans, EV-D120s were found exclusively in non-human primates, and the last virus type, EV-D111, was found in both. This and other observations have led to the hypothesis that EV-Ds could have a zoonotic origin. Previous studies have shown that EV-D68, EV-D70, and EV-D94 use sialic acids (Sias) as cellular attachment factors. We investigated the role of Sias in EV-D111 infection using sialidase treatments and loss-of-function experiments in human and simian cells. Assessing viral RNA yield by RT-qPCR analyses and infectious viral particle production by titration assays showed that the absence of Sias at the cell surface significantly slowed down EV-D111 infection kinetics without abolishing it. This suggests that Sia acts as an attachment factor. While EVs generally do not use Sias, EV-Ds seem to rely on them for optimal replication in cultured cells. Sia usage may therefore be an ancestral trait of this species. We also studied EV-B114, a simian enterovirus, and found that it does not use Sias. Our work provides new insight regarding an enterovirus that circulates in humans and exhibits unusual ecological traits.IMPORTANCEExcept for a few epidemics in the 1970s and 1980s, the impact of EV-Ds on human health remained modest until the 2010s. In 2014, EV-D68 was occasionally responsible for severe respiratory distress and fatal cases of muscular paralysis. EV-Ds have thus the ability to become pathogenic in humans, hence the importance of studying them. The recently discovered EV-D111, of which only a few isolates are available, has been detected in both human and simian samples, suggesting a potential zoonotic origin. We characterized the early steps of EV-D111 replication, with a focus on its ability to use Sias as attachment factors. We found that EV-D111, like other members of the EV-D species, but unlike most EVs, relies on Sia for optimal replication. Our work provides a better understanding of EV-D111 biology, which is essential to determine its tropism and its potential to emerge in humans.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0042925"},"PeriodicalIF":4.0,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144006377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Correction for Li et al., \"Mitochondria-mediated ferroptosis contributes to the inflammatory responses of bovine viral diarrhea virus (BVDV) <i>in vitro</i>\".","authors":"Zhijun Li, Bao Zhao, Ying Zhang, Wenqi Fan, Qinghong Xue, Xiwen Chen, Jingyu Wang, Xuefeng Qi","doi":"10.1128/jvi.00583-25","DOIUrl":"https://doi.org/10.1128/jvi.00583-25","url":null,"abstract":"","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0058325"},"PeriodicalIF":4.0,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143971197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Early short-course treatment fails to prevent disease or relapse upon discontinuance of treatment in infections in addition to COVID-19.","authors":"Arthur M Friedlander","doi":"10.1128/jvi.00381-25","DOIUrl":"https://doi.org/10.1128/jvi.00381-25","url":null,"abstract":"","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0038125"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144000104","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cellular transcription factor TFII-I represses adenovirus gene expression.","authors":"Rachel L White, Patrick Hearing","doi":"10.1128/jvi.00618-25","DOIUrl":"https://doi.org/10.1128/jvi.00618-25","url":null,"abstract":"<p><p>To successfully replicate, viruses must overcome innate cellular antiviral responses. With human adenovirus (HAdV), a key viral repressor of cellular antiviral responses is the early protein E4ORF3. E4ORF3 relocalizes numerous cellular antiviral proteins, particularly those involved in the interferon (IFN) and DNA damage response (DDR) pathways, to sequester them from viral replication sites in the nucleus. E4ORF3 also directs SUMO modification of a subset of its targets, some of which are subsequently targeted for proteasomal degradation. We previously identified TFII-I, a cellular transcription factor and DNA repair protein, as one of the proteins most highly SUMOylated by E4ORF3, as well as one of the E4ORF3 degradation targets. In this study, we characterized the effect of TFII-I knockout (KO) on HAdV replication. TFII-I KO significantly increased the infectious virus yield from infected cells, supporting the hypothesis that TFII-I acts as a restriction factor during HAdV infection. TFII-I KO also significantly increased viral genome replication, as well as both early and late gene and protein expression. Our results do not support TFII-I acting as a part of either the DDR or IFN responses during HAdV infection. Our results characterize a novel antiviral function for TFII-I against HAdV that occurs during the early stage of the viral replication cycle and highlight the importance of studying viral countermeasures to the cellular antiviral response, like E4ORF3, to better understand how cells restrict viral infection.IMPORTANCEThe cellular transcription factor TFII-I was previously shown to bind to HAdV late promoters and to E4-mutant viral genomes during replication. More recently, TFII-I was shown to be a degradation target of HAdV protein E4ORF3. Due to the long-established importance of E4ORF3 in countering cellular antiviral responses, this raised the question of whether TFII-I possesses an undiscovered antiviral role against HAdV. It was hypothesized that whether TFII-I played an antiviral role in HAdV infection, it was most likely to be as a repressor of the late transcriptional program. Here, we show the first direct evidence of TFII-I repressing HAdV infection and demonstrate that the inhibitory effect can be detected much earlier in the viral life cycle than previously predicted. Our findings provide insight into the role of TFII-I in the cellular antiviral response.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0061825"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144002740","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Determinants of susceptibility to SARS-CoV-2 infection in murine ACE2.","authors":"Takashi Kondo, Rigel Suzuki, Hisano Yajima, Sachiho Kawahara, Kodai Yamaya, Takaya Ichikawa, Shuhei Tsujino, Saori Suzuki, Tomokazu Tamura, Takao Hashiguchi, Takasuke Fukuhara","doi":"10.1128/jvi.00543-25","DOIUrl":"https://doi.org/10.1128/jvi.00543-25","url":null,"abstract":"<p><p>Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes angiotensin-converting enzyme 2 (ACE2) as a receptor to enter host cells, and primary receptor recognition of the spike protein is a major determinant of the host range of SARS-CoV-2. Since the emergence of SARS-CoV-2, a considerable number of variants have emerged. However, the determinants of host tropism of SARS-CoV-2 remain elusive. We conducted infection assays with chimeric recombinant SARS-CoV-2 carrying the spike protein from 10 viral variants, assessing their entry efficiency using mammalian ACE2 orthologs from species that have close contact with humans. We found that only murine ACE2 exhibited different susceptibilities to infection with the SARS-CoV-2 variants. Moreover, we revealed that the mutation N501Y in the viral spike protein has a crucial role in determining the infectivity of cells expressing murine ACE2 and of mice <i>in vivo</i>. Next, we identified six amino acid substitutions at 24, 30, 31, 82, 83, and 353 in murine ACE2 that allowed for viral entry of the variants to which murine ACE2 was previously resistant. Furthermore, we showed that ACE2 from a species closely related to mice, <i>Mus caroli</i>, is capable of supporting entry of the viral variants that could not use murine ACE2. These results suggest that few ACE2 orthologs have different susceptibility to infection with SARS-CoV-2 variants as observed for murine ACE2. Collectively, our study reveals critical amino acids in ACE2 and the SARS-CoV-2 spike protein that are involved in the host tropism of SARS-CoV-2, shedding light on interspecies susceptibility to infection.IMPORTANCESARS-CoV-2 can infect many species besides humans, leading to the evolution of the virus and adaptation to other animal hosts, which could trigger a new COVID-19 wave. The SARS-CoV-2 spike protein utilizes ACE2 as a receptor for entry into host cells. The interaction of ACE2 with the spike protein determines the host range of SARS-CoV-2. In this study, using chimeric viruses carrying the spike protein of SARS-CoV-2 variants to infect cells expressing different ACE2 orthologs from species humans come in close contact with, we confirmed murine ACE2 alone showed different susceptibility to the variants. We identified residues in murine ACE2 and the viral spike that restrict viral entry. Furthermore, an ACE2 ortholog from a species genetically close to mice mediated entry of SARS-CoV-2 variants incapable of infecting mice. This research highlights the uniquely limited susceptibility of mice to different SARS-CoV-2 variants and provides invaluable insights into the host tropism of SARS-CoV-2.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0054325"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144006375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shaohui Wang, Ujjaldeep Jaggi, Jay J Oh, Homayon Ghiasi
{"title":"IFNβ absence compensates for LAT functions in latency reactivation and T cell exhaustion.","authors":"Shaohui Wang, Ujjaldeep Jaggi, Jay J Oh, Homayon Ghiasi","doi":"10.1128/jvi.00374-25","DOIUrl":"https://doi.org/10.1128/jvi.00374-25","url":null,"abstract":"<p><p>Type I interferons (IFNs) generate strong antiviral immunity following viral infection in mice and humans. These type I IFNs are encoded by at least 14 IFNα genes and a single IFNβ gene. We showed that the absence of one of the IFNα genes (IFNα2A<sup>-/-</sup>) affected latency levels in herpes simplex virus type 1 (HSV-1) ocularly infected mice but not in infected control mice, and the absence of IFNα2A did not affect viral reactivation in ocularly infected mice. Since the role of IFNβ in HSV-1 latency reactivation and the potential effect of latency-associated transcript (LAT) on IFNβ activity is not known, we ocularly infected IFNβ<sup>-/-</sup> mice with different doses of LAT-plus [LAT(+)] and LAT-minus [LAT(-)] viruses. Wild-type (WT) control mice and IFNβ<sup>-/-</sup> mice were infected similarly. Virus titers in the eye, viral and cellular transcripts in the eye and trigeminal ganglia (TG) of infected mice on days 3 and 5 post-infection, eye disease, survival, latency-reactivation levels, and T cell exhaustion were measured in latently infected mice. Virus replication and viral and cellular transcripts in the eye of infected IFNβ<sup>-/-</sup> mice were similar to those in WT mice, while eye disease and survival in IFNβ<sup>-/-</sup> mice differed significantly from WT mice. WT mice infected with LAT(-) virus showed reduced latency, slower reactivation, and less T cell exhaustion than mice infected with LAT(+) virus. However, using different doses of each virus, latency levels, time of reactivation, and T cell exhaustion were similar between LAT(+) and LAT(-) viruses. These results suggest that the absence of IFNβ expression compensates for the function of LAT with regard to levels of latency, T cell exhaustion, and reactivation but does not affect viral and cellular transcripts during primary infection.IMPORTANCEInterferon β (IFNβ) is a type I interferon that plays an important role in controlling primary herpes simplex virus type 1 (HSV-1) infection. To evaluate the importance of IFNβ on HSV-1 latency reactivation and its relationship to LAT, we infected IFNβ<sup>-/-</sup> mice with LAT(+) and LAT(-) viruses. In the absence of IFNβ, latency levels in mice infected with LAT(-) virus were similar to those of mice infected with LAT(+) virus. The absence of IFNβ also reduced the time of reactivation in mice infected with LAT(-) virus to that of LAT(+) virus. Our results show a strong correlation between the functions of LAT and IFNβ during latent but not primary stages of HSV-1 infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0037425"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144016958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Rescue of naïve porcine circovirus type 3 and its pathogenesis in CD pigs.","authors":"Baoge Zhang, Jinshuang Cai, Chenguang Zhu, Yuyu Zhang, Jiaqiang Wu, Yufeng Li","doi":"10.1128/jvi.00341-25","DOIUrl":"https://doi.org/10.1128/jvi.00341-25","url":null,"abstract":"<p><p>The pathogenesis of naïve porcine circovirus type 3 (PCV3) in pigs is not accurately elucidated due to virus isolation failure <i>in vitro</i>. In this study, naïve PCV3 was successfully rescued and used to infect cesarean-derived (CD) pigs. The PCV3 genome was synthesized, cloned, and inserted into the pBluescript SK vector. PK-15 cells transfected with the recombinant plasmid pSK-PCV3 were passaged, and Cap protein expression was confirmed by immunofluorescence assay (IFA) and western blotting. Virus particles (~20 nm) were observed via transmission electron microscopy (TEM). The viral growth curve was plotted to determine the replication of the virus within the cells. Nocodazole treatment demonstrated that PCV3 replication is dependent on microtubule polymerization in the cell. Cells infected with PCV3 or PCV3-positive clinical samples (wPCV3) showed only cytoplasmic fluorescence. PCV3 can successfully infect CD pigs, resulting in persistent viremia, and increased viral loads in tissues and an antibody delay were observed. Inflammatory lesions were also observed in the lungs, lymph nodes, livers, and intestines of infected pigs. Stimulation of peripheral blood mononuclear cells (PBMCs) with virus-like particles (VLPs) containing the Cap protein significantly inhibited cell proliferation. scRNA-seq revealed a significant reduction in T helper 2 (Th2) cells and the migration of T helper 1 (Th1) cells toward the late differentiation stage following infection. In particular, the decrease in Th2 cells may indicate impaired humoral immunity, leading to delayed antibody production and immunosuppression. Our study is the first to observe PCV3 via TEM and to elucidate its immunosuppressive mechanisms in CD pigs.</p><p><strong>Importance: </strong>This study is of great significance as it successfully rescued naïve PCV3 and, for the first time, observed clear PCV3 viral particles using transmission electron microscopy. The successful infection of CD pigs deepened our understanding of its pathogenic mechanisms. The results revealed key aspects of viral replication, the impact of the virus on immune responses, and associated inflammatory lesions in various tissues. Notably, the study found that the reduction of Th2 cells leads to impaired humoral immunity and delayed antibody production, which may provide valuable insights for vaccine development and swine disease management.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0034125"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143989421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lilei Lv, Huaye Luo, Jingxuan Yi, Kang Zhang, Yanhua Li, Wu Tong, Yifeng Jiang, Yanjun Zhou, Guangzhi Tong, Changlong Liu
{"title":"IFITM proteins are key entry factors for porcine epidemic diarrhea coronavirus.","authors":"Lilei Lv, Huaye Luo, Jingxuan Yi, Kang Zhang, Yanhua Li, Wu Tong, Yifeng Jiang, Yanjun Zhou, Guangzhi Tong, Changlong Liu","doi":"10.1128/jvi.02028-24","DOIUrl":"https://doi.org/10.1128/jvi.02028-24","url":null,"abstract":"<p><p>Porcine epidemic diarrhea virus (PEDV) is a highly contagious coronavirus that poses a substantial threat to the global swine industry. However, our current understanding of the host factors crucial for PEDV infection remains limited. To identify these host factors, we conducted a genome-wide CRISPR/Cas9 gene knockout screen using a PEDV-permissive cell line. Our results indicate that the endogenous expression of human interferon-inducible transmembrane protein 3 (IFITM3) enhances PEDV entry and replication. Silencing or eliminating endogenous IFITM3 in Huh7 cells significantly suppressed PEDV entry, whereas reintroducing IFITM3 partially restored susceptibility to PEDV. Overexpression of human IFITM3 or IFITM2, but not IFITM1, in Huh7.5 cells substantially increased PEDV entry and replication. Importantly, our results suggest that human IFITM3 influences PEDV entry at a later stage. Furthermore, the overexpression of porcine IFITM1 significantly enhanced PEDV infection in LLC-PK1 cells, whereas the overexpression of porcine IFITM2/3 did not produce similar effects. Notably, removing the C-terminal 15 amino acids of porcine IFITM2/3 resulted in increased PEDV entry. Coimmunoprecipitation analyses showed that all IFITMs interacted with the PEDV S1 protein, indicating a direct role in the viral entry process. Additionally, porcine IFITM1 colocalized with the PEDV S protein at the cell nuclear periphery and enhanced PEDV infection in porcine small intestinal organoids. Overall, our results suggest that IFITMs are critical in facilitating PEDV entry into cells. Targeting IFITMs may provide a promising strategy for controlling PEDV transmission and developing interventions to mitigate the virus's impact on the swine industry.</p><p><strong>Importance: </strong>Understanding the mechanisms underlying porcine epidemic diarrhea virus (PEDV) infection is vital for addressing its significant impact on the swine industry. This study reveals that interferon-inducible transmembrane (IFITM) proteins, particularly human IFITM3 and porcine IFITM1, play crucial roles in facilitating PEDV entry and replication. By elucidating these molecular interactions, the research highlights the potential of IFITMs as therapeutic targets for managing PEDV infections and paves the way for antiviral strategies. Moreover, this research extends beyond PEDV management, underscoring the critical role of host factors in controlling the spread of pathogenic coronaviruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0202824"},"PeriodicalIF":4.0,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144042294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}