Zhen-Zhen Li, Yu-Lin Yang, Meng-Yao Sun, Hong-Bing Shu, Li-Bo Cao
{"title":"Lumpy skin disease virus LSDV087 positively regulates innate immune response by promoting oligomerization of MITA/STING.","authors":"Zhen-Zhen Li, Yu-Lin Yang, Meng-Yao Sun, Hong-Bing Shu, Li-Bo Cao","doi":"10.1128/jvi.01026-25","DOIUrl":"https://doi.org/10.1128/jvi.01026-25","url":null,"abstract":"<p><p>Lumpy skin disease (LSD), caused by the lumpy skin disease virus (LSDV), is a contagious disease in cattle that poses a major threat to the global cattle industry. The functions of most LSDV-encoded proteins remain poorly characterized, particularly regarding their roles in regulating innate immunity. In this study, we show that the LSDV-encoded protein LSDV087 positively regulates innate immune response independently of its decapping enzymatic activity. LSDV087 interacts with the adaptor protein MITA (also called STING) in the innate immune pathway, inhibits its degradation by reducing K48-linked polyubiquitination, and promotes its oligomerization and subsequent activation of downstream signaling events, leading to enhanced innate immune response. Consistently, LSDV087-deficient virus (LSDV∆087) exhibits an attenuated ability to activate cGAS-MITA-mediated innate immune response. Collectively, our study reveals regulatory mechanisms of LSDV-triggered innate immune response and points to the possibility of targeting LSDV087 for rational design of live-attenuated LSDV vaccines.IMPORTANCELumpy skin disease virus (LSDV), which causes a contagious disease in cattle, poses a significant threat to the global cattle industry. Despite its impact, the functions of most LSDV-encoded proteins remain poorly understood. In this study, we report that LSDV087 plays dual roles in both promoting the cGAS-MITA-mediated innate immune response and downregulating host gene transcription. LSDV087 interacts with the adaptor protein MITA in the innate immune pathway, inhibits its degradation by reducing K48-linked polyubiquitination, and promotes its oligomerization, leading to the subsequent activation of downstream signaling events and an enhanced innate immune response. Additionally, as an immediate-early protein, LSDV087 functions as a decapping enzyme, preferentially targeting host transcripts with multiple exons to facilitate viral replication. This dual functionality underscores the complex interplay between LSDV immune evasion strategies and host defense mechanisms and may inform the rational design of live-attenuated LSDV vaccines.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0102625"},"PeriodicalIF":3.8,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251605","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":"Ribosomal protein L35 negatively regulates FMDV replication by recruiting AMFR to promote the ubiquitination and degradation of VP2.","authors":"Wenhua Shao, Wei Zhang, Yang Yang, Xiaoyi Zhao, Weijun Cao, Chuangwei Chen, Wei Wang, Mengyao Huang, Tingting Zhou, Zixiang Zhu, Fan Yang, Haixue Zheng","doi":"10.1128/jvi.01453-25","DOIUrl":"https://doi.org/10.1128/jvi.01453-25","url":null,"abstract":"<p><p>The control of foot-and-mouth disease virus (FMDV) primarily relies on vaccine immunization; however, this approach is not always fully effective, underscoring the urgent need for novel antiviral strategies. This study identifies RPL35 as a host antiviral protein that targets FMDV. Further mechanistic investigations demonstrate that RPL35 directly interacts with the FMDV structural protein VP2, mediating its K48-linked polyubiquitination and subsequent degradation. The Lys217 residue of VP2 is critical for RPL35's antiviral activity, as evidenced by the increased viral virulence observed with the rO-VP2K217R mutant virus. Through an unbiased proteomic screen, we revealed that RPL35 recruits the E3 ligase AMFR to ubiquitinate and degrade VP2. Additionally, FMDV induces the degradation of KPNA3, thereby blocking RPL35's nuclear translocation. This study advances our understanding of host-virus interactions and provides new insights into developing antiviral drugs targeting the ubiquitin-proteasome pathway.IMPORTANCEThis investigation elucidated the antiviral role of RPL35 in the context of FMDV infection. Our results indicate that RPL35 facilitates the recruitment of AMFR, which, in turn, promotes K48-linked polyubiquitination and subsequent proteasomal degradation of the viral protein VP2. This process thereby mitigates viral infection. Further analysis identified Lys217 of VP2 as a critical ubiquitination site for RPL35, with the inhibitory effect of RPL35 being abolished in the recombinant mutant virus rO-VP2K217R. Additionally, we found that FMDV induces the degradation of KPNA3, which obstructs the nuclear translocation of RPL35. Collectively, these findings suggest that RPL35 functions as a potent antiviral effector in suppressing FMDV infection.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0145325"},"PeriodicalIF":3.8,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251639","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}
Danh C Lai, The N Nguyen, Giao P Trinh, David Steffen, Hiep L X Vu
{"title":"Lipid nanoparticle-encapsulated DNA vaccine induces balanced antibody and T-cell responses in pigs with maternally derived antibodies.","authors":"Danh C Lai, The N Nguyen, Giao P Trinh, David Steffen, Hiep L X Vu","doi":"10.1128/jvi.01123-25","DOIUrl":"https://doi.org/10.1128/jvi.01123-25","url":null,"abstract":"<p><p>Maternally derived antibodies (MDAs) play a vital role in protecting neonates from infectious diseases, but their presence at the time of vaccination can interfere with vaccine-induced immune responses, thereby reducing vaccine effectiveness. MDA interference has been documented in pigs vaccinated with whole-inactivated virus (WIV) vaccines against swine influenza A virus (IAV). This study evaluated the efficacy of a lipid nanoparticle-encapsulated DNA (LNP-DNA) vaccine against swine IAV in the presence and absence of MDAs, comparing it to a WIV vaccine. In MDA-negative piglets, both the LNP-DNA and WIV vaccines induced strong immune responses and effectively prevented the vaccinated animals from being infected with the homologous IAV strain. However, in MDA-positive piglets, the WIV vaccine failed to trigger significant antibody or T-cell responses and offered no protection against viral shedding or lung damage. In contrast, the LNP-DNA vaccine elicited stronger immune responses in MDA-positive pigs, reduced nasal viral shedding, and prevented lung lesions. These findings demonstrate that the LNP-DNA vaccine overcomes MDA interference, making it a promising strategy for enhancing vaccine efficacy in neonatal animals with maternal antibodies.</p><p><strong>Importance: </strong>Maternally derived antibody (MDA) interference is a major obstacle to developing effective vaccines for neonates. In pigs, MDAs significantly impair immune responses to a whole-inactivated virus vaccine. Here, we show that vaccination with a lipid nanoparticle (LNP)-encapsulated DNA vaccine can partially overcome MDA interference. These findings underscore the potential of the LNP-DNA vaccine as a viable strategy for effectively immunizing MDA-positive populations. Additionally, LNP-DNA vaccination in young pigs provides a valuable model for exploring the immunological mechanisms behind MDA-mediated suppression of vaccine-induced immunity.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0112325"},"PeriodicalIF":3.8,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251610","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}
Bruno Milhomem Pilati Rodrigues, Luis Janssen, Leonardo Assis da Silva, Suzane Suliane Vitorino Gomes Acacio, Mariana Tigano Magalhães, Bergmann Morais Ribeiro
{"title":"Erratum for Milhomem Pilati Rodrigues et al., \"Experimental and evolutionary evidence for horizontal transfer of an envelope fusion protein gene between thogotoviruses and baculoviruses\".","authors":"Bruno Milhomem Pilati Rodrigues, Luis Janssen, Leonardo Assis da Silva, Suzane Suliane Vitorino Gomes Acacio, Mariana Tigano Magalhães, Bergmann Morais Ribeiro","doi":"10.1128/jvi.01581-25","DOIUrl":"https://doi.org/10.1128/jvi.01581-25","url":null,"abstract":"","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0158125"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251626","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}
Jonathan K Mitchell, Vincent Mastrodomenico, Jim Hartnett, William J Heelan, Denise Garvin, Mei Cong, Jamison J Grailer
{"title":"A HiBiT-tagged pseudovirus-like particle platform for safe, rapid quantification of virus neutralization and antibody-dependent enhancement.","authors":"Jonathan K Mitchell, Vincent Mastrodomenico, Jim Hartnett, William J Heelan, Denise Garvin, Mei Cong, Jamison J Grailer","doi":"10.1128/jvi.00991-25","DOIUrl":"https://doi.org/10.1128/jvi.00991-25","url":null,"abstract":"<p><p>Accurate quantification of virus neutralization is essential for evaluating the efficacy of vaccines and antibody-based therapies. However, conventional neutralization assays require strict biocontainment or utilize surrogates of infection that increase biosafety but reduce biological relevance. These limitations create critical bottlenecks for the timely development of antiviral immunotherapeutics. Here, we describe a neutralization platform using non-replicating, HiBiT-tagged pseudovirus-like particles (HiBiT-PsVLPs) for safe yet biologically relevant assessment of neutralization. These HiBiT-PsVLPs package the HiBiT protein tag internally and incorporate glycoproteins from pathogenic viruses to recapitulate entry at reduced biosafety levels. Unlike traditional pseudoviruses, HiBiT-PsVLPs lack reporter genes and employ the NanoBiT split luciferase system for rapid, complementation-based luminescent readout of entry and neutralization. Using the SARS-CoV-2 Spike protein as proof-of-concept, we demonstrate efficient pseudotyping, entry, and neutralization of HiBiT-PsVLPs. HiBiT-PsVLP neutralization is specific, reproducible, and reflective of antibody potency and stability. Assay results align closely with an established surrogate virus neutralization test (sVNT), with HiBiT-PsVLPs capturing more diverse mechanisms-of-action (MoAs). Moving beyond SARS-CoV-2, we adapt the HiBiT-PsVLP platform for other clinically relevant viruses, including HIV-1 and emerging pathogens, such as Ebola, Marburg, Lassa, and Nipah viruses. By pairing HiBiT-PsVLPs with Fc gamma receptor (FcγR)-expressing cells, we also demonstrate the capability of this system to detect antibody-dependent enhancement (ADE) of virus entry as an important safety consideration for immunotherapies. Combined, these data establish the utility of the HiBiT-PsVLP platform for safely and rapidly measuring critical antibody activities across diverse viruses and drug development stages.</p><p><strong>Importance: </strong>Standard neutralization assays are often slow, labor-intensive, and restricted to high-containment facilities, thus complicating and delaying the development of vaccines and antibody-based treatments. Here, we present a novel neutralization assay system using HiBiT-tagged pseudovirus-like particles (HiBiT-PsVLPs). These particles incorporate entry proteins from diverse pathogenic viruses but are non-replicating and lack viral nucleic acids, thus mitigating the biosafety risks of conventional assays. The particles encapsulate the HiBiT peptide, enabling rapid, luminescent quantitation of entry and neutralization. We demonstrate that this platform accurately measures neutralizing activity of monoclonal antibodies across development stages and sensitively detects antibody-dependent enhancement, a critical safety consideration. Altogether, HiBiT-PsVLPs offer a safe, rapid, and scalable platform to accelerate the development of vaccines and antibody therapeutics targeting a broa","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0099125"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251643","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}
Samuel Medica, Nicole L Diggins, Michael Denton, Rebekah L Turner, Lydia J Pung, Adam T Mayo, Olivia Kramer-Hansen, Jennifer Mitchell, Luke Slind, Linh K Nguyen, Teresa A Beechwood, Gauthami Sulgey, Craig N Kreklywich, Daniel Malouli, Mette M Rosenkilde, Patrizia Caposio, Daniel N Streblow, Meaghan H Hancock
{"title":"Human cytomegalovirus UL78 is a nuclear-localized GPCR necessary for efficient reactivation from latent infection in CD34<sup>+</sup> hematopoietic progenitor cells.","authors":"Samuel Medica, Nicole L Diggins, Michael Denton, Rebekah L Turner, Lydia J Pung, Adam T Mayo, Olivia Kramer-Hansen, Jennifer Mitchell, Luke Slind, Linh K Nguyen, Teresa A Beechwood, Gauthami Sulgey, Craig N Kreklywich, Daniel Malouli, Mette M Rosenkilde, Patrizia Caposio, Daniel N Streblow, Meaghan H Hancock","doi":"10.1128/jvi.01402-25","DOIUrl":"https://doi.org/10.1128/jvi.01402-25","url":null,"abstract":"<p><p>Human cytomegalovirus (HCMV) is a ubiquitous pathogen that persists throughout the lifetime of the host due to the establishment of latency. HCMV encodes four putative G protein-coupled receptors (GPCRs): US27, US28, UL33, and UL78. A definitive role for UL78 in HCMV infection has yet to be elucidated. Utilizing an <i>in vitro</i> CD34<sup>+</sup> hematopoietic progenitor cell (HPC) model, we demonstrate that a recombinant virus lacking UL78 protein expression fails to efficiently reactivate from latent infection. Furthermore, we show that UL78 preferentially couples to the Gα<sub>i</sub> family of G proteins and that a recombinant HCMV containing mutations in the UL78 G protein-coupling DRL motif also fails to reactivate from latent infection. Together, our findings indicate that Gα<sub>i</sub> coupling is important for UL78 function during reactivation in latently infected CD34<sup>+</sup> HPCs. To better understand the role of UL78, we conducted proteomic analyses in HCMV-UL78-TurboID-infected fibroblasts and CD34<sup>+</sup> HPCs undergoing reactivation from latency. Congruent with our coupling data, we found that Gα<sub>i</sub> was the only heterotrimeric Gα protein in proximity to UL78. Pathway analysis of the UL78 interactome revealed that proteins associated with membrane trafficking, signaling, and the nuclear pore complex were enriched in both cell types. In addition, the UL78 interactome contained viral proteins with nuclear localization including viral transcription and DNA replication machinery. Nuclear localization of UL78 was validated using cell fractionation, immunofluorescence microscopy, and proteomic analysis of isolated nuclei. Together, our results provide novel insights into the localization and function of UL78, previously unknown to contribute to reactivation from latent infection.</p><p><strong>Importance: </strong>Human cytomegalovirus (HCMV) remains one of the most widespread viral infections globally. Primary HCMV infection is typically asymptomatic and leads to the establishment of latency in myeloid lineage cells, where the virus persists for the host's lifetime. Reactivation of latent HCMV can cause severe complications, particularly in immunocompromised individuals, such as transplant recipients and people living with HIV. Several factors influence the transition from latent to lytic infection, including signal transduction through the viral G protein-coupled receptors: US27, US28, UL33, and UL78. Using an advanced <i>in vitro</i> model, we show that recombinant viruses lacking UL78 fail to efficiently reactivate from latent infection. Moreover, we show that UL78 preferentially couples to the Gα<sub>i</sub> family of G proteins via a conserved DRL motif, and this coupling is required for efficient reactivation. These results were confirmed by proximity-dependent labeling experiments, where we identified Gα<sub>i</sub> and several other proteins involved in trafficking, signaling, transcription, and nuclear l","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0140225"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251674","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}
W Zaib, C Kaviani, X Kang, Y Gao, F Vanden Broucke, W Van den Broeck, S Coppens, S Theuns, H Nauwynck, K Laval
{"title":"Porcine hemagglutinating encephalomyelitis virus VW572 (not Gent/PS412 and Labadie) uses the CD81 receptor and MVB-derived exosomal pathway for efficient entry and spread in neuronal cells.","authors":"W Zaib, C Kaviani, X Kang, Y Gao, F Vanden Broucke, W Van den Broeck, S Coppens, S Theuns, H Nauwynck, K Laval","doi":"10.1128/jvi.01171-25","DOIUrl":"https://doi.org/10.1128/jvi.01171-25","url":null,"abstract":"<p><p>Porcine hemagglutinating encephalomyelitis virus (PHEV) is considered a neurotropic coronavirus that invades the peripheral (PNS) and central (CNS) nervous system of the pig and causes acute encephalomyelitis, also known as \"vomiting and wasting disease.\" Recently, PHEV has been proposed as a potential surrogate virus model to further elucidate the neuropathogenesis of other betacoronaviruses. In this study, we compared key steps in the replication cycle of three distinct PHEV isolates (VW572, Gent/PS412, and Labadie) in mouse neuronal (N2a) cells. We found that PHEV-VW572 replicates more efficiently in these cells compared to the other two isolates. Interestingly, PHEV-VW572 showed high intracellular virus titers without efficient extracellular release. Further investigation revealed that PHEV-VW572, but not PHEV-Gent/PS412, mainly uses multivesicular body (MVB)-derived exosomes for viral egress. Transmission electron microscopy confirmed the presence of complete PHEV-VW572 virions within intracellular vesicles and the release of fused PHEV-exosome structures near the plasma membrane. Finally, we showed that PHEV binding is restricted for all isolates. Still, we demonstrated that only PHEV-VW572 entry into cells is mediated by the tetraspanin CD81 receptor. Overall, these results suggest that PHEV-VW572 uses the MVB-derived exosomal pathway as a strategy to promote efficient infection and overcome the early restriction in neuronal cells. In addition, these findings highlight isolate-specific differences in PHEV neurotropism.IMPORTANCEThe neuropathogenesis of betacoronaviruses remains largely unclear despite the global impact of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. While these viruses are primarily known for their respiratory effects, mounting evidence suggests they can also cause significant neurological complications, ranging from mild symptoms such as headaches to severe outcomes, such as encephalitis and neurological diseases. The exact mechanisms by which coronaviruses affect the nervous system are still not fully understood, which hampers the development of adequate treatments and prevention strategies for these neurological disorders. In this study, we used the porcine hemagglutinating encephalomyelitis virus (PHEV) as a surrogate model for SARS-CoV-2 to further unravel the neuropathogenesis of betacoronaviruses.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0117125"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251660","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}
Theresa K Bessey, Yuhuan Wang, Sung-Sil Moon, Liliana Sanchez-Tacuba, Philippe H Jaïs, Harry B Greenberg, Baoming Jiang
{"title":"Mutations of two amino acids in VP5 mediate the attenuation of human rotavirus vaccine: evidence from <i>in vitro</i> and <i>in vivo</i> studies.","authors":"Theresa K Bessey, Yuhuan Wang, Sung-Sil Moon, Liliana Sanchez-Tacuba, Philippe H Jaïs, Harry B Greenberg, Baoming Jiang","doi":"10.1128/jvi.01067-25","DOIUrl":"https://doi.org/10.1128/jvi.01067-25","url":null,"abstract":"<p><p>Various vaccines, like polio, measles, and rotavirus, have been developed by serial-passaging in cell culture. Live oral rotavirus vaccines have been shown to be generally safe, but mechanisms of attenuation are not known. We have used a new, entirely plasmid-based reverse genetics system to artificially generate the novel human rotavirus vaccine strain CDC-9 (G1P[8]) and analyze the effect of the mutations within the VP4 gene on adaptation <i>in vitro</i> and attenuation <i>in vivo</i>. We demonstrated that out of the 6 amino acid mutations that appeared after serial passaging in Vero cells, mutations of wild-type CDC-9 P11 at VP4 AA331 and AA385 each or in combination were associated with increased replication <i>in vitro</i> comparable to cell-culture adapted CDC-9 P45. Neonatal rats infected with the single AA331 or AA385 mutant had reduced viral shedding, comparable to cell-culture passaged CDC-9 P45. We observed additional reduced shedding in neonatal rats that were infected with combination mutants harboring mutations at position AA331_385_388, indicative of a slight additive effect. Our data indicate that mutations in the VP5* region of the VP4 gene, particularly at position AA331 and AA385, are the determining factor for <i>in vitro</i> replication adaptation and <i>in vivo</i> attenuation of a G1P[8] rotavirus vaccine. This information provides great potential for targeted mutation in rotavirus vaccine generation instead of labor-consuming serial passaging in cell culture.IMPORTANCELive oral rotavirus vaccines have been developed through serial passaging in cell culture and found to be generally safe and efficacious in children. Live vaccines are also found to be associated with rare but severe adverse events, such as intussusception, in vaccinated children. Mechanisms for vaccine attenuation and adverse effects are unknown. We have developed a novel human rotavirus vaccine strain (CDC-9) and demonstrated several amino acid mutations in the VP4 gene of cell-passaged virus. In the present study, we identified two key amino acid mutations via reverse genetics technology in VP4 that mediated enhanced growth in cell culture, including a human intestinal cell line, reduced virus shedding, and downregulated inflammatory response in neonatal rats. This study is the first to identify the molecular signatures that define attenuation of human rotavirus vaccine and should help provide guidance for developing new generations of safe and effective vaccines.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0106725"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251613","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}
Jason K Perry, Samuel Itskanov, John P Bilello, Eric B Lansdon
{"title":"A structural roadmap for the formation of the coronavirus nsp3/nsp4 double membrane vesicle pore and its implications for polyprotein processing and replication/transcription.","authors":"Jason K Perry, Samuel Itskanov, John P Bilello, Eric B Lansdon","doi":"10.1128/jvi.01457-25","DOIUrl":"https://doi.org/10.1128/jvi.01457-25","url":null,"abstract":"<p><p>Coronavirus replication is understood to occur within double membrane vesicles (DMVs) that arise during viral infection. Prior work has determined that these DMVs have characteristic pores formed from the non-structural viral proteins nsp3 and nsp4, which facilitate export of newly synthesized viral RNA. Yet how the replication machinery, which is comprised of the non-structural proteins nsp7 to nsp16, is recruited to the DMV remains a mystery. Working from AlphaFold and previously determined structures, we constructed a series of models that link formation of the DMV pore to nsp5 protease processing of the polyprotein and trapping of the cleaved products within the DMV itself. We argue that the initial pore is formed from 12 subunits of nsp3 and six subunits each of the intermediate uncleaved polyproteins pp1a' (nsp4-nsp10) and pp1ab' (nsp4-nsp16). Formation of this initial structure activates the protease function of alternating nsp5 subunits within a close-packed ring, facilitating the initial <i>trans-</i>cleavage of the nsp4-nsp5 linkage. Maturation of the pore follows, as does formation of canonical nsp5 dimers, which can process the remainder of the polyproteins. When protease activation occurs subsequent to closure of the DMV, the cleavage products, whose stoichiometry is consistent with the previously proposed nsp15-centered hexameric replication complex, will be trapped inside.IMPORTANCECoronaviruses, like many other positive-sense single-stranded RNA viruses, rely on intracellular membrane remodeling to create a protected environment for efficient replication to occur. The resulting double membrane vesicles (DMVs) have characteristic pores formed from the non-structural viral proteins nsp3 and nsp4, while enzymes responsible for RNA replication, including the polymerase nsp12, are contained inside. Recent structural work has elucidated the nature of the pores, but how the polymerase and other viral proteins are recruited to the DMV represents a major gap in our current knowledge. Here we present a novel, step-by-step structural model of how the pores initially form from the uncleaved polyprotein, how protease activity is initiated, and how the viral replication machinery is trapped within the DMV.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0145725"},"PeriodicalIF":3.8,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145251603","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":"Single-cell RNA sequencing highlights the role of epithelial-immune dual features of proximal tubule cells in BK polyomavirus nephropathy.","authors":"Feng Yang, Xutao Chen, Hui Zhang, Shicong Yang, Huifei Yang, Peisong Chen, Guodong Zhao, Yingzhen He, Siyan Meng, Dongfeng Yin, Qian Li, Jiang Qiu, Gang Huang","doi":"10.1128/jvi.01394-25","DOIUrl":"https://doi.org/10.1128/jvi.01394-25","url":null,"abstract":"<p><p>Up to 10% of renal allograft failures are caused by BK polyomavirus nephropathy (BKPyVN). However, there is no specific antiviral agent for BKPyVN. The only treatment is to reduce the levels of immunosuppression, which is not always practical and increases the risk of rejection. Since targeting the microenvironment is a promising approach, we performed single-cell RNA sequencing (scRNA-seq) on BKPyVN samples and stable allografts to obtain BKPyVN microenvironmental atlases. Interestingly, we identified a novel subpopulation of proximal tubule cells (annotated as IGKC+ PT) with epithelial-immune dual features that may contribute to the progression of BKPyVN through T-cell exhaustion. Additionally, we determined that the IGKC+ PT subpopulation might serve as a non-invasive diagnostic marker through scRNA-seq of urine samples and co-immunofluorescence staining. These results improve our understanding of the BKPyVN microenvironment and may guide the development of new therapeutic and diagnostic approaches for a wide range of patients.IMPORTANCEBKPyVN severely threatens kidney transplant recipients. Due to the lack of effective drugs against BK polyomavirus (BKPyV), reducing immunosuppressant therapy is the only treatment. Unfortunately, this approach is not always effective and increases the acute rejection risk. A growing body of research suggests that potential therapeutic targets may be identified by studying the disease microenvironment. However, traditional methods have not explained why the large number of infiltrating T cells in the BKPyVN microenvironment does not effectively clear BKPyV. Newly available large-scale scRNA-seq technology can be used to study gene expression at a single-cell resolution, offering a new way to investigate the BKPyVN microenvironment. By combining scRNA-seq with experimental analysis, we found a novel subpopulation of proximal tubule cells (annotated as IGKC+ PT) with epithelial-immune dual features that may contribute to the progression of BKPyVN through T-cell exhaustion.</p>","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":" ","pages":"e0139425"},"PeriodicalIF":3.8,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145238867","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}