{"title":"Tradeoffs between proliferation and transmission in virus evolution- insights from evolutionary and functional analyses of SARS-CoV-2.","authors":"Jui-Hung Tai, Ding-Chin Lee, Hsin-Fu Lin, Tai-Ling Chao, Yongsen Ruan, Ya-Wen Cheng, Yu-Chi Chou, You-Yu Lin, Sui-Yuan Chang, Pei-Jer Chen, Shiou-Hwei Yeh, Hurng-Yi Wang","doi":"10.1186/s12985-025-02727-5","DOIUrl":"https://doi.org/10.1186/s12985-025-02727-5","url":null,"abstract":"<p><p>To be successful, a virus must maintain high between-host transmissibility while also effectively adapting within hosts. The impact of these potentially conflicting demands on viral genetic diversity and adaptation remains largely unexplored. These modes of adaptation can induce uncorrelated selection, bring mutations that enhance certain fitness aspects at the expense of others to high freqency, and contribute to the maintenance of genetic variation. The vast wealth of SARS-CoV-2 genetic data gathered from within and across hosts offers an unparalleled opportunity to test the above hypothesis. By analyzing a large set of SARS-CoV-2 sequences (~ 2 million) collected from early 2020 to mid-2021, we found that high frequency mutations within hosts are sometimes detrimental during between-host transmission. This highlights potential inverse selection pressures within- versus between-hosts. We also identified a group of nonsynonymous changes likely maintained by pleiotropy, as their frequencies are significantly higher than neutral expectation, yet they have never experienced clonal expansion. Analyzing one such mutation, spike M1237I, reveals that spike I1237 boosts viral assembly but reduces in vitro transmission, highlighting its pleiotropic effect. Though they make up about 2% of total changes, these types of variants represent 37% of SARS-CoV-2 genetic diversity. These mutations are notably prevalent in the Omicron variant from late 2021, hinting that pleiotropy may promote positive epistasis and new successful variants. Estimates of viral population dynamics, such as population sizes and transmission bottlenecks, assume neutrality of within-host variation. Our demonstration that these changes may affect fitness calls into question the robustness of these estimates.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"107"},"PeriodicalIF":4.0,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008902/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143985015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-19DOI: 10.1186/s12985-025-02729-3
Kylie H Van der Hoek, Tanja Jankovic-Karasoulos, Dylan McCullough, Rosa C Coldbeck-Shackley, Nicholas S Eyre, Claire T Roberts, Michael R Beard
{"title":"The first trimester human placenta responds to Zika virus infection inducing an interferon (IFN) and antiviral interferon stimulated gene (ISG) response.","authors":"Kylie H Van der Hoek, Tanja Jankovic-Karasoulos, Dylan McCullough, Rosa C Coldbeck-Shackley, Nicholas S Eyre, Claire T Roberts, Michael R Beard","doi":"10.1186/s12985-025-02729-3","DOIUrl":"https://doi.org/10.1186/s12985-025-02729-3","url":null,"abstract":"<p><strong>Background: </strong>Zika virus (ZIKV) is a positive-strand RNA virus of the Flaviviridae family. Maternal ZIKV infection during pregnancy can spread to the placenta and fetus causing severe neurological defects and infants born with microcephaly. Here, we investigated ZIKV infection and the cellular innate antiviral immune response in first trimester human placental explant cultures and isolated primary villus cytotrophoblasts (CTBs).</p><p><strong>Methods: </strong>Placentas were obtained with informed consent from women undergoing elective pregnancy termination and either cultured as placental explants or used to isolate primary CTBs. Explants and CTBs were both infected with ZIKV (PRVABC59), and samples evaluated for infection by qRT-PCR, viral plaque and ELISA assays, and immunohistochemical or immunocytochemical staining.</p><p><strong>Results: </strong>We demonstrate robust infection and production of ZIKV in placental explant and CTB cultures. Both displayed delayed upregulation of interferons (IFN), most notably IFNβ and IFNλ2/3, and a panel of interferon stimulated genes (ISG) (IFI6, IFIT1, IFIT2, IFITM1, ISG15, MX1, RSAD). Stimulation of explants and CTBs with the dsRNA mimic poly(I: C), caused immediate IFN and ISG upregulation, demonstrating the first trimester placenta is innate immune competent. This suggests that either ZIKV blocks the early innate response, or the placental response is inherently hindered.</p><p><strong>Conclusion: </strong>Together these data show that first trimester placenta is susceptible to ZIKV infection which induces a delayed type III IFN antiviral response. This delay likely creates an environment favourable to ZIKV replication and dissemination across the early gestation placenta to fetal tissue, causing pathologies associated with congenital ZIKV syndrome.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"108"},"PeriodicalIF":4.0,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008946/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144026098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-19DOI: 10.1186/s12985-025-02735-5
Sidhartha Hazari, Lizeth Taylor, Salima Haque, Robert F Garry, Sander Florman, Ronald Luftig, Frederic Regenstein, Srikanta Dash
{"title":"Retraction Note: Reduced expression of Jak-1 and Tyk-2 proteins leads to interferon resistance in hepatitis C virus replicon.","authors":"Sidhartha Hazari, Lizeth Taylor, Salima Haque, Robert F Garry, Sander Florman, Ronald Luftig, Frederic Regenstein, Srikanta Dash","doi":"10.1186/s12985-025-02735-5","DOIUrl":"https://doi.org/10.1186/s12985-025-02735-5","url":null,"abstract":"","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"110"},"PeriodicalIF":4.0,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008908/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144016094","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-19DOI: 10.1186/s12985-025-02734-6
Jinkai Li, Jianhua Mei, Jie Yu, Xiaolei Chen, Jianliang Zhu, Jiaji Ye, Deyong Zhang, Dongqing Cheng, Xiuying Chen
{"title":"Characteristics of molecular epidemiology and transmitted drug resistance among newly diagnosed HIV-1 infections in Lishui, China from 2020 to 2023.","authors":"Jinkai Li, Jianhua Mei, Jie Yu, Xiaolei Chen, Jianliang Zhu, Jiaji Ye, Deyong Zhang, Dongqing Cheng, Xiuying Chen","doi":"10.1186/s12985-025-02734-6","DOIUrl":"https://doi.org/10.1186/s12985-025-02734-6","url":null,"abstract":"<p><strong>Background: </strong>Transmitted drug resistance (TDR) is becoming an obstacle to the success of antiretroviral therapy (ART) as the HIV epidemic continues to spread. This study aimed to investigate the characteristics of TDR and the molecular epidemiology of ART-naive HIV-1 infections in Lishui.</p><p><strong>Methods: </strong>A total of 481 plasma samples were collected from ART-naive HIV-1 infections in Lishui between 2020 and 2023. The sequences acquired from infections were used to analyze the characteristics of genotype, TDR, and molecular transmission network.</p><p><strong>Results: </strong>This study discovered that the three most prevalent subtypes among the 455 sequences successfully obtained from infections in Lishui were CRF08_BC (35.8%), CRF07_BC (26.4%), and CRF01_AE (25.9%). The overall prevalence of TDR was 12.1%, and the K103N (2.4%) was the most frequent mutation. Multivariate analysis showed that CRF08_BC (OR = 5.401, P < 0.001) and CD4<sup>+</sup> cell concentration of 200-499 cells/µL (OR = 1.684, P = 0.030) were associated with a higher risk of entering the molecular transmission network and clustering, whereas the current address in other cities (OR = 0.328, P = 0.004), junior middle school (OR = 0.472, P = 0.006), and junior college or above (OR = 0.387, P = 0.045) were associated with a lower risk of clustering.</p><p><strong>Conclusions: </strong>This study revealed that the prevalence of TDR was at an intermediate level of drug resistance, and high levels of resistance were predominantly concentrated in efavirenz (EFV) and nevirapine (NVP) among the NNRTIs. Middle-aged and older infections represented a significant proportion of the molecular transmission network. This suggests that HIV surveillance and targeted prevention and treatment interventions are essential to reduce the risk of HIV transmission.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"111"},"PeriodicalIF":4.0,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008846/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144049047","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Emerging zoonotic potential of H4N1 avian influenza virus: enhanced human receptor binding and replication via novel mutations.","authors":"Xiaobin Peng, Xianxian Wen, Jing Deng, Mengyuan Yuan, Yang Liu, Yunqiu Zhou, XuBin Du, Han Yang, Guangqun Jiang, Xumei Li, Feng Yin, Ping Liu, Yun Tang, Zhangping Tan, Zhen Yu, Shuang Chen","doi":"10.1186/s12985-025-02736-4","DOIUrl":"https://doi.org/10.1186/s12985-025-02736-4","url":null,"abstract":"<p><strong>Background: </strong>Avian influenza virus (AIV), a zoonotic pathogen found worldwide, includes multiple subtypes, one of which is the H4 subtype frequently detected in wild birds and poultry. Despite its prevalence, research on H4 subtype AIV has been scarce, with a focus predominantly on the H4N2 and H4N6 subtypes. The zoonotic potential of H4N1 has not been investigated to date.</p><p><strong>Methods: </strong>In this study, we used gene sequencing in conjunction with bioinformatics methodologies to analyze wild-type H4N1 AIV strain and mutant strains emerging from serial passaging in cell culture. Furthermore, we assessed the zoonotic potential of H4N1 and the alterations caused by mutations via a series of phenotype assays, including evaluation of receptor binding affinity, immunofluorescence assays, analyses of growth kinetics across different animal cell cultures, and in vivo pathogenicity studies.</p><p><strong>Results: </strong>Our research reveals that H4N1 AIV can bind to human receptors and exhibits an affinity for human lung and tracheal tissues. In vitro experiments demonstrate that H4N1 replicates efficiently in human cell lines. Furthermore, animal studies demonstrate that H4N1 can induce pneumonia in mice without the need for prior adaptation to the host. Notably, during passage in cell culture, H4N1 rapidly acquired two previously unreported mutations. These mutations significantly enhanced the virus's ability to attach to human receptors and its capacity for replication.</p><p><strong>Conclusions: </strong>In summary, our study provides preliminary experimental evidence for the emerging zoonotic potential of H4N1 AIV. These findings expand our knowledge of the H4 subtype AIV and reinforce the critical need for continued surveillance of AIV to prevent and prepare for potential outbreaks affecting human health.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"106"},"PeriodicalIF":4.0,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008868/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-18DOI: 10.1186/s12985-025-02706-w
Jinni Chen, Yan Wang, Lingyun Liu, Hao Zhou, Pan Mao, Lingling Li, Ji Pu, Xuefang Xu, Jing Yang, Jingdong Song, Hui Sun, Xia Luo, Kui Dong, Changyun Ye
{"title":"Isolation and relationship analysis of Listeria phages with various serotype hosts and morphological characterization.","authors":"Jinni Chen, Yan Wang, Lingyun Liu, Hao Zhou, Pan Mao, Lingling Li, Ji Pu, Xuefang Xu, Jing Yang, Jingdong Song, Hui Sun, Xia Luo, Kui Dong, Changyun Ye","doi":"10.1186/s12985-025-02706-w","DOIUrl":"https://doi.org/10.1186/s12985-025-02706-w","url":null,"abstract":"<p><p>Listeriosis, caused by Listeria monocytogenes (Lm), is a severe foodborne illness with a high fatality rate. Listeria phages specifically target and lyse Lm, offer a promising alternative for biocontrol and phage therapy. However, most existing studies focus on the lytic characteristics of Listeria phages using limited sample sizes. In this study, a large number of Listeria phages were isolated from diverse sources, and their lytic profiles and morphology were characterized. A total of 317 Listeria phages were isolated from 90 food-related environmental samples and 196 natural environmental samples collected across seven provinces. The phages were tested for lytic activity against 35 Lm strains representing nine serotypes, and their morphology was characterized using transmission electron microscopy (TEM). Statistical analysis was conducted to evaluate the lytic patterns of phages. The phages were classified into three groups based on their total lysis ratios. Broad Host Range Phages (BHRP) were primarily members of the Myoviridae-like phages and demonstrated the ability to lyse a vast majority of nine serotype host strains. Medium Host Range Phages (MHRP) comprised both Siphoviridae-like and Myoviridae-like phages, and demonstrated lysis of 6-9 serotype strains. Narrow Host Range Phages (NHRP) belonged to the Siphoviridae-like phages and exhibited effective lysis of serotype 4 strains. Furthermore, phages isolated from food-related environmental sources demonstrated greater lytic activity against Listeria serotypes 1/2b, 4a, and 4c compared to those derived from natural environmental sources. The study first isolated a multitude of Listeria phages, elucidated their lytic patterns and ecological distribution, and provided a valuable resource for future research.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"104"},"PeriodicalIF":4.0,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12007378/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143998096","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-18DOI: 10.1186/s12985-025-02726-6
Caiyi Wen, Xiaohan Yang, Xianjin Wu
{"title":"HPV infection incidence and genotype distribution among male patients visiting outpatient departments in Huizhou from 2014 to 2023.","authors":"Caiyi Wen, Xiaohan Yang, Xianjin Wu","doi":"10.1186/s12985-025-02726-6","DOIUrl":"https://doi.org/10.1186/s12985-025-02726-6","url":null,"abstract":"<p><strong>Background: </strong>There have been no previous studies on male HPV infection in the Huizhou region. This research aims to investigate the HPV infection rate and genotype distribution among male patients in this area, offering valuable insights for developing targeted preventive strategies against HPV infection in male population.</p><p><strong>Methods: </strong>This study included 1009 male patients from Huizhou Central People's Hospital who underwent HPV genotype testing between 2014 and 2023. We analyzed the distribution of HPV genotypes by year, age group, and diagnosis. Additionally, clinical data from 308 HPV-positive patients were retrospectively collected, and differences in high-risk vs. low-risk types, single vs. multiple infections, and genotype correlations were analyzed.</p><p><strong>Results: </strong>The overall HPV positivity rate was 30.53%, with the positive rate(40.56%) in the 2014-2019 group being significantly higher than that in 2021 (25.56%) and 2022 (24.29%)(p<0.05). The most common genotypes were HPV6, HPV52, HPV11, and HPV16. HPV infection was most prevalent in the 41-50 age group, while males aged ≤ 30 were predominantly infected with low-risk types (41.73%). The 31-40 age group had a higher prevalence of high-risk types (52.07%), with males under 50 primarily infected with low-risk HPV6, while those aged 51 and above mostly had high-risk HPV52 infections. The highest HPV positivity rate was found in the viral wart group (79.01%). Single infections were more common (64.29%), with co-infection of HPV6 and HPV16 being the most prevalent type.</p><p><strong>Conclusion: </strong>The overall HPV infection rate was relatively high among outpatient male patients in Huizhou, with single infections being predominant. Additionally, HPV infection rates exhibited significant differences across various years, age groups, and diagnostic types, suggesting that these factors should be considered when formulating HPV prevention and control strategies.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"105"},"PeriodicalIF":4.0,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12007251/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-17DOI: 10.1186/s12985-025-02719-5
Tessa Nelemans, Ali Tas, Nina L de Beijer, George M C Janssen, Peter A van Veelen, Martijn J van Hemert, Marjolein Kikkert
{"title":"Usutu virus NS4A induces autophagy and is targeted by the selective autophagy receptor p62/SQSTM1 for degradation.","authors":"Tessa Nelemans, Ali Tas, Nina L de Beijer, George M C Janssen, Peter A van Veelen, Martijn J van Hemert, Marjolein Kikkert","doi":"10.1186/s12985-025-02719-5","DOIUrl":"https://doi.org/10.1186/s12985-025-02719-5","url":null,"abstract":"<p><p>Usutu virus (USUV) is an emerging orthoflavivirus, which mainly affects birds but in rare cases can cause severe neuroinvasive disease in humans. The virus relies on a multitude of host cell proteins, molecules and cellular processes for its replication, and must subvert host antiviral responses to establish a successful infection. Studying the complex network of virus-host protein interactions by proteomics approaches can therefore provide new insights in the replication cycle of USUV and its pathogenesis. We have previously shown that the USUV protein NS4A acts as an antagonist of the antiviral interferon response, and here we further map the host interaction partners of USUV NS4A using proximity labeling coupled to mass spectrometry. The resulting NS4A interactome revealed many host proteins involved in the autophagy pathway. We showed that both USUV infection and overexpression of USUV NS4A can indeed induce the autophagy pathway. However, stimulation or inhibition of the autophagy pathway in general did not affect USUV replication. Therefore, we decided to specifically analyze the role of the selective autophagy receptor sequestosome 1 (p62/SQSTM1), since we identified this protein as an important interaction partner of USUV NS4A. We found that p62 is involved in the degradation of USUV NS4A. In agreement with this, the knockdown of p62 enhanced replication of USUV in A549 cells. P62 thus plays an antiviral role during USUV infection, although this antiviral effect might also be related to its functions outside the autophagy pathway, such as modulation of the immune response. In conclusion, this study showed that USUV NS4A induces autophagy and is then targeted by p62 for degradation by the autophagic machinery, uncovering a new role of p62 in the antiviral defense against USUV.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"103"},"PeriodicalIF":4.0,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12004613/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144048518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cytomegalovirus infection initiates inflammatory bowel disease by activating a positive MyD88/NF-κB feedback loop in allogeneic skin transplantation mice.","authors":"Ming-Xian Chen, Yu Chen, Rui Fu, Jie-Yi Wang, Sai-Yue Liu, Tang-Biao Shen","doi":"10.1186/s12985-025-02725-7","DOIUrl":"https://doi.org/10.1186/s12985-025-02725-7","url":null,"abstract":"<p><p>Infection with the cytomegalovirus (CMV) is common. Inflammatory bowel disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract. CMV infection is involved in IBD pathogenesis. The abnormal activation of myeloid differentiation factor 88 (MyD88)/nuclear factor- kappa B (NF-κB) signaling, which results in inflammatory cytokine overexpression, is an important factor in IBD pathogenesis. The present study aimed to examine the effect of CMV infection on NF-κB activation and its role in IBD pathogenesis. Since BALB/c rather than C57BL/6 mice belong to the murine CMV (MCMV) susceptible strain, allogeneic skin transplantation was conducted between MyD88 (+/+) or MyD88-knockout Myd88 (-/-) BALB/c (recipient) mice and C57BL/6 (donor) mice. Thereafter, the immune function of the recipient mice was reduced by immunosuppressant cyclosporine, which is meaningful in the pathogenesis of IBD caused by MCMV in immunocompromised mice. MCMV strain K181-eGFP (eGFP K181) or hMIEP-eGFP K181 (knockout MCMV IE1-3 promoter) was used to infect MyD88 (+/+) BALB/c mice while eGFP K181 was also used to infect MyD88 (-/-) BALB/c mice on day 14 post allogeneic skin transplantation. MCMV DNA was detected via nested polymerase chain reaction. Hematoxylin-Eosin staining was used to assess colon necrosis and inflammatory cell infiltration. The serum levels of tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, IL-8, IL-12, flagellin, lipopolysaccharide, and myeloperoxidase were detected by ELISA and immune reaction. Immunoblots were applied to measure protein levels. eGFP K181 infection significantly induced colon permeability, necrosis, inflammatory cell infiltration, and inflammation in allogeneic skin transplantation mice. hMIEP-eGFP K181 infection significantly inhibited colon permeability, necrosis, inflammatory cell infiltration, and inflammation compared with eGFP K181 infection in allogeneic skin transplantation mice. Moreover, the MyD88-dependent NF-κB signaling pathway was involved in the pathogenesis of eGFP K181-induced colon permeability and inflammation in allogeneic skin transplantation mice. Our findings highlight the importance of CMV infection and the Myd88/NF-κB signaling pathway in IBD and might provide a new direction for the development of drugs for IBD.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"101"},"PeriodicalIF":4.0,"publicationDate":"2025-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12001603/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144014837","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virology JournalPub Date : 2025-04-16DOI: 10.1186/s12985-025-02722-w
Emily Kwan, Alistair R Legione, Carol A Hartley, Joanne M Devlin
{"title":"Transcriptomic profiles of Crandell-Rees feline kidney cells infected with Varicellovirus felidalpha-1 (FHV-1) field and vaccine strains.","authors":"Emily Kwan, Alistair R Legione, Carol A Hartley, Joanne M Devlin","doi":"10.1186/s12985-025-02722-w","DOIUrl":"https://doi.org/10.1186/s12985-025-02722-w","url":null,"abstract":"<p><strong>Background: </strong>Varicellovirus felidalpha-1 (FHV-1, previously Felid alphaherpesvirus-1) is a significant cause of upper respiratory tract disease in feline populations. Cats infected with FHV-1 show clinical signs that vary in severity. This can be due to differences in host responses and virus strain virulence. Investigating the gene transcription profiles during infections using FHV-1 strains could inform our understanding of host and viral factors contributing to disease outcomes. This study characterised the transcriptomes of Crandell-Rees feline kidney (CRFK) cells infected with field or vaccine FHV-1 strains to better understand the host response during infection.</p><p><strong>Methods: </strong>Crandell-Rees feline kidney cells were infected with either the FHV-1 F2 vaccine strain or the 384/75 field strain associated with severe disease. The transcriptomes were characterised using RNA-sequencing. To determine the host cellular transcription profile, the total transcripts were mapped to the cat genome and compared to uninfected cells. To characterise the viral transcription profile, the total reads were mapped to each FHV-1 strain. The differentially expressed host genes between infection strains were compared and further analysed using the PANTHER database to examine host pathway regulation.</p><p><strong>Results: </strong>The findings in this study show the differential host gene expressions induced by FHV-1 compared to uninfected CRFK cells. Genes encoding histone proteins were upregulated, while genes involved in cell adhesion and migration processes were downregulated during infections with FHV-1. Comparative analysis between field and vaccine strains showed similarities and differences in host gene expressions. Notably, upregulated genes unique to the field strain were associated with regulatory proteins involved in the cell cycle, while downregulated host genes in field and vaccine strains showed distinct host gene and pathway expressions involved in immune activation.</p><p><strong>Conclusions: </strong>This study demonstrates the host and viral gene expressions during FHV-1 infection shows the distinct host responses to field and vaccine strains using an in vitro model. These findings provide a foundation for future transcriptomic investigations in other cell types, including ex-vivo explants systems, to enhance our understanding of host and viral factors contributing to disease outcomes.</p>","PeriodicalId":23616,"journal":{"name":"Virology Journal","volume":"22 1","pages":"102"},"PeriodicalIF":4.0,"publicationDate":"2025-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12004785/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144050616","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}