Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28.680
Filter
1.
J Med Virol ; 96(7): e29777, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949212

ABSTRACT

Hepatitis E virus (HEV) is a prevalent pathogen responsible for acute viral hepatitis, HEV genotypes 3 and 4 infections causing zoonotic infections. Currently, the nucleotide similarity analysis between humans and pigs for HEV genotype 4 is limited. In this study, stool samples from an HEV-infected patient who is a pig farmer and from pigs were collected to obtain the near full-length genome of HEV, phylogenetic trees were constructed for genotyping, and similarity of HEV sequences was analyzed. The results showed that HEV-RNA was detected in the stool samples from the patient and six pigs (6/30, 20.0%). Both HEV subtype in the patient and pigs was 4b. Additionally, similarity analysis showed that the range was 99.875%-99.944% between the patient and pigs at the nucleotide level. Four isolates of amino acid sequences (ORFs 1-3) from pigs were 100% identical to the patient. Phylogenetic tree and similarity analysis of an additional nine HEV sequences isolated from other patients in this region showed that the HEV sequence from the pig farmer had the closest relationship with the pigs from his farm rather than other sources of infection in this region. This study provides indirect evidences for HEV subtype 4b can be transmitted from pigs to humans at the nucleotide level. Further research is needed to explore the characteristics of different HEV subtypes.


Subject(s)
Feces , Genome, Viral , Genotype , Hepatitis E virus , Hepatitis E , Phylogeny , RNA, Viral , Swine Diseases , Animals , Hepatitis E virus/genetics , Hepatitis E virus/classification , Hepatitis E virus/isolation & purification , Swine , Hepatitis E/virology , Hepatitis E/veterinary , Hepatitis E/epidemiology , China/epidemiology , Humans , Feces/virology , Swine Diseases/virology , RNA, Viral/genetics , Male , Sequence Analysis, DNA
3.
Emerg Microbes Infect ; 13(1): 2368202, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38970562

ABSTRACT

Influenza A viruses (IAV) impose significant respiratory disease burdens in both swine and humans worldwide, with frequent human-to-swine transmission driving viral evolution in pigs and highlighting the risk at the animal-human interface. Therefore, a comprehensive One Health approach (interconnection among human, animal, and environmental health) is needed for IAV prevention, control, and response. Animal influenza genomic surveillance remains limited in many Latin American countries, including Colombia. To address this gap, we genetically characterized 170 swine specimens from Colombia (2011-2017). Whole genome sequencing revealed a predominance of pandemic-like H1N1 lineage, with a minority belonging to H3N2 and H1N2 human seasonal-like lineage and H1N1 early classical swine lineages. Significantly, we have identified reassortant and recombinant viruses (H3N2, H1N1) not previously reported in Colombia. This suggests a broad genotypic viral diversity, likely resulting from reassortment between classical endemic viruses and new introductions established in Colombia's swine population (e.g. the 2009 H1N1 pandemic). Our study highlights the importance of a One Health approach in disease control, particularly in an ecosystem where humans are a main source of IAV to swine populations, and emphasizes the need for continued surveillance and enhanced biosecurity measures. The co-circulation of multiple subtypes in regions with high swine density facilitates viral exchange, underscoring the importance of monitoring viral evolution to inform vaccine selection and public health policies locally and globally.


Subject(s)
Evolution, Molecular , Genetic Variation , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , Phylogeny , Swine Diseases , Animals , Swine , Colombia/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/epidemiology , Swine Diseases/virology , Swine Diseases/epidemiology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/isolation & purification , One Health , Humans , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Whole Genome Sequencing , Genome, Viral , Epidemiological Monitoring , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/classification , Influenza, Human/virology , Influenza, Human/epidemiology
4.
Microb Biotechnol ; 17(7): e14518, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953907

ABSTRACT

Porcine epidemic diarrhoea virus (PEDV) infects pigs of all ages by invading small intestine, causing acute diarrhoea, vomiting, and dehydration with high morbidity and mortality among newborn piglets. However, current PEDV vaccines are not effective to protect the pigs from field epidemic strains because of poor mucosal immune response and strain variation. Therefore, it is indispensable to develop a novel oral vaccine based on epidemic strains. Bacillus subtilis spores are attractive delivery vehicles for oral vaccination on account of the safety, high stability, and low cost. In this study, a chimeric gene CotC-Linker-COE (CLE), comprising of the B. subtilis spore coat gene cotC fused to the core neutralizing epitope CO-26 K equivalent (COE) of the epidemic strain PEDV-AJ1102 spike protein gene, was constructed. Then recombinant B. subtilis displaying the CLE on the spore surface was developed by homologous recombination. Mice were immunized by oral route with B. subtilis 168-CLE, B. subtilis 168, or phosphate-buffered saline (PBS) as control. Results showed that the IgG antibodies and cytokine (IL-4, IFN-γ) levels in the B. subtilis 168-CLE group were significantly higher than the control groups. This study demonstrates that B. subtilis 168-CLE can generate specific systemic immune and mucosal immune responses and is a potential vaccine candidate against PEDV infection.


Subject(s)
Antibodies, Viral , Bacillus subtilis , Porcine epidemic diarrhea virus , Spores, Bacterial , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/immunology , Animals , Bacillus subtilis/genetics , Bacillus subtilis/immunology , Spores, Bacterial/genetics , Spores, Bacterial/immunology , Mice , Antibodies, Viral/blood , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/microbiology , Swine Diseases/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Administration, Oral , Cytokines/metabolism , Immunoglobulin G/blood , Mice, Inbred BALB C , Female , Cell Surface Display Techniques , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
5.
Vet Microbiol ; 295: 110160, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964034

ABSTRACT

Infection with Glaesserella parasuis, the primary pathogen behind Glässer's disease, is often associated with diverse clinical symptoms, including serofibrinous polyserositis, arthritis, and meningitis. Autophagy plays a dual role in bacterial infections, exerting either antagonistic or synergistic effects depending on the nature of the pathogen. Our previous studies have demonstrated that autophagy serves as a defense mechanism, combating inflammation and invasion caused by infection of highly virulent G. parasuis. However, the precise mechanisms remain to be elucidated. Pathogens exhibit distinct interactions with inflammasomes and autophagy processes. Herein, we explored the effect of autophagy on inflammasomes during G. parasuis infection. We found that G. parasuis infection triggers NLRP3-dependent pro-CASP-1-IL-18/IL-1ß processing and maturation pathway, resulting in increased release of IL-1ß and IL-18. Inhibition of autophagy enhances NLRP3 inflammasome activity, whereas stimulation of autophagy restricts it during G. parasuis infection. Furthermore, assembled NLRP3 inflammasomes undergo ubiquitination and recruit the autophagic adaptor, p62, facilitating their sequestration into autophagosomes during G. parasuis infection. These results suggest that the induction of autophagy mitigates inflammation by eliminating overactive NLRP3 inflammasomes during G. parasuis infection. Our research uncovers a mechanism whereby G. parasuis infection initiates inflammatory responses by promoting the assembly of the NLRP3 inflammasomes and activating NLRP3-CASP-1, both of which processes are downregulated by autophagy. This suggests that pharmacological manipulation of autophagy could be a promising approach to modulate G. parasuis-induced inflammatory responses.


Subject(s)
Autophagy , Caspase 1 , Haemophilus Infections , Haemophilus parasuis , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Haemophilus parasuis/immunology , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/genetics , Caspase 1/metabolism , Caspase 1/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Swine , Interleukin-18/metabolism , Interleukin-18/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Swine Diseases/microbiology , Swine Diseases/immunology , Mice
6.
Vet Microbiol ; 295: 110168, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964035

ABSTRACT

Glaesserella parasuis is an important porcine pathogen that commonly colonizes the upper respiratory tract of pigs and is prone to causing Glässer's disease under complex conditions. As yet, the disease has led to serious economic losses to the swine industry worldwide. Studies so far have found that several virulence factors are associated with the pathogenicity of G. parasuis, but the pathogenic mechanism is still not fully understood. Cytolethal distending toxin (CDT), a potential virulence factor in G. parasuis, is involved in cytotoxicity, serum resistance, adherence to and invasion of host cells in vitro. Here, to further investigate the pathogenic role of CDT during G. parasuis infection in vitro and in vivo, a double cdt1 and cdt2 deletion mutant (Δcdt1Δcdt2) without selectable marker was first generated in G. parasuis JS0135 strain by continuous natural transformations and replica plating. Morphological observation and lactate dehydrogenase assay showed that the Δcdt1Δcdt2 mutant was defective in cytotoxicity. Additionally, the Δcdt1Δcdt2 mutant was more susceptible to phagocytosis caused by 3D4/2 macrophages compared to the wild-type JS0135 strain. Moreover, by focusing on clinical signs, necropsy, bacterial recovery and pathological observation, we found that the deletion of cdt1 and cdt2 genes led to a significant attenuation of virulence in G. parasuis. Taken together, these findings suggest that as an important virulence factor, CDT can significantly affect the pathogenicity of G. parasuis.


Subject(s)
Bacterial Toxins , Haemophilus parasuis , Phagocytosis , Swine Diseases , Animals , Swine , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/genetics , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Swine Diseases/microbiology , Virulence , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Haemophilus Infections/immunology , Virulence Factors/genetics , Macrophages/microbiology , Cell Line
7.
Vet Res ; 55(1): 87, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982477

ABSTRACT

Hepatitis E virus (HEV) is a major cause of viral hepatitis worldwide. Pigs are the natural host of HEV genotype 3 and the main reservoir of HEV. As the host range of HEV genotype 3 expands, the possibility that HEV from various species can be transmitted to humans via pigs is increasing. We investigated the potential cross-species transmission of HEV by infecting minipigs with swine HEV (swHEV), rabbit HEV (rbHEV), and human HEV (huHEV) and examining their histopathological characteristics and distribution in various organs. Fifteen specific-pathogen-free Yucatan minipigs were infected with swHEV, rbHEV, huHEV, or a mock control. In the present study, we analysed faecal shedding, viremia, and serological parameters over a seven-week period. Our results indicated that swHEV exhibited more robust shedding and viremia than non-swHEVs. Only swHEV affected the serological parameters, suggesting strain-specific differences. Histopathological examination revealed distinct patterns in the liver, pancreas, intestine, and lymphoid tissues after infection with each HEV strain. Notably, all three HEVs induced histopathological changes in the pancreas, supporting the association of HEVs with acute pancreatitis. Our results also identified skeletal muscle as a site of HEV antigen presence, suggesting a potential link to myositis. In conclusion, this study provides valuable insights into the infection dynamics of different HEV strains in minipigs, emphasizing the strain-specific variations in virological, serological, and histological parameters. The observed differences in infection kinetics and tissue tropism will contribute to our understanding of HEV pathogenesis and the potential for cross-species transmission.


Subject(s)
Hepatitis E virus , Hepatitis E , Swine Diseases , Swine, Miniature , Animals , Swine , Hepatitis E/veterinary , Hepatitis E/virology , Hepatitis E/transmission , Hepatitis E virus/physiology , Swine Diseases/virology , Swine Diseases/transmission , Swine Diseases/pathology , Specific Pathogen-Free Organisms , Rabbits , Virus Shedding , Humans , Feces/virology , Female , Viremia/veterinary , Viremia/virology
8.
Acta Vet Scand ; 66(1): 34, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020377

ABSTRACT

Monitoring the use of antimicrobials and the emergence of resistance in animals and people is important for the control of antimicrobial resistance, and for establishing sustainable and effective disease management practices. In this study, we used Enterococcus spp. and Escherichia coli as indicator species to investigate antimicrobial susceptibility patterns and how these change over time, on ten Swedish pig farms. Indoor environmental sock sampling was performed once a month during the entire production cycle of one batch of pigs on each farm, resulting in 60 samples collected in total. Selective culture for E. coli and Enterococcus spp. resulted in 122 isolates of E. coli, 74 isolates of E. faecium, but no isolates of E. faecalis. Microdilution was used to determine minimum inhibitory concentrations for twelve antimicrobial substances in E. coli and fifteen substances in E. faecium. The overall prevalence of resistance was low. Among the E. coli isolates, the proportions non-wild type (resistant, NWT) isolates were as follows: azithromycin and amikacin 1% (n = 1), trimethoprim and sulfamethoxazole 2% (n = 3), ampicillin 6% (n = 7) and tetracycline 9% (n = 11). Among the E. faecium isolates, the NWT proportions were: teicoplanin, linezolid and gentamicin 1% (n = 1), daptomycin 3% (n = 2), erythromycin 26% (n = 19), tetracycline 27% (n = 20), quinupristin/dalfopristin 58% (n = 42). The resistance patterns differed between the farms, likely due to different antimicrobial use, biosecurity measures and source of the animals. The NWT prevalence among E. coli decreased over time, whereas no similar trend could be observed in E. faecium. The results of the current study illustrate the complex factors affecting the antimicrobial resistance patterns observed on each farm, indicating that specific practices and risk factors have an impact on the prevalence and type of antimicrobial resistance. Further studies of the farm environments in combination with antimicrobial use and other risk factor data are needed to elucidate the multifaceted drivers of antimicrobial resistance development on livestock farms.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Enterococcus faecium , Escherichia coli , Microbial Sensitivity Tests , Swine Diseases , Animals , Enterococcus faecium/drug effects , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Swine , Anti-Bacterial Agents/pharmacology , Sweden/epidemiology , Microbial Sensitivity Tests/veterinary , Swine Diseases/microbiology , Swine Diseases/epidemiology , Farms , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Escherichia coli Infections/epidemiology , Gram-Positive Bacterial Infections/veterinary , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Prevalence , Animal Husbandry/methods
12.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044581

ABSTRACT

This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 µg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 µg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 µg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 µg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Porcine epidemic diarrhea virus , Recombinant Fusion Proteins , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Mice , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Protein Domains , Immunogenicity, Vaccine , Immunity, Humoral
13.
Virol J ; 21(1): 160, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039549

ABSTRACT

Porcine Rotavirus (PoRV) is a significant pathogen affecting swine-rearing regions globally, presenting a substantial threat to the economic development of the livestock sector. At present, no specific pharmaceuticals are available for this disease, and treatment options remain exceedingly limited. This study seeks to design a multi-epitope peptide vaccine for PoRV employing bioinformatics approaches to robustly activate T-cell and B-cell immune responses. Two antigenic proteins, VP7 and VP8*, were selected from PoRV, and potential immunogenic T-cell and B-cell epitopes were predicted using immunoinformatic tools. These epitopes were further screened according to non-toxicity, antigenicity, non-allergenicity, and immunogenicity criteria. The selected epitopes were linked with linkers to form a novel multi-epitope vaccine construct, with the PADRE sequence (AKFVAAWTLKAAA) and RS09 peptide attached at the N-terminus of the designed peptide chain to enhance the vaccine's antigenicity. Protein-protein docking of the vaccine constructs with toll-like receptors (TLR3 and TLR4) was conducted using computational methods, with the lowest energy docking results selected as the optimal predictive model. Subsequently, molecular dynamics (MD) simulation methods were employed to assess the stability of the protein vaccine constructs and TLR3 and TLR4 receptors. The results indicated that the vaccine-TLR3 and vaccine-TLR4 docking models remained stable throughout the simulation period. Additionally, the C-IMMSIM tool was utilized to determine the immunogenic triggering capability of the vaccine protein, demonstrating that the constructed vaccine protein could induce both cell-mediated and humoral immune responses, thereby playing a role in eliciting host immune responses. In conclusion, this study successfully constructed a multi-epitope vaccine against PoRV and validated the stability and efficacy of the vaccine through computational analysis. However, as the study is purely computational, experimental evaluation is required to validate the safety and immunogenicity of the newly constructed vaccine protein.


Subject(s)
Antigens, Viral , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Dynamics Simulation , Rotavirus Infections , Rotavirus Vaccines , Rotavirus , Vaccines, Subunit , Animals , Swine , Rotavirus/immunology , Rotavirus/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Rotavirus Vaccines/immunology , Rotavirus Vaccines/chemistry , Rotavirus Vaccines/genetics , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Rotavirus Infections/virology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/chemistry , Antigens, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/chemistry , Molecular Docking Simulation , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Vaccine Development , Immunogenicity, Vaccine
14.
Vet Res ; 55(1): 91, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039559

ABSTRACT

The porcine epidemic diarrhea virus (PEDV) causes diarrhea in piglets, thereby causing very significant economic losses for the global swine industry. In previous studies, it has been confirmed that microRNAs (miRNAs) play an important role in the infection caused by PEDV. However, the precise molecular mechanism of miRNAs in the regulation of PEDV infection is still not fully understood. In the present study, we utilized miRNA-seq analysis to identify ssc-miR-1343 with differential expression between PEDV-infected and normal piglets. The expression of ssc-miR-1343 was detected in isolated exosomes, and it was found to be significantly higher than that in the controls following PEDV infection. The ssc-miR-1343 mimic was found to decrease PEDV replication, whereas the ssc-miR-1343 inhibitor was observed to increase PEDV replication, and ssc-miR-1343 was delivered by exosomes during PEDV infection. Mechanistically, ssc-miR-1343 binds to the 3'UTR region of FAM131C, down-regulating its expression, and FAM131C has been shown to enhance PEDV replication through simultaneously suppressing pathways associated with innate immunity. The ssc-miR-1343/FAM131C axis was found to upregulate the host immune response against PEDV infection. In conclusion, our findings indicate that the transport of ssc-miR-1343 in exosomes is involved in PEDV infection. This discovery presents a new potential target for the development of drugs to treat PEDV.


Subject(s)
Coronavirus Infections , Exosomes , MicroRNAs , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/physiology , Porcine epidemic diarrhea virus/genetics , Swine , MicroRNAs/metabolism , MicroRNAs/genetics , Swine Diseases/virology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Exosomes/metabolism , Virus Replication
15.
BMC Vet Res ; 20(1): 288, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961481

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) mainly causes acute and severe porcine epidemic diarrhea (PED), and is highly fatal in neonatal piglets. No reliable therapeutics against the infection exist, which poses a major global health issue for piglets. Luteolin is a flavonoid with anti-viral activity toward several viruses. RESULTS: We evaluated anti-viral effects of luteolin in PEDV-infected Vero and IPEC-J2 cells, and identified IC50 values of 23.87 µM and 68.5 µM, respectively. And found PEDV internalization, replication and release were significantly reduced upon luteolin treatment. As luteolin could bind to human ACE2 and SARS-CoV-2 main protease (Mpro) to contribute viral entry, we first identified that luteolin shares the same core binding site on pACE2 with PEDV-S by molecular docking and exhibited positive pACE2 binding with an affinity constant of 71.6 µM at dose-dependent increases by surface plasmon resonance (SPR) assay. However, pACE2 was incapable of binding to PEDV-S1. Therefore, luteolin inhibited PEDV internalization independent of PEDV-S binding to pACE2. Moreover, luteolin was firmly embedded in the groove of active pocket of Mpro in a three-dimensional docking model, and fluorescence resonance energy transfer (FRET) assays confirmed that luteolin inhibited PEDV Mpro activity. In addition, we also observed PEDV-induced pro-inflammatory cytokine inhibition and Nrf2-induced HO-1 expression. Finally, a drug resistant mutant was isolated after 10 cell culture passages concomitant with increasing luteolin concentrations, with reduced PEDV susceptibility to luteolin identified at passage 10. CONCLUSIONS: Our results push forward that anti-PEDV mechanisms and resistant-PEDV properties for luteolin, which may be used to combat PED.


Subject(s)
Antiviral Agents , Luteolin , Porcine epidemic diarrhea virus , Luteolin/pharmacology , Porcine epidemic diarrhea virus/drug effects , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Vero Cells , Swine , Molecular Docking Simulation , Virus Internalization/drug effects , Virus Replication/drug effects , Cell Line , Computer Simulation , Swine Diseases/virology , Swine Diseases/drug therapy
16.
Sci Rep ; 14(1): 15466, 2024 07 05.
Article in English | MEDLINE | ID: mdl-38965336

ABSTRACT

This study aimed to evaluate the efficacy of Lactiplantibacillus argentoratensis AGMB00912 (LA) in reducing Salmonella Typhimurium infection in weaned piglets. The investigation focused on the influence of LA on the gut microbiota composition, growth performance, and Salmonella fecal shedding. The results indicated that LA supplementation significantly improved average daily gain and reduced the prevalence and severity of diarrhea. Fecal analysis revealed reduced Salmonella shedding in the LA-supplemented group. Furthermore, LA notably altered the composition of the gut microbiota, increasing the levels of beneficial Bacillus and decreasing those of harmful Proteobacteria and Spirochaetes. Histopathological examination showed less intestinal damage in LA-treated piglets than in the controls. The study also observed that LA affected metabolic functions related to carbohydrate, amino acid, and fatty acid metabolism, thereby enhancing gut health and resilience against infection. Short-chain fatty acid concentrations in the feces were higher in the LA group, suggesting improved gut microbial activity. LA supplementation enriched the population of beneficial bacteria, including Streptococcus, Clostridium, and Bifidobacterium, while reducing the number of harmful bacteria, such as Escherichia and Campylobacter. These findings indicate the potential of LA as a probiotic alternative for swine nutrition, offering protective effects to the gut microbiota against Salmonella infection.


Subject(s)
Feces , Gastrointestinal Microbiome , Probiotics , Weaning , Animals , Gastrointestinal Microbiome/drug effects , Swine , Pilot Projects , Probiotics/administration & dosage , Feces/microbiology , Salmonella Infections, Animal/microbiology , Swine Diseases/microbiology , Swine Diseases/prevention & control , Lactobacillaceae , Salmonella typhimurium/drug effects
17.
PLoS One ; 19(7): e0306532, 2024.
Article in English | MEDLINE | ID: mdl-38968319

ABSTRACT

This study evaluated the use of endemic enteric coronaviruses polymerase chain reaction (PCR)-negative testing results as an alternative approach to detect the emergence of animal health threats with similar clinical diseases presentation. This retrospective study, conducted in the United States, used PCR-negative testing results from porcine samples tested at six veterinary diagnostic laboratories. As a proof of concept, the database was first searched for transmissible gastroenteritis virus (TGEV) negative submissions between January 1st, 2010, through April 29th, 2013, when the first porcine epidemic diarrhea virus (PEDV) case was diagnosed. Secondly, TGEV- and PEDV-negative submissions were used to detect the porcine delta coronavirus (PDCoV) emergence in 2014. Lastly, encountered best detection algorithms were implemented to prospectively monitor the 2023 enteric coronavirus-negative submissions. Time series (weekly TGEV-negative counts) and Seasonal Autoregressive-Integrated Moving-Average (SARIMA) were used to control for outliers, trends, and seasonality. The SARIMA's fitted and residuals were then subjected to anomaly detection algorithms (EARS, EWMA, CUSUM, Farrington) to identify alarms, defined as weeks of higher TGEV-negativity than what was predicted by models preceding the PEDV emergence. The best-performing detection algorithms had the lowest false alarms (number of alarms detected during the baseline) and highest time to detect (number of weeks between the first alarm and PEDV emergence). The best-performing detection algorithms were CUSUM, EWMA, and Farrington flexible using SARIMA fitted values, having a lower false alarm rate and identified alarms 4 to 17 weeks before PEDV and PDCoV emergences. No alarms were identified in the 2023 enteric negative testing results. The negative-based monitoring system functioned in the case of PEDV propagating epidemic and in the presence of a concurrent propagating epidemic with the PDCoV emergence. It demonstrated its applicability as an additional tool for diagnostic data monitoring of emergent pathogens having similar clinical disease as the monitored endemic pathogens.


Subject(s)
Coronavirus Infections , Porcine epidemic diarrhea virus , Swine Diseases , Transmissible gastroenteritis virus , Animals , Swine , Transmissible gastroenteritis virus/genetics , Transmissible gastroenteritis virus/isolation & purification , Porcine epidemic diarrhea virus/isolation & purification , Porcine epidemic diarrhea virus/genetics , Coronavirus Infections/diagnosis , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Swine Diseases/virology , Swine Diseases/diagnosis , Retrospective Studies , Gastroenteritis, Transmissible, of Swine/diagnosis , Gastroenteritis, Transmissible, of Swine/virology , Gastroenteritis, Transmissible, of Swine/epidemiology , Polymerase Chain Reaction/methods , Deltacoronavirus/genetics , Deltacoronavirus/isolation & purification , United States/epidemiology
18.
Res Vet Sci ; 176: 105351, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38963994

ABSTRACT

Sarcoptic mange is a widely distributed disease, with numerous potential hosts among domestic and wild animals. Nowadays it is considered a neglected re-emergent infection in humans. As a difference with domestic pigs, and even with several clinical cases reported in some European countries, it seems that Eurasian wild boars (Sus scrofa) have a low susceptibility to clinical mange. However, because of a case of confirmed transmission from Spanish ibex (Capra pyrenaica) to wild boar in the province of Tarragona, we planned a large-scale ELISA survey in the neighboring Valencian Community (SE Spain). We compared 419 wild boar sera from different management systems (fenced vs. open game estates), different ages (piglets, juveniles, and adults), with different behaviour (gregarious females of all ages and male piglets vs. solitary juveniles and adult males), from areas with different wild boar densities, different wild ruminant densities and different sarcoptic mange epidemiologic situations. The whole prevalence of antibodies against sarcoptic mange in the tested wild boars was 10.5%. No significant differences were found when comparing fenced and free ranging wild boars, males and females, gregarious vs. solitary individuals or among different ages. However, wild boar density was a relevant factor. In areas with a hunting bag of <1 wild boar/km2, considered as a low density of suids, the seroprevalence was 2.94%, but rose to 11.52% in high density districts, constituting a significant difference (p = 0.037). Low wild boar populations would act as a protective factor (OR 0.233; p = 0.049) against coming into contact with the mite. The wild ruminant densities or their sarcoptic mange status did not show any effect on wild boars seroprevalence against this disease. These results reinforce the suggested host-taxon Sarcoptes scabiei specificity and the independence of host-species foci.


Subject(s)
Scabies , Sus scrofa , Swine Diseases , Animals , Scabies/veterinary , Scabies/epidemiology , Sus scrofa/parasitology , Male , Female , Swine , Spain/epidemiology , Swine Diseases/epidemiology , Swine Diseases/parasitology , Animals, Wild/parasitology , Seroepidemiologic Studies , Sarcoptes scabiei , Goats , Enzyme-Linked Immunosorbent Assay/veterinary , Prevalence
19.
Can Vet J ; 65(7): 707-711, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38952762

ABSTRACT

A swine production system had 3 sections located a few kilometers apart. Sections A and C contained several thousand sows and nursery and finishing pigs. Section B, located between the other 2 sections, was the smallest and had 6 finishing sites and 2 sow sites. The entire system was infected with porcine reproductive and respiratory syndrome virus, Mycoplasma hyopneumoniae, and Actinobacillus pleuropneumoniae. Section B was depopulated, cleaned, disinfected, and repopulated with negative gilts. Despite extreme measures, recontamination occurred for each pathogen, with aerosol considered the most plausible contamination source.


Transmission suspectée d'agents pathogènes porcins par aérosol : un cas de terrainUn système de production porcine comportait 3 sections situées à quelques kilomètres l'une de l'autre. Les sections A et C contenaient plusieurs milliers de truies et de porcs en maternité et en finition. La section B, située entre les 2 autres sections, était la plus petite et comptait 6 sites de finition et 2 sites de truies. L'ensemble du système était infecté par le virus du syndrome reproducteur et respiratoire porcin, Mycoplasma hyopneumoniae et Actinobacillus pleuropneumoniae. La section B a été dépeuplée, nettoyée, désinfectée et repeuplée de cochettes négatives. Malgré des mesures extrêmes, une recontamination s'est produite pour chaque agent pathogène, les aérosols étant considérés comme la source de contamination la plus plausible.(Traduit par Dr Serge Messier).


Subject(s)
Actinobacillus pleuropneumoniae , Aerosols , Mycoplasma hyopneumoniae , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Swine , Swine Diseases/transmission , Swine Diseases/microbiology , Swine Diseases/virology , Mycoplasma hyopneumoniae/isolation & purification , Actinobacillus pleuropneumoniae/isolation & purification , Porcine respiratory and reproductive syndrome virus/isolation & purification , Actinobacillus Infections/veterinary , Actinobacillus Infections/transmission , Actinobacillus Infections/microbiology , Pneumonia of Swine, Mycoplasmal/transmission , Female , Porcine Reproductive and Respiratory Syndrome/transmission , Animal Husbandry
20.
Nat Commun ; 15(1): 5811, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987310

ABSTRACT

Extraintestinal Pathogenic Escherichia coli (ExPEC) pose a significant threat to human and animal health. However, the diversity and antibiotic resistance of animal ExPEC, and their connection to human infections, remain largely unexplored. The study performs large-scale genome sequencing and antibiotic resistance testing of 499 swine-derived ExPEC isolates from China. Results show swine ExPEC are phylogenetically diverse, with over 80% belonging to phylogroups B1 and A. Importantly, 15 swine ExPEC isolates exhibit genetic relatedness to human-origin E. coli strains. Additionally, 49 strains harbor toxins typical of enteric E. coli pathotypes, implying hybrid pathotypes. Notably, 97% of the total strains are multidrug resistant, including resistance to critical human drugs like third- and fourth-generation cephalosporins. Correspondingly, genomic analysis unveils prevalent antibiotic resistance genes (ARGs), often associated with co-transfer mechanisms. Furthermore, analysis of 20 complete genomes illuminates the transmission pathways of ARGs within swine ExPEC and to human pathogens. For example, the transmission of plasmids co-harboring fosA3, blaCTX-M-14, and mcr-1 genes between swine ExPEC and human-origin Salmonella enterica is observed. These findings underscore the importance of monitoring and controlling ExPEC infections in animals, as they can serve as a reservoir of ARGs with the potential to affect human health or even be the origin of pathogens infecting humans.


Subject(s)
Anti-Bacterial Agents , Escherichia coli Infections , Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Phylogeny , Swine Diseases , Animals , Swine , China/epidemiology , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/drug effects , Extraintestinal Pathogenic Escherichia coli/isolation & purification , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/veterinary , Swine Diseases/microbiology , Escherichia coli Proteins/genetics , Anti-Bacterial Agents/pharmacology , Humans , Drug Resistance, Multiple, Bacterial/genetics , Plasmids/genetics , Genome, Bacterial/genetics , Whole Genome Sequencing , Microbial Sensitivity Tests , Drug Resistance, Bacterial/genetics , beta-Lactamases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL