Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.304
Filtrer
1.
Microb Biotechnol ; 17(7): e14518, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953907

RÉSUMÉ

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.


Sujet(s)
Anticorps antiviraux , Bacillus subtilis , Virus de la diarrhée porcine épidémique , Spores bactériens , Virus de la diarrhée porcine épidémique/génétique , Virus de la diarrhée porcine épidémique/immunologie , Animaux , Bacillus subtilis/génétique , Bacillus subtilis/immunologie , Spores bactériens/génétique , Spores bactériens/immunologie , Souris , Anticorps antiviraux/sang , Suidae , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins antiviraux/administration et posologie , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/prévention et contrôle , Maladies des porcs/prévention et contrôle , Maladies des porcs/virologie , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Antigènes viraux/génétique , Antigènes viraux/immunologie , Administration par voie orale , Cytokines/métabolisme , Immunoglobuline G/sang , Souris de lignée BALB C , Femelle , Techniques d'exposition à la surface cellulaire , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
2.
Vet Microbiol ; 295: 110160, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964034

RÉSUMÉ

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.


Sujet(s)
Autophagie , Caspase-1 , Infections à Haemophilus , Haemophilus parasuis , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Inflammasomes/immunologie , Inflammasomes/métabolisme , Haemophilus parasuis/immunologie , Haemophilus parasuis/pathogénicité , Haemophilus parasuis/génétique , Caspase-1/métabolisme , Caspase-1/génétique , Infections à Haemophilus/médecine vétérinaire , Infections à Haemophilus/immunologie , Infections à Haemophilus/microbiologie , Suidae , Interleukine-18/métabolisme , Interleukine-18/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Souris
3.
Sci Rep ; 14(1): 15997, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987322

RÉSUMÉ

Pork is the most widely consumed meat on the planet, placing swine health as a critical factor for both the world economy and the food industry. Infectious diseases in pigs not only threaten these sectors but also raise zoonotic concerns, as pigs can act as "mixing vessels" for several animals and human viruses and can lead to the emergence of new viruses that are capable of infecting humans. Several efforts are ongoing to develop pig vaccines, albeit with limited success. This has been largely attributed to the complex nature of pig infections and incomplete understanding of the pig immune responses. Additionally, pig has been suggested to be a good experimental model to study viral infections (e.g., human influenza). Despite the significant importance of studying pig immunology for developing infection models, zoonosis, and the crucial need to develop better swine vaccines, there is still very limited information on the response of the swine adaptive immune system to several emerging pathogens. Particularly, very little is known about the pig B cell repertoire upon infection. Understanding the B cell repertoire is especially crucial towards designing better vaccines, predicting zoonosis and can provide insights into developing new diagnostic agents. Here, we developed methods for performing parallel single pig B cell (up to 10,000 B cells) global and immunoglobulin transcriptome sequencing. We then adapted a computational pipeline previously built for human/mouse sequences, to now analyze pig sequences. This allowed us to comprehensively map the B cell repertoire and get paired antibody sequences from pigs in a single parallel sequencing experiment. We believe that these approaches will have significant implications for swine diseases, particularly in the context of swine mediated zoonosis and swine and human vaccine development.


Sujet(s)
Lymphocytes B , Transcriptome , Animaux , Suidae , Lymphocytes B/immunologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Analyse sur cellule unique/méthodes , Analyse de profil d'expression de gènes/méthodes
4.
Viruses ; 16(6)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38932231

RÉSUMÉ

Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.


Sujet(s)
Betacoronavirus-1 , Cellules épithéliales , Analyse de profil d'expression de gènes , Interférons , Animaux , Suidae , Cellules épithéliales/virologie , Cellules épithéliales/immunologie , Interférons/génétique , Interférons/métabolisme , Interférons/immunologie , Betacoronavirus-1/immunologie , Betacoronavirus-1/génétique , Immunité innée , Réplication virale , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Infections à coronavirus/médecine vétérinaire , Cytokines/métabolisme , Cytokines/génétique , Cytokines/immunologie , Transcriptome , Muqueuse respiratoire/virologie , Muqueuse respiratoire/immunologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Maladies des porcs/génétique , Cellules cultivées , Deltacoronavirus (genre)
5.
Viruses ; 16(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38932247

RÉSUMÉ

Influenza A virus (IAV) infections in swine are usually subclinical, but they can reach high morbidity rates. The mortality rate is normally low. In this study, six vaccinated, spontaneously deceased sows revealed IAV infection and enhanced neutrophilic bronchopneumonia with unexpectedly large numbers of infiltrating eosinophils. The purpose of this study was to characterize these lung lesions with special emphasis on the phenotypes of inflammatory cells, the presence of eosinophilic peroxidase (EPO), and neutrophil extracellular traps (NETs). The number of Sirius red-stained eosinophils was significantly higher in the lungs of IAV-infected sows compared to healthy pigs, indicating a migration of eosinophils from blood vessels into the lung tissue stimulated by IAV infection. The detection of intra- and extracellular EPO in the lungs suggests its contribution to pulmonary damage. The presence of CD3+ T lymphocytes, CD20+ B lymphocytes, and Iba-1+ macrophages indicates the involvement of cell-mediated immune responses in disease progression. Furthermore, high numbers of myeloperoxidase-positive cells were detected. However, DNA-histone-1 complexes were reduced in IAV-infected sows, leading to the hypothesis that NETs are not formed in the IAV-infected sows. In conclusion, our findings in the lungs of IAV-infected vaccinated sows suggest the presence of so far unreported field cases of vaccine-associated enhanced respiratory disease.


Sujet(s)
Virus de la grippe A , Vaccins antigrippaux , Poumon , Infections à Orthomyxoviridae , Maladies des porcs , Animaux , Suidae , Poumon/anatomopathologie , Poumon/virologie , Poumon/immunologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Femelle , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Virus de la grippe A/immunologie , Épidémies de maladies/médecine vétérinaire , Granulocytes éosinophiles/immunologie , Pièges extracellulaires/immunologie , Vaccination/médecine vétérinaire , Eosinophil Peroxidase/métabolisme
6.
Open Vet J ; 14(5): 1224-1242, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38938443

RÉSUMÉ

Background: Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), is associated with high mortality and morbidity rates, especially in neonatal pigs. This has resulted in significant economic losses for the pig industry. PEDV genotype II-based vaccines were found to confer better immunity against both heterologous and homologous challenges; specifically, spike (S) proteins, which are known to play a significant role during infection, are ideal for vaccine development. Aim: This study aims to design a multi-epitope subunit vaccine targeting the S protein of the PEDV GIIa strain using an immunoinformatics approach. Methods: Various bioinformatics tools were used to predict HTL, CTL, and B-cell epitopes. The epitopes were connected using appropriate linkers and conjugated with the CTB adjuvant and M-ligand. The final multiepitope vaccine construct (fMEVc) was then docked to toll-like receptor 4 (TLR4). The stability of the fMEVc-TLR4 complex was then simulated using GROMACS. C-immsim was then used to predict the in vitro immune response of the fMEVc. Results: Six epitopes were predicted to induce antibody production, ten epitopes were predicted to induce CTL responses, and four epitopes were predicted to induce HTL responses. The assembled epitopes conjugated with the CTB adjuvant and M-ligand, fMEVc, is antigenic, non-allergenic, stable, and soluble. The construct showed a favorable binding affinity for TLR4, and the protein complex was shown to be stable through molecular dynamics simulations. A robust immune response was induced after immunization, as demonstrated through immune stimulation. Conclusion: In conclusion, the multi-epitope subunit vaccine construct for PEDV designed in this study exhibits promising antigenicity, stability, and immunogenicity, eliciting robust immune responses and suggesting its potential as a candidate for further vaccine development.


Sujet(s)
Biologie informatique , Infections à coronavirus , Virus de la diarrhée porcine épidémique , Glycoprotéine de spicule des coronavirus , Maladies des porcs , Vaccins sous-unitaires , Vaccins antiviraux , Animaux , Virus de la diarrhée porcine épidémique/immunologie , Vaccins sous-unitaires/immunologie , Suidae , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Vaccins antiviraux/immunologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie , Génotype , Épitopes/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes B/immunologie , Simulation de docking moléculaire ,
7.
Sci Rep ; 14(1): 14586, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918457

RÉSUMÉ

Natural killer (NK) cells play a key role in defense against Salmonella infections during the early phase of infection. Our previous work showed that the excretory/secretory products of Ascaris suum repressed NK activity in vitro. Here, we asked if NK cell functionality was influenced in domestic pigs during coinfection with Ascaris and Salmonella enterica serotype Typhimurium. Ascaris coinfection completely abolished the IL-12 and IL-18 driven elevation of IFN-γ production seen in CD16 + CD8α + perforin + NK cells of Salmonella single-infected pigs. Furthermore, Ascaris coinfection prohibited the Salmonella-driven rise in NK perforin levels and CD107a surface expression. In line with impaired effector functions, NK cells from Ascaris-single and coinfected pigs displayed elevated expression of the inhibitory KLRA1 and NKG2A receptors genes, contrasting with the higher expression of the activating NKp46 and NKp30 receptors in NK cells during Salmonella single infection. These differences were accompanied by the highly significant upregulation of T-bet protein expression in NK cells from Ascaris-single and Ascaris/Salmonella coinfected pigs. Together, our data strongly indicate a profound repression of NK functionality by an Ascaris infection which may hinder infected individuals from adequately responding to a concurrent bacterial infection.


Sujet(s)
Ascaridiose , Co-infection , Cellules tueuses naturelles , Maladies des porcs , Animaux , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Ascaridiose/immunologie , Ascaridiose/médecine vétérinaire , Ascaridiose/parasitologie , Co-infection/immunologie , Co-infection/microbiologie , Co-infection/parasitologie , Suidae , Maladies des porcs/parasitologie , Maladies des porcs/immunologie , Maladies des porcs/microbiologie , Salmonelloses animales/immunologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Ascaris suum/immunologie , Interféron gamma/métabolisme , Perforine/métabolisme , Interleukine-12/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Interleukine-18/métabolisme
8.
Anim Sci J ; 95(1): e13964, 2024.
Article de Anglais | MEDLINE | ID: mdl-38831612

RÉSUMÉ

This study evaluated the effects of supplementation with Antrodia cinnamomea mycelium by-product (ACBP) on growth performance and immune response in weaning piglets. Total available content and antioxidant capacity of ACBP were determined. Ninety-six black pigs were randomly distributed to 24 pens. Study compared four groups which were supplemented with ACBP at 0%, 2.5%, 5%, or 10% for 6 weeks after weaning at 4 weeks. Results showed that ACBP on total phenolic, total flavonoid, and total triterpenoids contents were 13.68 mg GAE/g DW, 1.67 µg QE/g DW, and 15.6 mg/g, respectively. Weaning piglets fed 2.5% ACBP showed a significant decreased body weight gain compared with those supplemented with 5% ACBP, 10% ACBP, and control groups. Results showed that all ACBP groups increased the villi height of jejunum significantly. Incidence of diarrhea in 11 weeks with supplementation with 5% and 10% ACBP diets were lower than in control group. The 10% ACBP group showed significantly lower expression of immune response genes (IL-1ß, IL-6, IL-8, TNF-α, and IFN-γ) than the 2.5% and 5% ACBP groups. Based on results, dietary supplementation with 10% ACBP did not significantly affect body weight but could decrease piglet diarrhea condition and expression of IL-1ß and IL-6 genes.


Sujet(s)
Aliment pour animaux , Antioxydants , Régime alimentaire , Compléments alimentaires , Mycelium , Sevrage , Prise de poids , Animaux , Suidae/croissance et développement , Suidae/immunologie , Prise de poids/effets des médicaments et des substances chimiques , Régime alimentaire/médecine vétérinaire , Antioxydants/métabolisme , Diarrhée/médecine vétérinaire , Triterpènes/pharmacologie , Triterpènes/administration et posologie , Expression des gènes/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Jéjunum/métabolisme , Phénols/analyse , Phénomènes physiologiques nutritionnels chez l'animal , Maladies des porcs/microbiologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Polyporales/composition chimique
9.
BMC Vet Res ; 20(1): 239, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831363

RÉSUMÉ

The porcine epidemic diarrhea virus (PEDV) infection inflicted substantial economic losses upon the global pig-breeding industry. This pathogen can infect all pigs and poses a particularly high fatality risk for suckling piglets. The S1 subunit of spike protein is a crucial target protein for inducing the particularly neutralizing antibodies that can intercept the virus-host interaction and neutralize virus infectivity. In the present study, the HEK293F eukaryotic expression system was successfully utilized to express and produce recombinant S1 protein. Through quantitative analysis, five monoclonal antibodies (mAbs) specifically targeting the recombinant S1 protein of PEDV were developed and subsequently evaluated using enzyme-linked immunosorbent assay (ELISA), indirect immunofluorescence assay (IFA), and flow cytometry assay (FCA). The results indicate that all five mAbs belong to the IgG1 isotype, and their half-maximal effective concentration (EC50) values measured at 84.77, 7.42, 0.89, 14.64, and 7.86 pM. All these five mAbs can be utilized in ELISA, FCA, and IFA for the detection of PEDV infection. MAb 5-F9 exhibits the highest sensitivity to detect as low as 0.3125 ng/mL of recombinant PEDV-S1 protein in ELISA, while only 0.096 ng/mL of mAb 5-F9 is required to detect PEDV in FCA. The results from antigen epitope analysis indicated that mAb 8-G2 is the sole antibody capable of recognizing linear epitopes. In conclusion, this study has yielded a highly immunogenic S1 protein and five high-affinity mAbs specifically targeting the S1 protein. These findings have significant implications for early detection of PEDV infection and provide a solid foundation for further investigation into studying virus-host interactions.


Sujet(s)
Anticorps monoclonaux , Infections à coronavirus , Test ELISA , Virus de la diarrhée porcine épidémique , Glycoprotéine de spicule des coronavirus , Virus de la diarrhée porcine épidémique/immunologie , Anticorps monoclonaux/immunologie , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Suidae , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Test ELISA/médecine vétérinaire , Anticorps antiviraux/immunologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Cellules HEK293 , Humains , Protéines recombinantes/immunologie , Souris de lignée BALB C , Souris , Technique d'immunofluorescence indirecte/médecine vétérinaire
10.
Front Immunol ; 15: 1361323, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835763

RÉSUMÉ

Introduction: Swine influenza viruses (SIVs) pose significant economic losses to the pig industry and are a burden on global public health systems. The increasing complexity of the distribution and evolution of different serotypes of influenza strains in swine herds escalates the potential for the emergence of novel pandemic viruses, so it is essential to develop new vaccines based on swine influenza. Methods: Here, we constructed a self-assembling ferritin nanoparticle vaccine based on the hemagglutinin (HA) extracellular domain of swine influenza A (H1N1) virus using insect baculovirus expression vector system (IBEVS), and after two immunizations, the immunogenicities and protective efficacies of the HA-Ferritin nanoparticle vaccine against the swine influenza virus H1N1 strain in mice and piglets were evaluated. Results: Our results demonstrated that HA-Ferritin nanoparticle vaccine induced more efficient immunity than traditional swine influenza vaccines. Vaccination with the HA-Ferritin nanoparticle vaccine elicited robust hemagglutinin inhibition titers and antigen-specific IgG antibodies and increased cytokine levels in serum. MF59 adjuvant can significantly promote the humoral immunity of HA-Ferritin nanoparticle vaccine. Furthermore, challenge tests showed that HA-Ferritin nanoparticle vaccine conferred full protection against lethal challenge with H1N1 virus and significantly decreased the severity of virus-associated lung lesions after challenge in both BALB/c mice and piglets. Conclusion: Taken together, these results indicate that the hemagglutinin extracellular-based ferritin nanoparticle vaccine may be a promising vaccine candidate against SIVs infection.


Sujet(s)
Anticorps antiviraux , Ferritines , Glycoprotéine hémagglutinine du virus influenza , Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Nanoparticules , Infections à Orthomyxoviridae , Animaux , Sous-type H1N1 du virus de la grippe A/immunologie , Ferritines/immunologie , Vaccins antigrippaux/immunologie , Suidae , Souris , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie , Femelle ,
11.
Front Immunol ; 15: 1381026, 2024.
Article de Anglais | MEDLINE | ID: mdl-38919620

RÉSUMÉ

Introduction: Porcine deltacoronavirus (PDCoV) is a zoonotic pathogen with a global distribution, capable of infecting both pigs and humans. To mitigate the risk of cross-species transmission and potential outbreaks, it is crucial to characterize novel antiviral genes, particularly those from human hosts. Methods: This research used HIEC-6 to investigate PDCoV infection. HIEC-6 cells were infected with PDCoV. Samples were collected 48 h postinfection for proteomic analysis. Results: We discovered differential expression of MRPS6 gene at 48 h postinfection with PDCoV in HIEC-6 cells. The gene expression initially increased but then decreased. To further explore the role of MRPS6 in PDCoV infection, we conducted experiments involving the overexpression and knockdown of this gene in HIEC-6 and Caco2 cells, respectively. Our findings revealed that overexpression of MRPS6 significantly inhibited PDCoV infection in HIEC-6 cells, while knockdown of MRPS6 in Caco2 cells led to a significant increase of virus titer. Furthermore, we investigated the correlation between PDCoV infection and the expression of MRPS6. Subsequent investigations demonstrated that MRPS6 exerted an augmentative effect on the production of IFN-ß through interferon pathway activation, consequently impeding the progression of PDCoV infection in cellular systems. In conclusion, this study utilized proteomic analysis to investigate the differential protein expression in PDCoV-infected HIEC-6 cells, providing evidence for the first time that the MRPS6 gene plays a restrictive role in PDCoV virus infection. Discussion: Our findings initially provide the validation of MRPS6 as an upstream component of IFN-ß pathway, in the promotion of IRF3, IRF7, STAT1, STAT2 and IFN-ß production of HIEC-6 via dual-activation from interferon pathway.


Sujet(s)
Deltacoronavirus (genre) , Humains , Animaux , Suidae , Deltacoronavirus (genre)/physiologie , Deltacoronavirus (genre)/génétique , Cellules Caco-2 , Infections à coronavirus/virologie , Infections à coronavirus/immunologie , Lignée cellulaire , Interactions hôte-pathogène/immunologie , Protéomique/méthodes , Transduction du signal , Maladies des porcs/virologie , Maladies des porcs/immunologie
12.
Methods Mol Biol ; 2815: 115-119, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884914

RÉSUMÉ

Streptococcus suis is a swine bacterial pathogen that predominantly causes disease in weaned piglets characterized by swelling of joints, arthritis, septicemia, meningitis, and sudden death. Intravenous, intramuscular, intraperitoneal, and intranasal infection models were developed to study the bacterial pathogenicity and efficacy of vaccines and various therapeutics. The selection of the appropriate infection model is a critical step in any study, as it may impact the outcomes of the study. Here we describe a method for infecting weaned piglets with S. suis using intraperitoneal route as a reliable, consistent, and reproducible animal model to evaluate vaccine protection against systemic bacterial infection.


Sujet(s)
Modèles animaux de maladie humaine , Infections à streptocoques , Streptococcus suis , Maladies des porcs , Animaux , Suidae , Streptococcus suis/pathogénicité , Infections à streptocoques/médecine vétérinaire , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Injections péritoneales
13.
Methods Mol Biol ; 2815: 131-142, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884916

RÉSUMÉ

Streptococcus suis is a bacterial pathogen that can cause significant economic losses in the swine industry due to high morbidity and mortality rates in infected animals. Vaccination with bacterins, which consist of inactivated bacteria and adjuvants to enhance the pig's immune response, is an effective approach to control S. suis infections in piglets. Here we provide a description of S. suis bacterins and the methods for vaccine preparation. Moreover, this chapter also describes the addition of recombinant Sao (rSao-L) protein to the S. suis bacterin, aiming to enhance the efficacy of the bacterins against S. suis in piglets. Furthermore, the methods for evaluating the immune response elicited by the bacterins are also covered in this chapter.


Sujet(s)
Streptococcus suis , Animaux , Suidae , Streptococcus suis/immunologie , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/médecine vétérinaire , Maladies des porcs/microbiologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Vaccination/méthodes , Vaccins antibactériens/immunologie , Adjuvants immunologiques/pharmacologie , Anticorps antibactériens/immunologie , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , Vaccins antistreptococciques/immunologie , Vaccins antistreptococciques/administration et posologie
14.
Nat Commun ; 15(1): 5025, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871701

RÉSUMÉ

Influenza A viruses in swine have considerable genetic diversity and continue to pose a pandemic threat to humans due to a potential lack of population level immunity. Here we describe a pipeline to characterize and triage influenza viruses for their pandemic risk and examine the pandemic potential of two widespread swine origin viruses. Our analysis reveals that a panel of human sera collected from healthy adults in 2020 has no cross-reactive neutralizing antibodies against a α-H1 clade strain (α-swH1N2) but do against a γ-H1 clade strain. The α-swH1N2 virus replicates efficiently in human airway cultures and exhibits phenotypic signatures similar to the human H1N1 pandemic strain from 2009 (H1N1pdm09). Furthermore, α-swH1N2 is capable of efficient airborne transmission to both naïve ferrets and ferrets with prior seasonal influenza immunity. Ferrets with H1N1pdm09 pre-existing immunity show reduced α-swH1N2 viral shedding and less severe disease signs. Despite this, H1N1pdm09-immune ferrets that became infected via the air can still onward transmit α-swH1N2 with an efficiency of 50%. These results indicate that this α-swH1N2 strain has a higher pandemic potential, but a moderate level of impact since there is reduced replication fitness and pathology in animals with prior immunity.


Sujet(s)
Furets , Sous-type H1N1 du virus de la grippe A , Sous-type H1N2 du virus de la grippe A , Grippe humaine , Infections à Orthomyxoviridae , Pandémies , Animaux , Furets/virologie , Humains , Suidae , Grippe humaine/virologie , Grippe humaine/épidémiologie , Grippe humaine/immunologie , Grippe humaine/sang , Grippe humaine/transmission , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/transmission , Infections à Orthomyxoviridae/sang , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/isolement et purification , Sous-type H1N2 du virus de la grippe A/génétique , Sous-type H1N2 du virus de la grippe A/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Maladies des porcs/virologie , Maladies des porcs/épidémiologie , Maladies des porcs/immunologie , Maladies des porcs/transmission , Maladies des porcs/sang , Femelle , Excrétion virale , Mâle , Adulte , Réplication virale
15.
Nat Commun ; 15(1): 5330, 2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38909062

RÉSUMÉ

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that has recently been detected in humans. Despite this zoonotic concern, the antigenic structure of PDCoV remains unknown. The virus relies on its spike (S) protein for cell entry, making it a prime target for neutralizing antibodies. Here, we generate and characterize a set of neutralizing antibodies targeting the S protein, shedding light on PDCoV S interdomain crosstalk and its vulnerable sites. Among the four identified antibodies, one targets the S1A domain, causing local and long-range conformational changes, resulting in partial exposure of the S1B domain. The other antibodies bind the S1B domain, disrupting binding to aminopeptidase N (APN), the entry receptor for PDCoV. Notably, the epitopes of these S1B-targeting antibodies are concealed in the prefusion S trimer conformation, highlighting the necessity for conformational changes for effective antibody binding. The binding footprint of one S1B binder entirely overlaps with APN-interacting residues and thus targets a highly conserved epitope. These findings provide structural insights into the humoral immune response against the PDCoV S protein, potentially guiding vaccine and therapeutic development for this zoonotic pathogen.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Deltacoronavirus (genre) , Épitopes , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/composition chimique , Animaux , Anticorps neutralisants/immunologie , Suidae , Anticorps antiviraux/immunologie , Épitopes/immunologie , Humains , Deltacoronavirus (genre)/immunologie , Deltacoronavirus (genre)/métabolisme , Antigènes CD13/métabolisme , Antigènes CD13/immunologie , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Domaines protéiques , Liaison aux protéines , Maladies des porcs/virologie , Maladies des porcs/immunologie , Cellules HEK293
16.
Vet Res ; 55(1): 79, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886840

RÉSUMÉ

Porcine deltacoronavirus (PDCoV) is an enteropathogenic coronavirus that has been reported to use various strategies to counter the host antiviral innate immune response. The cGAS-STING signalling pathway plays an important role in antiviral innate immunity. However, it remains unclear whether PDCoV achieves immune evasion by regulating the cGAS-STING pathway. Here, we demonstrated that the nonstructural protein 2 (nsp2) encoded by PDCoV inhibits cGAS-STING-mediated type I and III interferon (IFN) responses via the regulation of porcine STING (pSTING) stability. Mechanistically, ectopically expressed PDCoV nsp2 was found to interact with the N-terminal region of pSTING. Consequently, pSTING was degraded through K48-linked ubiquitination and the proteasomal pathway, leading to the disruption of cGAS-STING signalling. Furthermore, K150 and K236 of pSTING were identified as crucial residues for nsp2-mediated ubiquitination and degradation. In summary, our findings provide a basis for elucidating the immune evasion mechanism of PDCoV and will contribute to the development of targets for anti-coronavirus drugs.


Sujet(s)
Deltacoronavirus (genre) , Protéines virales non structurales , Animaux , Suidae , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Deltacoronavirus (genre)/génétique , Deltacoronavirus (genre)/physiologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/virologie , Infections à coronavirus/immunologie , Infections à coronavirus/métabolisme , Interféron de type I/métabolisme , Interféron de type I/génétique , Immunité innée , Cellules HEK293 , Échappement immunitaire , Ubiquitination
17.
Vet Microbiol ; 295: 110151, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870752

RÉSUMÉ

Porcine circovirus type 2 (PCV2) stands as a predominant etiological agent in porcine circovirus-associated diseases. To manage the spread of the disease, it is necessary to develop a next-generation vaccine expressing PCV2 antigens that target the prevailing genotype such as PCV2d. A bacterial-mediated vaccine delivery by live-attenuated Salmonella has attracted interest for its low-cost production and highly effective vaccine delivery. Thus, in this study, we utilized the advantages of the Salmonella-mediated vaccine delivery by cloning PCV2d cap and rep into a eukaryotic expression plasmid pJHL204 and electroporation into an engineered live-attenuated Salmonella Typhimurium JOL2500 (Δlon, ΔcpxR, ΔsifA, Δasd). The eukaryotic antigen expression by JOL2995 (p204:cap) and JOL2996 (p204:rep) was confirmed in vitro and in vivo which showed efficient antigen delivery. Furthermore, vaccination of mice model with the vaccine candidates elicited humoral and cell-mediated immune responses as depicted by high levels of PCV2-specific antibodies, CD4+ and CD8+ T cells, and neutralizing antibodies, especially by JOL2995 (p204:cap) which correlated with the significant decrease in the viral load in PCV2d-challenged mice. Interestingly, JOL2996 (p204:rep) may not have elicited high levels of neutralizing antibodies and protective efficacy, but it elicited considerably higher cell-mediated immune responses. This study demonstrated Salmonella-mediated vaccine delivery system coupled with the eukaryotic expression vector can efficiently deliver and express the target PCV2d antigens for strong induction of immune response and protective efficacy in mice model, further supporting the potential application of the Salmonella-mediated vaccine delivery system as an effective novel approach in vaccine strategies for PCV2d.


Sujet(s)
Infections à Circoviridae , Circovirus , Vecteurs génétiques , Salmonella typhimurium , Vaccins antiviraux , Animaux , Circovirus/immunologie , Circovirus/génétique , Souris , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Infections à Circoviridae/prévention et contrôle , Infections à Circoviridae/médecine vétérinaire , Infections à Circoviridae/immunologie , Suidae , Antigènes viraux/immunologie , Antigènes viraux/génétique , Souris de lignée BALB C , Anticorps antiviraux/sang , Femelle , Anticorps neutralisants/sang , Protéines de capside/immunologie , Protéines de capside/génétique , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie
18.
Methods Mol Biol ; 2813: 145-165, 2024.
Article de Anglais | MEDLINE | ID: mdl-38888777

RÉSUMÉ

As an alternative to traditional serological markers, that is, antibodies, for serum-based specific diagnosis of infections, circulating non-antibody markers may be used to monitor active disease. Acute phase proteins (APPs) are a prominent class of such markers widely used for diagnosing ongoing inflammation and infection. In this chapter, basic theoretical and practical considerations on developing APP assays and using APPs as markers of ongoing infection are presented with a specific focus on intracellular infections in pigs. Examples on APP-based monitoring of infection in pigs with viruses such as porcine respiratory and reproductive syndrome virus (PRRSV), porcine endemic diarrhea virus (PEDV), and influenza A virus (IAV), as well as intracellular bacteria (Lawsonia intracellularis) and the protozoan intracellular parasites Toxoplasma gondii and Cryptosporidium parvum are presented, with an emphasis on major pig APPs C-reactive protein (CRP), haptoglobin, serum amyloid A (SAA), and pig major acute phase protein (pig-MAP). The performance of these APPs as biomarkers in a range of experimental infection studies in pigs is described as examples on their use for estimating the severity of infection, vaccine efficacy, herd health characterization, and differential diagnosis.


Sujet(s)
Protéine de la phase aigüe , Marqueurs biologiques , Maladies des porcs , Animaux , Suidae , Protéine de la phase aigüe/métabolisme , Protéine de la phase aigüe/immunologie , Marqueurs biologiques/sang , Maladies des porcs/diagnostic , Maladies des porcs/virologie , Maladies des porcs/immunologie , Maladies des porcs/parasitologie , Maladies des porcs/sang
19.
Front Immunol ; 15: 1396446, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799456

RÉSUMÉ

Ascaris spp. undergo extensive migration within the body before establishing patent infections in the small intestinal tract of humans and pigs. However, whether larval migration is critical for inducing efficient type 2 responses remains poorly understood. Therefore, we investigated systemic versus local adaptive immune responses along the hepato-tracheal migration of Ascaris suum during primary, single infections in conventionally raised pigs. Neither the initial invasion of gut tissue nor migration through the liver resulted in discernable Th2 cell responses. In contrast, lung-stage larvae elicited a Th2-biased pulmonary response, which declined after the larvae had left the lungs. In the small intestine, we observed an accumulation of Th2 cells upon the arrival of fourth-stage larvae (L4) to the small intestinal lumen. In parallel, we noticed robust and increasing Th1 responses in circulation, migration-affected organs, and draining lymph nodes. Phenotypic analysis of CD4+ T cells specifically recognizing A. suum antigens in the circulation and lung tissue of infected pigs confirmed that the majority of Ascaris-specific T cells produced IL-4 (Th2) and, to a much lesser extent, IL-4/IFN-g (Th2/1 hybrids) or IFN-g alone (Th1). These data demonstrate that lung-stage but not the early liver-stage larvae lead to a locally restricted Th2 response. Significant Th2 cell accumulation in the small intestine occurs only when L4 complete the body migration. In addition, Th2 immunity seems to be hampered by the concurrent, nonspecific Th1 bias in growing pigs. Together, the late onset of Th2 immunity at the site of infection and the Th1-biased systemic immunity likely enable the establishment of intestinal infections by sufficiently large L4 stages and pre-adult worms, some of which resist expulsion mechanisms.


Sujet(s)
Ascaridiose , Ascaris suum , Lymphocytes auxiliaires Th1 , Lymphocytes auxiliaires Th2 , Animaux , Ascaris suum/immunologie , Ascaridiose/immunologie , Ascaridiose/parasitologie , Lymphocytes auxiliaires Th2/immunologie , Suidae , Lymphocytes auxiliaires Th1/immunologie , Maladies des porcs/immunologie , Maladies des porcs/parasitologie , Poumon/immunologie , Poumon/parasitologie , Larve/immunologie , Cytokines/métabolisme
20.
Viruses ; 16(5)2024 04 24.
Article de Anglais | MEDLINE | ID: mdl-38793542

RÉSUMÉ

The suboptimal performance of rotavirus (RV) vaccines in developing countries and in animals necessitates further research on the development of novel therapeutics and control strategies. To initiate infection, RV interacts with cell-surface O-glycans, including histo-blood group antigens (HBGAs). We have previously demonstrated that certain non-pathogenic bacteria express HBGA- like substances (HBGA+) capable of binding RV particles in vitro. We hypothesized that HBGA+ bacteria can bind RV particles in the gut lumen protecting against RV species A (RVA), B (RVB), and C (RVC) infection in vivo. In this study, germ-free piglets were colonized with HBGA+ or HBGA- bacterial cocktail and infected with RVA/RVB/RVC of different genotypes. Diarrhea severity, virus shedding, immunoglobulin A (IgA) Ab titers, and cytokine levels were evaluated. Overall, colonization with HBGA+ bacteria resulted in reduced diarrhea severity and virus shedding compared to the HBGA- bacteria. Consistent with our hypothesis, the reduced severity of RV disease and infection was not associated with significant alterations in immune responses. Additionally, colonization with HBGA+ bacteria conferred beneficial effects irrespective of the piglet HBGA phenotype. These findings are the first experimental evidence that probiotic performance in vivo can be improved by including HBGA+ bacteria, providing decoy epitopes for broader/more consistent protection against diverse RVs.


Sujet(s)
Antigènes de groupe sanguin , Axénie , Infections à rotavirus , Rotavirus , Animaux , Infections à rotavirus/immunologie , Infections à rotavirus/virologie , Suidae , Rotavirus/immunologie , Antigènes de groupe sanguin/métabolisme , Antigènes de groupe sanguin/immunologie , Diarrhée/virologie , Diarrhée/microbiologie , Diarrhée/prévention et contrôle , Maladies des porcs/virologie , Maladies des porcs/microbiologie , Maladies des porcs/immunologie , Excrétion virale , Bactéries/classification , Immunoglobuline A/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Cytokines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...