Persistent infection with foot-and-mouth disease virus (FMDV) develops in over 50% of infected ruminants, presenting major obstacles to disease control and eradication. To clarify host immune correlates of FMDV persistence, we characterized systemic T- and B-cell responses, as well as mucosal responses in 15 vaccinated cattle following experimental FMDV challenge. The prevalence of FMDV persistence was 53.3%. While peripheral CD4+, CD8+, and γδ T-cell populations and their respective naïve/memory/effector subpopulations showed comparable frequencies between carriers and noncarriers, carriers exhibited significantly lower frequencies of IFN-γ-producing CD4+ and CD8+ T lymphocytes during early infection, indicating compromised cell-mediated immune responses essential for viral clearance. During persistent infection, carriers displayed a distinctive immunological profile characterized by significantly reduced peripheral B-cell frequencies and increased secretory IgA (sIgA) levels in oropharyngeal fluid (OPF), with comparable systemic antigen-specific and neutralizing antibody titers across groups throughout the study period. Notably, the combination of peripheral B cell frequencies with OPF sIgA levels demonstrated superior diagnostic specificity for vaccinated carrier identification compared to either indicator alone. Our findings highlight key immune features of FMDV persistence and propose a dual-biomarker approach for detecting asymptomatic carriers.
{"title":"Distinctive Immunological Signatures Define Foot-and-Mouth Disease Virus Persistence in Vaccinated Cattle","authors":"Zhihui Zhang, Zhidong Teng, Shuang Wang, Suyu Mu, Sumin Wei, Hu Dong, Shuanghui Yin, Yun Zhang, Yaozhong Ding, Yijing Li, Shiqi Sun, Huichen Guo","doi":"10.1155/tbed/4010309","DOIUrl":"10.1155/tbed/4010309","url":null,"abstract":"<p>Persistent infection with foot-and-mouth disease virus (FMDV) develops in over 50% of infected ruminants, presenting major obstacles to disease control and eradication. To clarify host immune correlates of FMDV persistence, we characterized systemic T- and B-cell responses, as well as mucosal responses in 15 vaccinated cattle following experimental FMDV challenge. The prevalence of FMDV persistence was 53.3%. While peripheral CD4<sup>+</sup>, CD8<sup>+</sup>, and <i>γδ</i> T-cell populations and their respective naïve/memory/effector subpopulations showed comparable frequencies between carriers and noncarriers, carriers exhibited significantly lower frequencies of IFN-<i>γ</i>-producing CD4<sup>+</sup> and CD8<sup>+</sup> T lymphocytes during early infection, indicating compromised cell-mediated immune responses essential for viral clearance. During persistent infection, carriers displayed a distinctive immunological profile characterized by significantly reduced peripheral B-cell frequencies and increased secretory IgA (sIgA) levels in oropharyngeal fluid (OPF), with comparable systemic antigen-specific and neutralizing antibody titers across groups throughout the study period. Notably, the combination of peripheral B cell frequencies with OPF sIgA levels demonstrated superior diagnostic specificity for vaccinated carrier identification compared to either indicator alone. Our findings highlight key immune features of FMDV persistence and propose a dual-biomarker approach for detecting asymptomatic carriers.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12745507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861346","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yafang Lin, Yan Ouyang, Jiayang Zheng, Changguang Xiao, Yang Yang, Qianming Zhao, Yan Zhang, Zongjie Li, Ke Liu, Beibei Li, Donghua Shao, Yafeng Qiu, Zhiyong Ma, Jianchao Wei
Porcine reproductive and respiratory syndrome virus (PRRSV) causes major economic losses in swine production worldwide, characterized by reproductive failure and respiratory disease. In China, the cocirculation of diverse strains drives ongoing viral evolution and the emergence of novel recombinants, posing serious challenges for disease control. Here, we report the isolation and characterization of a novel PRRSV strain (JS-YZ/2023) from a PRRS-suspected pig farm. Genomic analysis revealed a unique recombinant pattern between NADC30-like and HP-PRRSV lineages, distinct from previously reported recombinants. Experimental infection in piglets demonstrated significant pathogenicity, with persistent fever, severe anorexia, progressive weight loss, and pathological lesions including interstitial pneumonia with lymphocyte infiltration and marked disruption of intestinal villi. Notably, JS-YZ/2023 exhibited strong intestinal tropism, inducing a local cytokine storm in the small intestine and showing PRRSV-N protein antigen accumulation in crypt epithelial cells, as detected by immunohistochemistry. In addition, we successfully constructed a full-length infectious cDNA clone of JS-YZ/2023 incorporating a green fluorescent protein (GFP) reporter gene (rGFP-JS-YZ/2023). This engineered virus provides a valuable tool for investigating the pathogenic mechanisms of NADC30-like recombinants and facilitating vaccine development.
{"title":"Molecular Characterization and Pathogenicity of a Novel Recombinant NADC30-Like PRRSV Strain Isolated From Piglets Inducing Intestinal Inflammation, and Construction of Its Infectious Clone","authors":"Yafang Lin, Yan Ouyang, Jiayang Zheng, Changguang Xiao, Yang Yang, Qianming Zhao, Yan Zhang, Zongjie Li, Ke Liu, Beibei Li, Donghua Shao, Yafeng Qiu, Zhiyong Ma, Jianchao Wei","doi":"10.1155/tbed/4893251","DOIUrl":"10.1155/tbed/4893251","url":null,"abstract":"<p>Porcine reproductive and respiratory syndrome virus (PRRSV) causes major economic losses in swine production worldwide, characterized by reproductive failure and respiratory disease. In China, the cocirculation of diverse strains drives ongoing viral evolution and the emergence of novel recombinants, posing serious challenges for disease control. Here, we report the isolation and characterization of a novel PRRSV strain (JS-YZ/2023) from a PRRS-suspected pig farm. Genomic analysis revealed a unique recombinant pattern between NADC30-like and HP-PRRSV lineages, distinct from previously reported recombinants. Experimental infection in piglets demonstrated significant pathogenicity, with persistent fever, severe anorexia, progressive weight loss, and pathological lesions including interstitial pneumonia with lymphocyte infiltration and marked disruption of intestinal villi. Notably, JS-YZ/2023 exhibited strong intestinal tropism, inducing a local cytokine storm in the small intestine and showing PRRSV-N protein antigen accumulation in crypt epithelial cells, as detected by immunohistochemistry. In addition, we successfully constructed a full-length infectious cDNA clone of JS-YZ/2023 incorporating a green fluorescent protein (GFP) reporter gene (rGFP-JS-YZ/2023). This engineered virus provides a valuable tool for investigating the pathogenic mechanisms of NADC30-like recombinants and facilitating vaccine development.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12741902/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145848564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Duck Tembusu virus (DTMUV) is an emerging avian pathogenic flavivirus that causes neurological disorders and acute egg drop syndrome in ducks. Multiple genotypes of DTMUV, including clusters 1, 2, and 3, have been identified, and recent evidence suggests a correlation between viral genotype and disease severity. Our recent study demonstrated that DTMUV cluster 2.1 induces more severe clinical disease in ducks than cluster 1; however, the underlying mechanisms responsible for this difference remain unclear. Macrophages are key target cells for DTMUV replication and play a crucial role during the early stage of viral pathogenesis. Therefore, we hypothesized that distinct DTMUV genotypes may differentially regulate macrophage apoptosis, thereby influencing viral replication and modulating T lymphocyte responses. To investigate this, we developed an in vitro protocol to generate duck monocyte-derived macrophages (MDMs) and assessed their response to infection with two different DTMUV genotypes: cluster 1 and cluster 2.1. Our results showed that DTMUV cluster 1 infection significantly increased macrophage apoptosis and reduced cell viability. In contrast, DTMUV cluster 2.1-infected MDM exhibited higher cell viability and fewer apoptotic cells. Correspondingly, viral titers in supernatants from DTMUV cluster 2.1-infected cultures were significantly higher than those from cluster 1. Furthermore, culture with DTMUV cluster 2.1 resulted in enhanced proliferation of Th (CD3+CD4+BrdU+) and cytotoxic T lymphocyte (CTL; CD3+CD8+BrdU+) lymphocytes in both naïve peripheral blood mononuclear cells (PBMCs) and macrophage-peripheral blood lymphocytes (PBLs) co-culture systems. These results suggest that the inhibition of apoptosis in DTMUV cluster 2.1-infected macrophages may enhance antigen-specific T lymphocyte responses. Overall, our findings reveal a genotype-dependent mechanism by which DTMUV modulates macrophage survival, thereby influencing both viral replication and T cell activation, and ultimately contributing to genotype-specific immunopathogenesis. These insights deepen our understanding of DTMUV immunopathogenesis and may inform the development of more effective genotype-targeted vaccination and disease control strategies.
{"title":"Genotype-Dependent Modulation of Macrophage Apoptosis and T Lymphocyte Responses by Duck Tembusu Virus Potentially Contributes to Genotype-Specific Immunopathogenesis","authors":"Teerawut Nedumpun, Kanana Rungprasert, Benchaphorn Limcharoen, Aunyaratana Thontiravong","doi":"10.1155/tbed/3076122","DOIUrl":"https://doi.org/10.1155/tbed/3076122","url":null,"abstract":"<p>Duck Tembusu virus (DTMUV) is an emerging avian pathogenic flavivirus that causes neurological disorders and acute egg drop syndrome in ducks. Multiple genotypes of DTMUV, including clusters 1, 2, and 3, have been identified, and recent evidence suggests a correlation between viral genotype and disease severity. Our recent study demonstrated that DTMUV cluster 2.1 induces more severe clinical disease in ducks than cluster 1; however, the underlying mechanisms responsible for this difference remain unclear. Macrophages are key target cells for DTMUV replication and play a crucial role during the early stage of viral pathogenesis. Therefore, we hypothesized that distinct DTMUV genotypes may differentially regulate macrophage apoptosis, thereby influencing viral replication and modulating T lymphocyte responses. To investigate this, we developed an in vitro protocol to generate duck monocyte-derived macrophages (MDMs) and assessed their response to infection with two different DTMUV genotypes: cluster 1 and cluster 2.1. Our results showed that DTMUV cluster 1 infection significantly increased macrophage apoptosis and reduced cell viability. In contrast, DTMUV cluster 2.1-infected MDM exhibited higher cell viability and fewer apoptotic cells. Correspondingly, viral titers in supernatants from DTMUV cluster 2.1-infected cultures were significantly higher than those from cluster 1. Furthermore, culture with DTMUV cluster 2.1 resulted in enhanced proliferation of Th (CD3<sup>+</sup>CD4<sup>+</sup>BrdU<sup>+</sup>) and cytotoxic T lymphocyte (CTL; CD3<sup>+</sup>CD8<sup>+</sup>BrdU<sup>+</sup>) lymphocytes in both naïve peripheral blood mononuclear cells (PBMCs) and macrophage-peripheral blood lymphocytes (PBLs) co-culture systems. These results suggest that the inhibition of apoptosis in DTMUV cluster 2.1-infected macrophages may enhance antigen-specific T lymphocyte responses. Overall, our findings reveal a genotype-dependent mechanism by which DTMUV modulates macrophage survival, thereby influencing both viral replication and T cell activation, and ultimately contributing to genotype-specific immunopathogenesis. These insights deepen our understanding of DTMUV immunopathogenesis and may inform the development of more effective genotype-targeted vaccination and disease control strategies.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/tbed/3076122","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145825266","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ahmad I. Al-Mustapha, Veronica Adetunji, Oluwaseun A. Ogundijo, Ismail A. Odetokun, Lateefah Oyafajo, Hauwa W. Abali, Muftau Oyewo, Ahmed Tijani Abubakar, Shuaibu Osu Muhammad, Deborah Adeolu Adetunji, Adesoji Odukoya, Anasin Haruna, Folashade Bamidele, Nusirat Elelu, Folorunso O. Fasina
<p>Decision-makers in animal health require reliable, evidence-based, timely, yet sensitive data to design disease contingency and preparedness plans, make informed decisions, and prioritize health interventions. Using Nigeria-specific animal health disease reports from the World Animal Health Information System (WAHIS), a global animal disease reporting platform, between 2006 and 2023, we conducted descriptive statistics to summarize the animal disease burden. We used a zero-inflated negative binomial (ZINB) regression model to forecast annual estimates of new outbreaks for the top 10 most reported diseases [Newcastle disease (NCD), infectious bursal disease (IBD), highly pathogenic avian influenza (HPAI), fowl typhoid, contagious bovine pleuropneumonia (CBPP), foot and mouth disease (FMD), African swine fever (ASF), peste des petits ruminants (PPR), rabies, and trypanosomiasis]. We analyzed 3362 official reports that were retrieved from the WAHIS platform and represented 9331 outbreaks of notifiable disease events that occurred in Nigeria. In summary, our analyses revealed that 3,248,945 cases and 4,911,495 culls were linked to the outbreaks, and ~2.2 million doses of vaccines were administered to animals. The most frequently reported non-zoonotic diseases were IBD (446 outbreaks) in poultry, PPR (2836 outbreaks) in small ruminants, and 642 outbreaks of CBPP in cattle. During the period under review, 3822 outbreaks (10 diseases) were reported to be zoonotic. Based on the animal species, there were 10 poultry diseases with HPAI (1230 outbreaks), NCD (1605 outbreaks), and fowl typhoid (241 outbreaks) being most frequently reported. In cattle, there were 11 diseases, with CBPP (642 outbreaks) and trypanosomiasis (233 outbreaks) being the most prevalent. The data revealed spatial variations in disease burden, with 20.7% (1934 outbreaks) reported from North Central Nigeria. Using data from 2006 to 2023, our model forecasted an increasing annual trend in the number of NCD outbreaks from 413 (95% CI: 246–679) in 2025 to 772 (95% CI: 473–1283) by 2030. There was a moderate increase in forecasted estimates for the vaccine-preventable diseases, rabies and FMD. The model forecasted some 68 FMD outbreaks (95% CI: 25–146) in 2025 and 144 (95% CI: 58–295) outbreaks by 2030 and Nigeria should plan against some 157 rabies outbreaks (95% CI: 79–289) in 2025, and this could rise to 252 outbreaks (95% CI: 133–457) by 2030. Outbreaks of ASF and the protozoan, tsetse-fly-transmitted disease trypanosomiasis were forecasted to have steady but slower increases, with ASF outbreaks estimated to range from 18 (95% CI: 5–46) in 2025 to 38 (95% CI: 13–93) by 2030y. Some 52 (95% CI: 19–112) outbreaks of the trypanosomiasis were forecasted for 2025. This is expected to increase to 95 (95% CI: 37–201) by 2030. The model estimated fewer than 10 (95% CI: 1–9) cases of HPAI annually. Finally, the model forecasted a modest but consistent rise in outbreaks of CBPP and fowl typhoid
{"title":"Animal Disease Burden in Nigeria, 2006–2023","authors":"Ahmad I. Al-Mustapha, Veronica Adetunji, Oluwaseun A. Ogundijo, Ismail A. Odetokun, Lateefah Oyafajo, Hauwa W. Abali, Muftau Oyewo, Ahmed Tijani Abubakar, Shuaibu Osu Muhammad, Deborah Adeolu Adetunji, Adesoji Odukoya, Anasin Haruna, Folashade Bamidele, Nusirat Elelu, Folorunso O. Fasina","doi":"10.1155/tbed/1694850","DOIUrl":"10.1155/tbed/1694850","url":null,"abstract":"<p>Decision-makers in animal health require reliable, evidence-based, timely, yet sensitive data to design disease contingency and preparedness plans, make informed decisions, and prioritize health interventions. Using Nigeria-specific animal health disease reports from the World Animal Health Information System (WAHIS), a global animal disease reporting platform, between 2006 and 2023, we conducted descriptive statistics to summarize the animal disease burden. We used a zero-inflated negative binomial (ZINB) regression model to forecast annual estimates of new outbreaks for the top 10 most reported diseases [Newcastle disease (NCD), infectious bursal disease (IBD), highly pathogenic avian influenza (HPAI), fowl typhoid, contagious bovine pleuropneumonia (CBPP), foot and mouth disease (FMD), African swine fever (ASF), peste des petits ruminants (PPR), rabies, and trypanosomiasis]. We analyzed 3362 official reports that were retrieved from the WAHIS platform and represented 9331 outbreaks of notifiable disease events that occurred in Nigeria. In summary, our analyses revealed that 3,248,945 cases and 4,911,495 culls were linked to the outbreaks, and ~2.2 million doses of vaccines were administered to animals. The most frequently reported non-zoonotic diseases were IBD (446 outbreaks) in poultry, PPR (2836 outbreaks) in small ruminants, and 642 outbreaks of CBPP in cattle. During the period under review, 3822 outbreaks (10 diseases) were reported to be zoonotic. Based on the animal species, there were 10 poultry diseases with HPAI (1230 outbreaks), NCD (1605 outbreaks), and fowl typhoid (241 outbreaks) being most frequently reported. In cattle, there were 11 diseases, with CBPP (642 outbreaks) and trypanosomiasis (233 outbreaks) being the most prevalent. The data revealed spatial variations in disease burden, with 20.7% (1934 outbreaks) reported from North Central Nigeria. Using data from 2006 to 2023, our model forecasted an increasing annual trend in the number of NCD outbreaks from 413 (95% CI: 246–679) in 2025 to 772 (95% CI: 473–1283) by 2030. There was a moderate increase in forecasted estimates for the vaccine-preventable diseases, rabies and FMD. The model forecasted some 68 FMD outbreaks (95% CI: 25–146) in 2025 and 144 (95% CI: 58–295) outbreaks by 2030 and Nigeria should plan against some 157 rabies outbreaks (95% CI: 79–289) in 2025, and this could rise to 252 outbreaks (95% CI: 133–457) by 2030. Outbreaks of ASF and the protozoan, tsetse-fly-transmitted disease trypanosomiasis were forecasted to have steady but slower increases, with ASF outbreaks estimated to range from 18 (95% CI: 5–46) in 2025 to 38 (95% CI: 13–93) by 2030y. Some 52 (95% CI: 19–112) outbreaks of the trypanosomiasis were forecasted for 2025. This is expected to increase to 95 (95% CI: 37–201) by 2030. The model estimated fewer than 10 (95% CI: 1–9) cases of HPAI annually. Finally, the model forecasted a modest but consistent rise in outbreaks of CBPP and fowl typhoid","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12721471/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145814815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yang Zhao, Jinling Wang, Qiang Liu, Jiang Wu, Qixin Huang, Bingwu Zhang, Yunze Guo, Chang Liu, Xing Guo, Kui Guo, Weiguo Zhang, Xiaohua Ma, Xue-Feng Wang, Xiaojun Wang
Enzootic nasal tumor virus (ENTV) is the etiological agent responsible for enzootic nasal adenocarcinoma (ENA), a chronic and contagious disease predominantly affecting sheep and goats. ENTV is classified into two distinct types: ENTV-1, which infects sheep, and ENTV-2, which infects goats. ENA has been globally reported in small ruminant-rearing regions, causing significant mortality and substantial economic impacts in affected flocks. There is currently no standardized detection method for ENA. In this study, we successfully generated a monoclonal antibody (mAb) and a polyclonal antibody (pAb) against the ENTV-2 capsid protein (p27), and identified the epitope of the mAb, which was found to be highly conserved among different ENTV-2 isolates. An antigen capture ELISA (acELISA) was then successfully developed using the mAb as the capture antibody and the pAb as the detection antibody to specifically detect p27 of ENTV-2 in nasal secretions. The cut-off value of the acELISA was determined to be 0.1052 by analyzing S/P values. The detection limit of this assay was 0.16 ng/mL of rp27 protein and equivalent to 844 copies/μL of ENTV-2 RNA. Specificity tests showed that the method had no cross-reaction with other prevalent small ruminant pathogens. The coincidence rates of the developed acELISA compared with western blotting and qRT-PCR assays were 98.95% (189/191) and 96.34% (184/191), respectively. Furthermore, the acELISA was applied to assess ENTV-2 in 1228 clinical nasal swab samples collected from seven provinces in China. The results demonstrated that the positivity rate varied between 0.00% and 28.21%. In conclusion, we successfully developed an acELISA with high specificity, sensitivity and reproducibility for the detection of ENTV-2 antigen. This high-throughput method for the detection of ENTV-2 represents a significant advancement in the field and may contribute to the prevention and control of ENTV-2.
{"title":"Development and Application of an Antigen Capture ELISA for the Detection of Enzootic Nasal Tumor Virus-2","authors":"Yang Zhao, Jinling Wang, Qiang Liu, Jiang Wu, Qixin Huang, Bingwu Zhang, Yunze Guo, Chang Liu, Xing Guo, Kui Guo, Weiguo Zhang, Xiaohua Ma, Xue-Feng Wang, Xiaojun Wang","doi":"10.1155/tbed/5514208","DOIUrl":"10.1155/tbed/5514208","url":null,"abstract":"<p>Enzootic nasal tumor virus (ENTV) is the etiological agent responsible for enzootic nasal adenocarcinoma (ENA), a chronic and contagious disease predominantly affecting sheep and goats. ENTV is classified into two distinct types: ENTV-1, which infects sheep, and ENTV-2, which infects goats. ENA has been globally reported in small ruminant-rearing regions, causing significant mortality and substantial economic impacts in affected flocks. There is currently no standardized detection method for ENA. In this study, we successfully generated a monoclonal antibody (mAb) and a polyclonal antibody (pAb) against the ENTV-2 capsid protein (p27), and identified the epitope of the mAb, which was found to be highly conserved among different ENTV-2 isolates. An antigen capture ELISA (acELISA) was then successfully developed using the mAb as the capture antibody and the pAb as the detection antibody to specifically detect p27 of ENTV-2 in nasal secretions. The cut-off value of the acELISA was determined to be 0.1052 by analyzing S/P values. The detection limit of this assay was 0.16 ng/mL of rp27 protein and equivalent to 844 copies/μL of ENTV-2 RNA. Specificity tests showed that the method had no cross-reaction with other prevalent small ruminant pathogens. The coincidence rates of the developed acELISA compared with western blotting and qRT-PCR assays were 98.95% (189/191) and 96.34% (184/191), respectively. Furthermore, the acELISA was applied to assess ENTV-2 in 1228 clinical nasal swab samples collected from seven provinces in China. The results demonstrated that the positivity rate varied between 0.00% and 28.21%. In conclusion, we successfully developed an acELISA with high specificity, sensitivity and reproducibility for the detection of ENTV-2 antigen. This high-throughput method for the detection of ENTV-2 represents a significant advancement in the field and may contribute to the prevention and control of ENTV-2.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717442/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145802767","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cato Van Herzele, Sieglinde Coppens, Sebastiaan Theuns, Lina De Smet, Jolien Van Cleemput, Dirk C. de Graaf, Hans Nauwynck
Honey bees are dying due to a disease complex consisting of viruses, parasites, chemicals, nutritional deficiencies, and management problems. In the present study, pathogens and hemocytes were analyzed in honey bee hemolymph samples using third-generation sequencing and flow cytometry in three apiaries over a honey bee season. Using nanopore sequencing, several viruses and bacteria were identified, including the first reported presence in European honey bees of La Jolla virus and Apis rhabdovirus 5. Seasonal pathogen peaks were found, with spring and summer showing the highest loads which coincide with the main flowering periods. Notably, viral infections often did not persist after initial detection despite the continuous appearance of new bees, hinting at colony-wide transgenerational immunity. However, Hubei partiti-like virus 34 remained endemic in one apiary and was found even in young bees, raising concerns about its ability to evade the transgenerational immunity. Additionally, dynamics in total hemocyte counts (THCs) were identified. Understanding pathogen and immune factor dynamics throughout the bee season will help identify weak colonies and provide valuable insights into colony collapse and its prevention.
{"title":"Pathogen and Hemocyte Dynamics in Three Apiaries Across a Bee Season","authors":"Cato Van Herzele, Sieglinde Coppens, Sebastiaan Theuns, Lina De Smet, Jolien Van Cleemput, Dirk C. de Graaf, Hans Nauwynck","doi":"10.1155/tbed/5065424","DOIUrl":"10.1155/tbed/5065424","url":null,"abstract":"<p>Honey bees are dying due to a disease complex consisting of viruses, parasites, chemicals, nutritional deficiencies, and management problems. In the present study, pathogens and hemocytes were analyzed in honey bee hemolymph samples using third-generation sequencing and flow cytometry in three apiaries over a honey bee season. Using nanopore sequencing, several viruses and bacteria were identified, including the first reported presence in European honey bees of La Jolla virus and Apis rhabdovirus 5. Seasonal pathogen peaks were found, with spring and summer showing the highest loads which coincide with the main flowering periods. Notably, viral infections often did not persist after initial detection despite the continuous appearance of new bees, hinting at colony-wide transgenerational immunity. However, Hubei partiti-like virus 34 remained endemic in one apiary and was found even in young bees, raising concerns about its ability to evade the transgenerational immunity. Additionally, dynamics in total hemocyte counts (THCs) were identified. Understanding pathogen and immune factor dynamics throughout the bee season will help identify weak colonies and provide valuable insights into colony collapse and its prevention.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717445/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145802909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The emergence of the NADC30-like (Lineage1.8) porcine reproductive and respiratory syndrome virus (PRRSV) has accelerated the evolutionary divergence of PRRSV, causing significant economic losses to Chinese swine industry. In the present study, a novel Lineage 7 (L7/Prime Pac-like) PRRSV strain FJZHK-2025 was isolated in China. Full-length genome analysis revealed that FJZHK-2025 is closely related to L7 PRRSV and exhibits the highest nucleotide homology (96.4%–96.5%) with the L7 PRRSV reference strains. Notably, FJZHK-2025 contained a 36 amino acid (aa) insertion within Nsp2 that was identical to the Prime Pac PRRS vaccine strain. Phylogenetic analysis, recombination detection, and regional genomic homology comparisons revealed that FJZHK-2025 originated from inter-lineage recombination between L7 and lineage 1.8 (NADC30-like) PRRSV-2 strains. Animal challenge experiments showed that piglets infected with FJZHK-2025 developed a persistent high fever, displayed mild to moderate clinical symptoms and presented with moderate lung pathological lesions, indicating that the FJZHK-2025 strain is pathogenic to piglets. Therefore, it is critical to develop an effective strategy to prevent and control the spread of L7 PRRSV in China.
{"title":"Genetic and Pathogenic Characteristics of an Emerging Lineage 7 PRRSV-2 (Prime Pac-Like) Strain in China","authors":"Yuan Yang, Haoyu Chen, Feimin Xie, Xuejing Li, Xin Lan, Hua Wang, Ziyi Wang, Kewei Fan, Wei Wei, Cheng Luo, Ailing Dai, Xiaohua Li, Manlin Luo, Chunhua Wei, Jiankui Liu","doi":"10.1155/tbed/8104154","DOIUrl":"10.1155/tbed/8104154","url":null,"abstract":"<p>The emergence of the NADC30-like (Lineage1.8) porcine reproductive and respiratory syndrome virus (PRRSV) has accelerated the evolutionary divergence of PRRSV, causing significant economic losses to Chinese swine industry. In the present study, a novel Lineage 7 (L7/Prime Pac-like) PRRSV strain FJZHK-2025 was isolated in China. Full-length genome analysis revealed that FJZHK-2025 is closely related to L7 PRRSV and exhibits the highest nucleotide homology (96.4%–96.5%) with the L7 PRRSV reference strains. Notably, FJZHK-2025 contained a 36 amino acid (aa) insertion within Nsp2 that was identical to the Prime Pac PRRS vaccine strain. Phylogenetic analysis, recombination detection, and regional genomic homology comparisons revealed that FJZHK-2025 originated from inter-lineage recombination between L7 and lineage 1.8 (NADC30-like) PRRSV-2 strains. Animal challenge experiments showed that piglets infected with FJZHK-2025 developed a persistent high fever, displayed mild to moderate clinical symptoms and presented with moderate lung pathological lesions, indicating that the FJZHK-2025 strain is pathogenic to piglets. Therefore, it is critical to develop an effective strategy to prevent and control the spread of L7 PRRSV in China.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12715340/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145802888","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hanyun Wang, Yujing Zhang, Zhenyu Wang, Yawen Xu, Le Zhou, Jing Wang, Xinan Jiao, Lin Sun
Tigecycline is considered a last-line therapeutic option for treating infections caused by multidrug-resistant (MDR) Gram-negative bacteria. The emergence of tmexCD1-toprJ1, a plasmid-mediated resistance-nodulation-division (RND) efflux pump gene cluster, poses a growing threat to tigecycline efficacy. While this gene cluster has primarily been identified in Klebsiella pneumoniae, its presence in other species remains poorly characterized. In this study, we investigated the occurrence, genetic features, resistance profile, and virulence potential of tmexCD-toprJ-positive Raoultella ornithinolytica isolates from a swine farm in China. A total of 126 samples were collected from a swine farm and screened for tigecycline-resistant isolates. Species identification was performed using MALDI-TOF MS, while PCR and sequencing were applied to detect tmexCD-toprJ. Antimicrobial susceptibility testing was assessed by agar and broth microdilution methods. Whole-genome sequencing with hybrid assembly provided insights into genetic organization. Conjugation and electroporation experiments were conducted to assess plasmid mobility, and virulence was assessed using the G. mellonella infection model. Six R. ornithinolytica isolates (4.76%) were identified as carrying tmexCD1-toprJ1 and exhibited multidrug resistance, including reduced susceptibility to tigecycline (minimal inhibitory concentrations [MICs] 4–16 mg/L). The resistance genes were located on highly similar IncFIB plasmids, which lacked typical conjugative elements. However, IS26 sequences flanking the gene cluster suggested potential for horizontal transfer. Phylogenetic analysis indicated possible clonal dissemination. All isolates carried chromosomally encoded virulence genes, and in vivo assays in G. mellonella revealed moderate to high pathogenicity. These findings expand the ecological distribution of tmexCD1-toprJ1 to R. ornithinolytica in livestock environments, underscoring the role of swine farm water systems as potential reservoirs. The coexistence of antimicrobial resistance and virulence determinants highlights the urgent need for strengthened surveillance and containment strategies to prevent further dissemination of tigecycline resistance.
{"title":"Emergence of Mobilized Tigecycline Resistance Gene Cluster tmexCD1-toprJ1 in Raoultella ornithinolytica From a Swine Farm, China","authors":"Hanyun Wang, Yujing Zhang, Zhenyu Wang, Yawen Xu, Le Zhou, Jing Wang, Xinan Jiao, Lin Sun","doi":"10.1155/tbed/6690944","DOIUrl":"10.1155/tbed/6690944","url":null,"abstract":"<p>Tigecycline is considered a last-line therapeutic option for treating infections caused by multidrug-resistant (MDR) Gram-negative bacteria. The emergence of <i>tmexCD1-toprJ1</i>, a plasmid-mediated resistance-nodulation-division (RND) efflux pump gene cluster, poses a growing threat to tigecycline efficacy. While this gene cluster has primarily been identified in <i>Klebsiella pneumoniae</i>, its presence in other species remains poorly characterized. In this study, we investigated the occurrence, genetic features, resistance profile, and virulence potential of <i>tmexCD-toprJ</i>-positive <i>Raoultella ornithinolytica</i> isolates from a swine farm in China. A total of 126 samples were collected from a swine farm and screened for tigecycline-resistant isolates. Species identification was performed using MALDI-TOF MS, while PCR and sequencing were applied to detect <i>tmexCD-toprJ</i>. Antimicrobial susceptibility testing was assessed by agar and broth microdilution methods. Whole-genome sequencing with hybrid assembly provided insights into genetic organization. Conjugation and electroporation experiments were conducted to assess plasmid mobility, and virulence was assessed using the <i>G. mellonella</i> infection model. Six <i>R. ornithinolytica</i> isolates (4.76%) were identified as carrying <i>tmexCD1-toprJ1</i> and exhibited multidrug resistance, including reduced susceptibility to tigecycline (minimal inhibitory concentrations [MICs] 4–16 mg/L). The resistance genes were located on highly similar IncFIB plasmids, which lacked typical conjugative elements. However, IS26 sequences flanking the gene cluster suggested potential for horizontal transfer. Phylogenetic analysis indicated possible clonal dissemination. All isolates carried chromosomally encoded virulence genes, and in vivo assays in <i>G. mellonella</i> revealed moderate to high pathogenicity. These findings expand the ecological distribution of <i>tmexCD1-toprJ1</i> to <i>R. ornithinolytica</i> in livestock environments, underscoring the role of swine farm water systems as potential reservoirs. The coexistence of antimicrobial resistance and virulence determinants highlights the urgent need for strengthened surveillance and containment strategies to prevent further dissemination of tigecycline resistance.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12714096/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145802870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hong Lin, Xiaoyang Zhu, Jianzhong Zhu, Nanhua Chen, Wenbin Bao
The red-crowned crane (Grus japonensis) is one of the rarest cranes with a global population of less than 4000 individuals. The population of red-crowned crane could be influenced by health threats, including metabolic and infectious diseases. In the Wildlife Rescue Center of Suining County of Jiangsu Province, gouty arthritis (GA) was observed in all four red-crowned cranes since March 2024. A pooled fecal supernatant was first submitted to metagenomics sequencing for screening disease-associated pathogens. Enterobacteria phage phiEcoM-GJ1 was detected as the predominant virus while Escherichia coli and Aeromonas hydrophila were the dominated bacteria in the mixed fecal sample from red-crowned cranes. The 16S rRNA gene sequencing was further performed on both the mixed fecal sample and four individual samples, which showed that Escherichia-Shigella, Lactobacillus, and Enterococcus were the most abundant gut flora in both mixed and individual fecal samples. Furthermore, bacteria isolation and identification with matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF/MS) confirmed that Escherichia coli was predominant (19/29 colonies, 65.52%) in the feces. Therefore, anti-uricacid and antibacteria treatments using plantain herb, doxycycline, Vitamin AD3 and multivitamin B were adopted, leading to a full behavioral recovery within 1 month. Overall, this case-based observational study provides first clue on the gut-joint axis in red-crowned cranes, supporting that gut microbiota dysbiosis is closely associated with GA in red-crowned cranes.
{"title":"High-Throughput Sequencings Revealed That Gut Microbiota Dysbiosis is Implicated in Gouty Arthritis of Red-Crowned Crane (Grus japonensis)","authors":"Hong Lin, Xiaoyang Zhu, Jianzhong Zhu, Nanhua Chen, Wenbin Bao","doi":"10.1155/tbed/2422900","DOIUrl":"10.1155/tbed/2422900","url":null,"abstract":"<p>The red-crowned crane (<i>Grus japonensis</i>) is one of the rarest cranes with a global population of less than 4000 individuals. The population of red-crowned crane could be influenced by health threats, including metabolic and infectious diseases. In the Wildlife Rescue Center of Suining County of Jiangsu Province, gouty arthritis (GA) was observed in all four red-crowned cranes since March 2024. A pooled fecal supernatant was first submitted to metagenomics sequencing for screening disease-associated pathogens. Enterobacteria phage phiEcoM-GJ1 was detected as the predominant virus while <i>Escherichia coli</i> and <i>Aeromonas hydrophila</i> were the dominated bacteria in the mixed fecal sample from red-crowned cranes. The 16S rRNA gene sequencing was further performed on both the mixed fecal sample and four individual samples, which showed that <i>Escherichia-Shigella</i>, <i>Lactobacillus</i>, and <i>Enterococcus</i> were the most abundant gut flora in both mixed and individual fecal samples. Furthermore, bacteria isolation and identification with matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF/MS) confirmed that <i>Escherichia coli</i> was predominant (19/29 colonies, 65.52%) in the feces. Therefore, anti-uricacid and antibacteria treatments using plantain herb, doxycycline, Vitamin AD3 and multivitamin B were adopted, leading to a full behavioral recovery within 1 month. Overall, this case-based observational study provides first clue on the gut-joint axis in red-crowned cranes, supporting that gut microbiota dysbiosis is closely associated with GA in red-crowned cranes.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12703207/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145766716","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yao Rong, Yidan Cheng, Hongbo Zhang, Xichen Zhang, Yu Zheng, Jianhua Li, Pengtao Gong, Xiaocen Wang, Xin Li, Nan Zhang
Pentatrichomonas hominis (P. hominis) has traditionally been regarded as a commensal or opportunistic inhabitant of the intestine. However, a growing number of studies have identified this protozoan as the sole pathogen in cases of intestinal disorders, including diarrhea, in both humans and animals, suggesting it may be a neglected zoonotic pathogen. To investigate the pathogenicity of P. hominis, we here systematically evaluated its effects on healthy immunocompetent BALB/c mice. Our findings revealed that infection with P. hominis induced injury in the large intestine, marked by inflammatory cell infiltration, epithelial cell necrosis, and intestinal mucosal sloughing. These pathological changes persisted and worsened throughout the 90-day observation period. Furthermore, infection appeared to disrupt goblet cell maturation or secretion, as indicated by increased periodic acid–Schiff (PAS) staining alongside decreased MUC2 production. Elevated levels of pro-inflammatory cytokines were also detected in both intestinal lavage fluid and serum. Finally, P. hominis infection altered the composition of the gut microbiota, increasing both its richness and diversity. Notably, it raised the relative abundance of the inflammation-associated genus Desulfovibrio, while reducing the abundance of beneficial bacteria, such as Akkermansia, Roseburia, and Lactobacillus. Collectively, these results provide compelling evidence that P. hominis acts as a zoonotic pathogen capable of inducing chronic intestinal inflammation. Therefore, the clinical significance of P. hominis infection warrants attention.
{"title":"Pentatrichomonas hominis Infection Induces Chronic Intestinal Inflammation in Immunocompetent Mice","authors":"Yao Rong, Yidan Cheng, Hongbo Zhang, Xichen Zhang, Yu Zheng, Jianhua Li, Pengtao Gong, Xiaocen Wang, Xin Li, Nan Zhang","doi":"10.1155/tbed/8820836","DOIUrl":"https://doi.org/10.1155/tbed/8820836","url":null,"abstract":"<p><i>Pentatrichomonas hominis</i> (<i>P. hominis</i>) has traditionally been regarded as a commensal or opportunistic inhabitant of the intestine. However, a growing number of studies have identified this protozoan as the sole pathogen in cases of intestinal disorders, including diarrhea, in both humans and animals, suggesting it may be a neglected zoonotic pathogen. To investigate the pathogenicity of <i>P. hominis</i>, we here systematically evaluated its effects on healthy immunocompetent BALB/c mice. Our findings revealed that infection with <i>P. hominis</i> induced injury in the large intestine, marked by inflammatory cell infiltration, epithelial cell necrosis, and intestinal mucosal sloughing. These pathological changes persisted and worsened throughout the 90-day observation period. Furthermore, infection appeared to disrupt goblet cell maturation or secretion, as indicated by increased periodic acid–Schiff (PAS) staining alongside decreased MUC2 production. Elevated levels of pro-inflammatory cytokines were also detected in both intestinal lavage fluid and serum. Finally, <i>P. hominis</i> infection altered the composition of the gut microbiota, increasing both its richness and diversity. Notably, it raised the relative abundance of the inflammation-associated genus <i>Desulfovibrio</i>, while reducing the abundance of beneficial bacteria, such as <i>Akkermansia</i>, <i>Roseburia</i>, and <i>Lactobacillus</i>. Collectively, these results provide compelling evidence that <i>P. hominis</i> acts as a zoonotic pathogen capable of inducing chronic intestinal inflammation. Therefore, the clinical significance of <i>P. hominis</i> infection warrants attention.</p>","PeriodicalId":234,"journal":{"name":"Transboundary and Emerging Diseases","volume":"2025 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1155/tbed/8820836","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145739755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}