Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 771
Filter
1.
mSphere ; 9(9): e0023124, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39194201

ABSTRACT

Glaesserella parasuis is a commensal bacterial organism found in the upper respiratory tract of healthy pigs and the etiological agent of Glässer's disease, which causes severe economic losses in the swine industry. This study aimed to better understand the epidemiological characteristics of this opportunistic pathogen. We investigated the prevalence and distribution of sequence types (STs), serovars, antimicrobial resistance genes (ARGs), and potential virulence factors (VFs) in 764 G. parasuis isolates collected from diseased and healthy pigs from 19 countries, including China. Multilocus sequence typing showed a high degree of variation with 334 STs, of which 93 were not previously recognized. Phylogenetic analysis revealed two major clades distinguished by isolation year, source, country, and serovar. The dominant serovars of G. parasuis were serovars 4 (19.50%), 7 (15.97%), 5/12 (13.87%), and 13 (12.30%). Serovar 7 gradually became one of the dominant serovars in G. parasuis with more VFs and fewer ARGs. Serovars 4 and 5/12 were the most frequent serovars in diseased pigs, whereas serovars 2, 8, and 11 were predominant in healthy pigs. Serovars 7 and 13 possessed more VFs than the other serovars. This study provides novel insights into the global prevalence and epidemiology of G. parasuis and valuable clues for further investigation into the pathogenicity of G. parasuis, which will facilitate the development of effective vaccines.IMPORTANCEGlaesserella parasuis is a clinically important gram-negative opportunistic pathogen, which causes serious financial losses in swine industry on a global scale. No vaccine is known that provides cross-protection against all 15 serovars; furthermore, the correlation between serovar and virulence is largely unknown. This study provides a large number of sequenced strains in 19 countries and compares the genomic diversity of G. parasuis between diseased and healthy pigs. We found a slight change in the dominant serovar of G. parasuis in the world, with serovar 7 gradually emerging as one of the predominant serovars. The observed higher average number of VFs in this particular serovar strain challenges the previously held notion that serovar 7 is non-virulent, indicating a more complex virulence landscape than previously understood. Our analysis indicating that six ARGs [tet(B), sul2, aph(3')-Ia, aph (6)-Id, blaROB-1, and aph(3'')-Ib] are likely to be transmitted horizontally in their entirety. By analyzing VFs, we provided an improved understanding of the virulence of G. parasuis, and these key findings suggest that vaccine development will be challenging.


Subject(s)
Genetic Variation , Haemophilus Infections , Multilocus Sequence Typing , Phylogeny , Serogroup , Swine Diseases , Virulence Factors , Animals , Swine , Swine Diseases/microbiology , Swine Diseases/epidemiology , Virulence Factors/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Haemophilus Infections/epidemiology , Haemophilus parasuis/genetics , Haemophilus parasuis/classification , Haemophilus parasuis/isolation & purification , Haemophilus parasuis/pathogenicity , Pasteurellaceae/genetics , Pasteurellaceae/classification , Pasteurellaceae/isolation & purification , Pasteurellaceae/pathogenicity , Genome, Bacterial , China/epidemiology , Genomics , Drug Resistance, Bacterial/genetics
2.
Genes (Basel) ; 15(8)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39202454

ABSTRACT

Glaesserella parasuis (GPS) can cause severe systemic inflammation in pigs, resulting in huge economic losses to the pig industry. At present, no effective method is available for the prevention and control of GPS infection. Molecular breeding for disease resistance is imminent, but disease-resistance genes have not been identified. To study the mechanism of systemic acute inflammation caused by GPS, we established three in vitro infection models (3D4/21 cells, PK15 cells, and PAVEC cells) according to its infection path. There was no significant difference in apoptosis among the three kinds of cells after 12 h of continuous GPS stimulation, while inflammatory factors were significantly upregulated. Subsequent transcriptome analysis revealed 1969, 1207, and 3564 differentially expressed genes (DEGs) in 3D4/21 cells, PK15 cells, and PAVEC cells, respectively, after GPS infection. Many of the DEGs were predicted to be associated with inflammatory responses (C3, CD44, etc.); cell proliferation, growth and apoptosis; gene expression; and protein phosphorylation. Key signaling pathways, including S100 family signaling, bacteria and virus recognition, and pathogen-induced cytokine storm signaling, were enriched based on Ingenuity Pathway Analysis (IPA). Furthermore, a total of three putative transmembrane receptors and two putative G-protein-coupled receptors, namely F3, ICAM1, PLAUR, ACKR3, and GPRC5A, were identified by IPA among the three types of cells. ACKR3 and GPRC5A play pivotal roles in bacterial adhesion, invasion, host immune response and inflammatory response through the S100 family signaling pathway. Our findings provide new insights into the pathological mechanisms underlying systemic inflammation caused by GPS infection in pigs, and they lay a foundation for further research on disease-resistance breeding to GPS.


Subject(s)
Haemophilus parasuis , Inflammation , Signal Transduction , Swine Diseases , Animals , Swine , Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Signal Transduction/genetics , Inflammation/genetics , Inflammation/microbiology , Swine Diseases/microbiology , Swine Diseases/genetics , Swine Diseases/immunology , Haemophilus Infections/veterinary , Haemophilus Infections/genetics , Haemophilus Infections/microbiology , Haemophilus Infections/immunology , Transcriptome/genetics , Gene Expression Profiling , Cell Line , Apoptosis/genetics
3.
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
4.
Vet Res ; 55(1): 96, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075542

ABSTRACT

Glaesserella parasuis (G. parasuis) induces vascular damage and systemic inflammation. However, the mechanism by which it causes vascular damage is currently unclear. Baicalin has important anti-inflammatory, antibacterial and immunomodulatory functions. In this study, we explored the ability of baicalin and probenecid to protect against G. parasuis challenge in a piglet model. Sixty piglets were randomly divided into a control group; an infection group; a probenecid group; and 25 mg/kg, 50 mg/kg and 100 mg/kg baicalin groups. The probenecid group and the 25 mg/kg, 50 mg/kg and 100 mg/kg baicalin groups were injected intramuscularly with 20 mg/kg body weight (BW) probenecid and 25 mg/kg BW, 50 mg/kg BW and 100 mg/kg BW baicalin, respectively. All piglets except those from the control group were injected intraperitoneally with 1 × 108 CFU of G. parasuis. The control group was injected intraperitoneally with TSB. The results showed baicalin and probenecid protected piglets against G. parasuis challenge, improved body weight and decreased temperature changes in piglets. Baicalin and probenecid attenuated IL-1ß, IL-10, IL-18, TNF-α and IFN-γ mRNA levels in the blood for 48 h, inhibited the production of the nucleosides ATP, ADP, AMP and UMP from 24 to 72 h, reduced Panx-1/P2Y6/P2X7 expression, weakened NF-kB, AP-1, NLRP3/Caspase-1 and ROCK/MLCK/MLC signalling activation, and upregulated VE-cadherin expression in the blood vessels of piglets challenged with G. parasuis. Baicalin and probenecid alleviated pathological tissue damage in piglets induced by G. parasuis. Our results might provide a promising strategy to control and treat G. parasuis infection in the clinical setting.


Subject(s)
Flavonoids , Haemophilus parasuis , Probenecid , Swine Diseases , Animals , Probenecid/pharmacology , Flavonoids/pharmacology , Flavonoids/administration & dosage , Swine , Swine Diseases/microbiology , Swine Diseases/prevention & control , Haemophilus parasuis/drug effects , Haemophilus Infections/veterinary , Haemophilus Infections/prevention & control
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.
Comp Immunol Microbiol Infect Dis ; 111: 102214, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002176

ABSTRACT

MicroRNAs (miRNAs) have been shown to play an important regulatory role in the process of pathogenic infection. However, the miRNAs that regulate the pathogenic process of G. parasuis and their functions are still unknown. Here, high-throughput sequencing was used to quantify the expression of miRNA in piglet lung tissue after G. parasuis XX0306 strain infection. A total of 25 differentially expressed microRNAs (DEmiRNAs) were identified. GO and KEGG pathway enrichment analysis showed that many of the functions of genes that may be regulated by DEmiRNA are related to inflammatory response and immune regulation. Further studies found that ssc-miR-135 may promote the expression of inflammatory factors through NF-κB signaling pathway. Whereas, ssc-miR-155-3p inhibited the inflammatory response induced by G. parasuis, and its regulatory mechanism remains to be further investigated. This study provides a valuable reference for revealing the regulatory effects of miRNAs on the pathogenesis of G. parasuis. DATA AVAILABILITY: The datasets generated during the current study are not publicly available due to this study is currently in the ongoing research stage, and some of the data cannot be made public sooner yet, but are available from the corresponding author on reasonable request.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Inflammation , Lung , MicroRNAs , Swine Diseases , Animals , MicroRNAs/genetics , Swine , Lung/microbiology , Lung/immunology , Swine Diseases/microbiology , Swine Diseases/genetics , Swine Diseases/immunology , Inflammation/genetics , Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Haemophilus Infections/veterinary , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus Infections/genetics , Gene Expression Profiling , NF-kappa B/metabolism , NF-kappa B/genetics , Signal Transduction , High-Throughput Nucleotide Sequencing , Gene Expression Regulation , Transcriptome , Metastrongyloidea/genetics
7.
Vet Res ; 55(1): 93, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075605

ABSTRACT

Glaesserella parasuis is usually a benign swine commensal in the upper respiratory tract, but virulent strains can cause systemic infection characterized by pneumonia, meningitis, and fibrinous polyserositis. The intensive pulmonary inflammatory response following G. parasuis infection is the main cause of lung injury and death in pigs. Vaccination has failed to control the disease due to the lack of extended cross-protection. Accumulating evidence indicates that the heme-binding protein A (HbpA) is a potential virulence determinant and a promising antigen candidate for the development of a broader range of vaccines. However, it is not yet known whether HbpA contributes to G. parasuis virulence or has any potential immune protective effects against G. parasuis. Here, we show that HbpA can induce the transcription and secretion of proinflammatory cytokines (IL-6, TNF-α, and MCP-1) in porcine alveolar macrophages (PAM, 3D4/31). The HbpA protein is recognized by Toll-like receptors 2 and 4 on 3D4/21 macrophages, resulting in the activation of MAP kinase and NF-κB signalling cascades and the transcription and secretion of proinflammatory cytokines. HbpA contributes to virulence and bacterial pulmonary colonization in C57BL/6 mice and plays a role in adhesion to host cells and evasion of the bactericidal effect of pulmonary macrophages. In addition, mice immunized with HbpA were partially protected against challenge by G. parasuis SC1401. The results suggest that HbpA plays an important role in the pathogenesis of disease caused by G. parasuis and lay a foundation for the development of a subunit or chimeric anti-G. parasuis vaccine.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , NF-kappa B , Signal Transduction , Swine Diseases , Animals , Mice , Haemophilus parasuis/immunology , Haemophilus Infections/veterinary , Haemophilus Infections/prevention & control , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , NF-kappa B/metabolism , Swine Diseases/prevention & control , Swine Diseases/microbiology , Swine Diseases/immunology , Swine , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Pasteurellaceae/immunology , Inflammation/prevention & control , Inflammation/veterinary , Female
8.
Vet Microbiol ; 294: 110106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776767

ABSTRACT

Glaesserella parasuis (G. parasuis) is the causative agent of porcine Glässer's disease, resulting in high mortality rates in pigs due to excessive inflammation-induced tissue damage. Previous studies investigating the protective effects of G. parasuis vaccination indicated a possible role of ApoA1 in reflecting disease progression following G. parasuis infection. However, the mechanisms of ApoA1 expression and its role in these infections are not well understood. In this investigation, newborn porcine tracheal (NPTr) epithelial cells infected with G. parasuis were used to elucidate the molecular mechanism and role of ApoA1. The study revealed that the AMPK pathway activation inhibited ApoA1 expression in NPTr cells infected with G. parasuis for the first time. Furthermore, Egr1 was identified as a core transcription factor regulating ApoA1 expression using a CRISPR/Cas9-based system. Importantly, it was discovered that APOA1 protein significantly reduced apoptosis, pyroptosis, necroptosis, and inflammatory factors induced by G. parasuis in vivo. These findings not only enhance our understanding of ApoA1 in response to bacterial infections but also highlight its potential in mitigating tissue damage caused by G. parasuis infection.


Subject(s)
AMP-Activated Protein Kinases , Apolipoprotein A-I , Early Growth Response Protein 1 , Haemophilus parasuis , Signal Transduction , Swine Diseases , Animals , Swine , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Haemophilus parasuis/genetics , Swine Diseases/microbiology , Swine Diseases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Epithelial Cells/microbiology , Gene Expression Regulation , Trachea/microbiology , Trachea/metabolism , Apoptosis , Animals, Newborn
9.
Vet Microbiol ; 294: 110127, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797057

ABSTRACT

Glaesserella parasuis (G. parasuis) is a common Gram-negative commensal bacterium in the upper respiratory tract of swine that can cause Glässer's disease under stress conditions. Pyroptosis is an important immune defence mechanism of the body that plays a crucial role in clearing pathogen infections and endogenous danger signals. This study aimed to investigate the mechanism of G. parasuis serotype 5 SQ (GPS5-SQ)-induced pyroptosis in swine tracheal epithelial cells (STECs). The results of the present study demonstrated that GPS5-SQ infection induces pyroptosis in STECs by enhancing the protein level of the N-terminal domain of gasdermin D (GSDMD-N) and activating the NOD-like receptor protein 3 (NLRP3) inflammasome. Furthermore, the levels of pyroptosis-related proteins, including GSDMD-N and cleaved caspase-1 were considerably decreased in STECs after the knockdown of retinoic acid inducible gene-I (RIG-I) and mitochondrial antiviral signaling protein (MAVS). These results indicated that GPS5-SQ might trigger pyroptosis through the activation of the RIG-I/MAVS/NLRP3 signaling pathway. More importantly, the reactive oxygen species (ROS) scavenger N-acetylcysteine (NAC) repressed the activation of the RIG-I/MAVS/NLRP3 signaling and rescued the decrease in Occludin and zonula occludens-1 (ZO-1) after GPS5-SQ infection. Overall, our findings show that GPS5-SQ can activate RIG-I/MAVS/NLRP3 signaling and destroy the integrity of the epithelial barrier by inducing ROS generation in STECs, shedding new light on G. parasuis pathogenesis.


Subject(s)
Epithelial Cells , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Signal Transduction , Animals , Epithelial Cells/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Swine , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/genetics , Trachea/microbiology , Trachea/cytology , Swine Diseases/microbiology , Serogroup , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Inflammasomes/metabolism , Inflammasomes/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology
10.
Poult Sci ; 103(6): 103751, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38652951

ABSTRACT

Infectious coryza (IC) is an acute infectious respiratory disease in chickens that is caused by Avibacterium paragallinarum (A. paragallinarum). A. paragallinarum poses a significant threat to poultry health due to its virulence and multidrug resistance. This study isolated and identified 21 A. paragallinarum isolates from Guangdong between 2022 and 2023. Biochemical tests showed that 100% of A. paragallinarum isolates fermented glucose but did not ferment alginate and galactose, and only YZ18 was nicotinamide adenine dinucleotide independent. To determine the genetic relatedness between these isolates and NCBI reference strains, whole-genome-based phylogenetic analysis was employed. In addition, analysis of the 2,000 bp-length hmtp210 gene showed that the hmtp210 gene was strongly associated with A. paragallinarum serotypes. Meanwhile, a PCR assay for serotyping A. paragallinarum was developed based on the hmtp210 gene, this assay has high sensitivity and specificity. The antimicrobial susceptibility of isolates was assessed using the disk diffusion method. The antibiotic resistance genes of isolates were analyzed using the genomic method. Phenotypic resistance to ampicillin (95.2%), streptomycin (95.2%), methotrexate-sulfamethoxazole (90.5%), and tetracycline (85.7%) was most frequent among the isolates. All of the isolates exhibited resistance to multiple drugs, and furthermore, the isolates possessed a collective total of 14 genes associated with antibiotic resistance. This study will contribute to advancing our knowledge of A. paragallinarum antibiotic resistance and provide a scientific basis for the prophylaxis and treatment of IC, and the subsequent rational design of potential clinical therapeutics.


Subject(s)
Anti-Bacterial Agents , Chickens , Poultry Diseases , Poultry Diseases/microbiology , Poultry Diseases/epidemiology , Animals , China/epidemiology , Anti-Bacterial Agents/pharmacology , Prevalence , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Haemophilus Infections/epidemiology , Pasteurellaceae/genetics , Pasteurellaceae/drug effects , Drug Resistance, Bacterial/genetics , Phylogeny , Haemophilus paragallinarum/genetics , Haemophilus paragallinarum/drug effects , Haemophilus paragallinarum/physiology , Genome, Bacterial
11.
BMC Vet Res ; 20(1): 141, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582846

ABSTRACT

Glaesserella parasuis, an important respiratory bacterial pathogen, causes Glässer's disease in piglets, with potential immunosuppression. We established a piglet infection model and explored the immunosuppression mechanism to improve our understanding of the host immune response to G. parasuis. Twenty piglets were randomly divided into two groups (n = 10). The infection group was intraperitoneally challenged with 2 × 108 CFU of G. parasuis in 2 mL TSB. The control group was intraperitoneally injected with equivalent TSB. After 72 h, the piglets were sacrificed, and spleen tissue was collected. PD-1/PD-L1 expression was determined. The splenocytes were isolated to detect CD3+ T, CD3+CD4+ T, CD3+CD8+ T and CD3-CD21+cell differentiation. Via data-independent acquisition (DIA), we compared the proteomics of healthy and infected spleen tissues. Glaesserella parasuis modified CD3+ T, CD3+CD4+ T, CD3+CD8+ T and CD3-CD21+ cell differentiation and PD-1/PD-L1 expression in the spleen. The infection group had 596 proteins with significant differences in expression, of which 301 were significantly upregulated and 295 downregulated. Differentially expressed proteins (DEPs) were mainly related to immune responses. This is the first study on PD-1/PD-L1 expression in the spleen associated with immunosuppression in a piglet model to explore the protein changes related to immune responses via DIA.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Animals , B7-H1 Antigen , Haemophilus Infections/microbiology , Haemophilus Infections/veterinary , Immunosuppression Therapy/veterinary , Phosphatidylinositol 3-Kinases , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-akt , Swine , Swine Diseases/microbiology , TOR Serine-Threonine Kinases
12.
Avian Pathol ; 53(4): 291-302, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38385975

ABSTRACT

ABSTRACTThe study describes three clinical cases of infection with Avibacterium spp.. In case no. 1, respiratory clinical signs and high mortality (0.7-4.2% daily; total 21.2%) in Ross 308 broiler chickens were shown to be caused by coinfection with sequence type 9 of O. rhinotracheale presumptive serotype A and A. paragallinarum presumptive serotype B. The identical (pulsed-field gel electrophoresis) restriction pattern (pulsotype) of seven A. paragallinarum isolates indicated that infectious coryza in broilers was caused by the same clone. In cases 2 and 3, sudden increased deaths in Ross 308 broiler breeders (especially males) with lesions in the endocardium (valvular or mural endocarditis) were shown to be caused by A. endocarditis. Among nine antibiotics tested, florfenicol was the only antibiotic to which all A. paragallinarum and O. rhinotracheale isolates were susceptible. Out of the eight antibiotics tested, 11 A. endocarditis isolates from both clinical cases of infective endocarditis were susceptible to penicillin, amoxicillin, doxycycline and florfenicol. The A. endocarditis isolates tested in both clinical cases had different PFGE patterns (pulsotypes), but identical within a case. The causes of infectious coryza and infective endocarditis in the cases presented have not been determined. In the prevention of infectious diseases in large-scale livestock farming, it is very important to follow the rules of biosecurity.


Subject(s)
Anti-Bacterial Agents , Chickens , Coinfection , Flavobacteriaceae Infections , Haemophilus Infections , Ornithobacterium , Poultry Diseases , Animals , Poultry Diseases/microbiology , Poultry Diseases/pathology , Chickens/microbiology , Ornithobacterium/genetics , Ornithobacterium/isolation & purification , Female , Coinfection/veterinary , Coinfection/microbiology , Flavobacteriaceae Infections/veterinary , Flavobacteriaceae Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Male , Poland/epidemiology , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Haemophilus paragallinarum/genetics , Haemophilus paragallinarum/isolation & purification , Endocarditis, Bacterial/veterinary , Endocarditis, Bacterial/microbiology , Pasteurellaceae/isolation & purification , Pasteurellaceae/genetics , Microbial Sensitivity Tests/veterinary
13.
J Anim Sci ; 1012023 Jan 03.
Article in English | MEDLINE | ID: mdl-37882211

ABSTRACT

Infectious coryza (IC) is an important respiratory infectious disease in chickens. In this study, an Avibacterium paragallinarum Page serovar C strain, named ZJ-C, was isolated from a local layer flock that was routinely vaccinated with an inactivated trivalent vaccine, using reference strain Modesto as the serovar C immunogen. The pathogenicity, immunogenicity, and genetic characteristics of ZJ-C were studied. The minimum pathogenic dose of the isolate was 100 CFU, which was 1/1,000 of the dose of the serovar C reference strain Modesto. The vaccination-challenge trial in specific pathogen-free (SPF) chickens showed that the ZJ-C bacterin could provide 100% protection against challenge from both ZJ-C and Modesto strains, whereas Modesto provided 100% protection against challenge from itself, but only 70% protection against ZJ-C. Sequence analysis of the HMTp210 hypervariable region (region 2) showed that the homology of region 2 between ZJ-C and Modesto was 96.14%, whereas the homology between ZJ-C and the Kume serovar C-4 reference strain HP60 was 99.83%. Phylogenetic analysis of region 2 showed that ZJ-C was most closely related to cluster C-4, represented by HP60. The experimental data obtained in this study will help the selection of optimal vaccine strains and assist serotyping studies of Av. paragallinarum.


Vaccination with inactivated multivalent vaccines is a primary strategy to control Infectious coryza. Avibacterium paragallinarum serotyping is important for effective protection as inactivated whole-cell vaccines provide protection against only the serogroup or serovar from which the vaccine was derived. In this study, a novel serovar within the serogroup C Avibacterium paragallinarum isolate ZJ-C has been characterized first time in China. It was highly virulent and induced 100% cross-protection to Modesto bacterin vaccinated chickens, but not the other way around.


Subject(s)
Haemophilus Infections , Haemophilus paragallinarum , Poultry Diseases , Animals , Bacterial Vaccines , Chickens/microbiology , Haemophilus Infections/prevention & control , Haemophilus Infections/veterinary , Haemophilus paragallinarum/genetics , Phylogeny , Poultry Diseases/prevention & control , Poultry Diseases/microbiology , Vaccines, Inactivated
14.
Microbiol Spectr ; 11(6): e0150823, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37882555

ABSTRACT

IMPORTANCE: The key bacterial pathogen Glaesserella parasuis, which can cause Glässer's disease, has caused significant financial losses to the swine industry worldwide. Capsular polysaccharide (CPS) is an important virulence factor for bacteria, providing the ability to avoid recognition and killing by the host immune system. Exploring the alteration of CPS synthesis in G. parasuis in response to epinephrine stimulation can lay the groundwork for revealing the pathogenic mechanism of G. parasuis as well as providing ideas for Glässer's disease control.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Animals , Swine , Virulence Factors , Haemophilus parasuis/genetics , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Swine Diseases/microbiology
15.
Avian Dis ; 67(2): 153-159, 2023 06.
Article in English | MEDLINE | ID: mdl-37556294

ABSTRACT

Avibacterium paragallinarum is an important respiratory pathogen of domestic chickens. Avibacterium paragallinarum has been subtyped into three serogroups and nine serovars according to the Page and revised Kume schemes. The major hemagglutinin antigen of A. paragallinarum is HMTp210, which is a large protein of about 2000 amino acids (aa), including a 70-aa signal peptide at its N-terminal end. However, the regions important for the hemagglutination (HA) activity and serotypes of HMTp210 remain unclear. In this study we constructed a series of A. paragallinarum strains expressing HMTp210 in-frame deletion mutants and determined their HA titers to identify the regions important for the HA activity and serotypes of HMTp210. Two distinct types of HA activities were found in HMTp210. The type 1 HA activity resided in the region spanning the full-length HA (aa 71-2084), whereas the type 2 resided in the region spanning aa 1003-2084. The putative ligand binding of the type 1 HA activity was located at aa 176-360, which had a structure similar to YadA of Yersinia enterocolitica. The putative ligand binding site of the type 2 HA activity was located at aa 1003-1125, which had a structure similar to UspA1 from Moraxella catarrhalis. The type 1 HA activity appeared to be Page serogroup specific, whereas type 2 appeared to be Kume serovar specific. Finally, sequence analyses of the regions spanning aa 1-400 and aa 1100-1600 of HMTp210 could be useful for the molecular serotyping (the Page and revised Kume schemes) of A. paragallinarum isolates.


Regiones importantes para la actividad de hemaglutinación y serotipos de la proteína HMTp210 de Avibacterium paragallinarum. La bacteria Avibacterium paragallinarum es un patógeno respiratorio importante de los pollos domésticos. Avibacterium paragallinarum se subtipificó en tres serogrupos y nueve serovares de acuerdo con los esquemas revisados de Page y Kume. El principal antígeno de la hemaglutinina de A. paragallinarum es la proteína HMTp210, que es una proteína grande de unos 2000 aminoácidos (aa), que incluye un péptido señal de 70 aminoácidos en su extremo N-terminal. Sin embargo, las regiones importantes para la actividad de hemaglutinación (HA) y de los serotipos de la proteína HMTp210 siguen sin estar determinados. En este estudio, se construyó una serie de cepas de A. paragallinarum que expresaban mutantes de deleción en marco de lectura de HMTp210 y se determinaron sus títulos de hemaglutinación para identificar las regiones importantes para la actividad de hemaglutinación y de los serotipos de HMTp210. Se encontraron dos tipos distintos de actividades hemaglutinación en la proteína HMTp210. La actividad de hemaglutinación de tipo 1 residía en la región que abarcaba la longitud completa (aminoácidos 71­2084), mientras que la de tipo 2 residía en la región que abarcaba entre los aminoácidos 1003­2084. El sitio supuesto de unión al ligando de la actividad de hemaglutinación tipo 1 se ubicó entre los aminoácidos 176­360, que tenía una estructura similar a la proteína YadA de Yersinia enterocolitica. El supuesto sitio de unión del ligando de la actividad de hemaglutinación tipo 2 se ubicó entre los aminoácidos 1003­1125, que tenía una estructura similar a la proteína UspA1 de Moraxella catarrhalis. La actividad de hemaglutinación tipo 1 parecía ser específica del serogrupo Page, mientras que la hemaglutinación tipo 2 parecía ser específica del serovar Kume. Finalmente, los análisis de secuencias de las regiones que abarcan los aminácidos 1­400 y aminoácidos 1100­1600 de HMTp210 podrían ser útiles para la serotipificación molecular (por el esquema revisado de Page y Kume revisado) de aislamientos de A. paragallinarum.


Subject(s)
Haemophilus Infections , Haemophilus paragallinarum , Poultry Diseases , Animals , Serogroup , Hemagglutination , Haemophilus Infections/veterinary , Ligands , Chickens/microbiology , Poultry Diseases/microbiology , Haemophilus paragallinarum/genetics , Amino Acids
16.
Avian Pathol ; 52(5): 362-376, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37470411

ABSTRACT

Avibacterium paragallinarum (A. paragallinarum) is the aetiological agent of infectious coryza (IC) in chickens and characterized by acute respiratory distress and severe drop in egg production. Vaccination is important in the control of IC outbreaks and the efficacy of vaccination is dependent on A. paragallinarum serovars included in the vaccine. Classical serotyping of A. paragallinarum is laborious and hampered by poor availability of antigens and antisera. The haemagglutinin, important in classical serotyping, is encoded by the HMTp210 gene. HMTp210 gene analysis has been shown to have potential as alternative to classical serotyping. The aim of the present study was to further investigate the potential of sequence analyses of partial region 1 of the HMTp210 gene, the HMTp210 hypervariable region and the concatenated sequences of both fragments. For this analysis, 123 HMTp210 gene sequences (field isolates, A. paragallinarum serovar reference strains and vaccine strains) were included. Evaluation of serovar references and vaccine strains revealed a need for critical evaluation, especially within Page serovar B and C. Phylogenetic analysis of HMTp210 region 1 resulted in a separation of Page serovar A, B and C strains. Analysis of the HMTp210 HVR alone was not sufficient to discriminate all nine different Kume serovar references. The concatenated sequences of HMTp210 region 1 and HMTp210 HVR resulted in 14 clusters with a high correlation with Page serovar and with the nine currently known Kume serovars and is therefore proposed as a novel genotyping method that could be used as an alternative for classical serotyping of A. paragallinarum.


Subject(s)
Haemophilus Infections , Haemophilus paragallinarum , Poultry Diseases , Animals , Serotyping/veterinary , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Genotype , Phylogeny , Chickens , Haemophilus paragallinarum/genetics , Poultry Diseases/microbiology
17.
Microbiol Spectr ; 11(3): e0520922, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37212663

ABSTRACT

Avibacterium paragallinarum is the pathogen involved in infectious coryza (IC), an acute infectious upper respiratory disease in chickens. The prevalence of IC has increased in China in recent years. There is a lack of reliable and effective procedures for gene manipulation, which has limited the research on the bacterial genetics and pathogenesis of A. paragallinarum. Natural transformation has been developed as a method of gene manipulation in Pasteurellaceae by the introduction of foreign genes or DNA fragments into bacterial cells, but there has been no report on natural transformation in A. paragallinarum. In this study, we analyzed the existence of homologous genetic factors and competence proteins underlying natural transformation in A. paragallinarum and established a method for transformation in it. Through bioinformatics analysis, we identified 16 homologs of Haemophilus influenzae competence proteins in A. paragallinarum. We found that the uptake signal sequence (USS) was overrepresented in the genome of A. paragallinarum (1,537 to 1,641 copies of the core sequence ACCGCACTT). We then constructed a plasmid, pEA-KU, that carries the USS and a plasmid, pEA-K, without the USS. These plasmids can be transferred via natural transformation into naturally competent strains of A. paragallinarum. Significantly, the plasmid that carries USS showed a higher transformation efficiency. In summary, our results demonstrate that A. paragallinarum has the ability to undergo natural transformation. These findings should prove to be a valuable tool for gene manipulation in A. paragallinarum. IMPORTANCE Natural transformation is an important mechanism for bacteria to acquire exogenous DNA molecules during the process of evolution. Additionally, it can also be used as a method to introduce foreign genes into bacteria under laboratory conditions. Natural transformation does not require equipment such as an electroporation apparatus. It is easy to perform and is similar to gene transfer under natural conditions. However, there have been no reports on natural transformation in Avibacterium paragallinarum. In this study, we analyzed the presence of homologous genetic factors and competence proteins underlying natural transformation in A. paragallinarum. Our results indicate that natural competence could be induced in A. paragallinarum serovars A, B, and C. Furthermore, the method that we established to transform plasmids into naturally competent A. paragallinarum strains was stable and efficient.


Subject(s)
Haemophilus Infections , Haemophilus paragallinarum , Pasteurellaceae , Poultry Diseases , Animals , Haemophilus Infections/veterinary , Haemophilus Infections/microbiology , Poultry Diseases/microbiology , Chickens/microbiology , Pasteurellaceae/genetics , Haemophilus paragallinarum/genetics
18.
Res Vet Sci ; 158: 226-234, 2023 May.
Article in English | MEDLINE | ID: mdl-37031471

ABSTRACT

Glaesserella parasuis (G. parasuis) is a part of the normal upper respiratory microbiota of healthy swine. In many studies, the serovars 1, 4, 5, and 12 of G. parasuis are considered to be highly virulent and its serovars 3, 6, 7, 9, and 11 are considered to be non-virulent. Until now, researchers have found that non-virulent strains of G. parasuis cause an increasing number of diseases. However, little is known concerning why non-virulent strains cause disease with the virulence changes. In present study, four G. parasuis strains were evaluated for their cytotoxicity property, which aims to compare their virulence. The results showed that highly virulent strains XX0306 and CY1201, as well as, non-virulent strains HLD0115 and YK1603 caused a series of pathological changes, increased lactate dehydrogenase (LDH) release, and decreased cell activity. In addition, compared to the control group, both highly and non-virulent strains showed similar trends, demonstrating that the method of classifying the virulence of G. parasuis based on its serovar is worth further deliberation. Hence, we investigated the adhesion capacity and invasion rate of G. parasuis, the results indicated that XX0306 and HLD0115 had the strongest adhesion and invasion ability, which contradicts the classification of the virulence of G. parasuis based on its serovar. The apoptosis degree induced by highly virulent strains was more intensive than non-virulent strains, as measured by annexin V and propidium iodide (PI) double staining. Through testing the expression of apoptosis-related genes Bcl-2 and Bax, we found highly virulent strains induced apoptosis by inhibiting the expression of Bcl-2.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Swine , Animals , Virulence/genetics , Haemophilus Infections/veterinary , Swine Diseases/epidemiology , Swine Diseases/pathology , Serogroup , Haemophilus parasuis/genetics , China/epidemiology
19.
Vet Microbiol ; 282: 109748, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37120968

ABSTRACT

Glaesserella parasuis is an important pathogen that causes fibrinous polyserositis, peritonitis and meningitis in pigs, leading to considerable economic losses to the swine industry worldwide. It is well established that the serine protease HtrA is closely associated with bacterial virulence, but the role of HtrA in G. parasuis pathogenesis remains largely unknown. To characterize the function of the htrA gene in G. parasuis, a ΔhtrA mutant was constructed. We found that the ΔhtrA mutant showed significant growth inhibition under heat shock and alkaline stress conditions, indicating HtrA is involved in stress tolerance and survival of G. parasuis. In addition, deletion of htrA gene resulted in decreased adherence to PIEC and PK-15 cells and increased phagocytic resistance to 3D4/2 macrophages, suggesting that htrA is essential for adherence of G. parasuis. Scanning electron microscopy revealed morphological surface changes of the ΔhtrA mutant, and transcription analysis confirmed that a number of adhesion-associated genes are downregulated, which corroborated the aforementioned phenomenon. Furthermore, G. parasuis HtrA induced a potent antibody response in piglets with Glässer's disease. These observations confirmed that the htrA gene is related to the survival and pathogenicity of G. parasuis.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Animals , Swine , Serogroup , Virulence/genetics , Haemophilus Infections/microbiology , Haemophilus Infections/veterinary , Swine Diseases/microbiology
20.
Res Vet Sci ; 157: 35-39, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36863230

ABSTRACT

Glaesserella parasuis (Gp) is the etiological agent of Glässer's disease (GD), which causes important economic losses for the pig intensive production worldwide. This organism uses a smart protein-based receptor to acquire specifically iron from the porcine transferrin. This surface receptor consists of transferrin-binding protein A (TbpA) and transferrin-binding protein B (TbpB). TbpB has been considered the most promising antigen to formulate a based-protein vaccine with broad-spectrum of protection against GD. The purpose of our study was to determine the capsular diversity of Gp clinical isolates collected in different Spanish regions between 2018 and 2021. A total of 68 Gp isolates were recovered from porcine respiratory or systemic samples. A species-specific PCR based on tbpA gene, followed by multiplex PCR for typing Gp isolates were performed. Serovars 5, 10, 2, 4 and 1 were the most prevalent and involved almost 84% of isolates. TbpB amino acid sequences from 59 of these isolates were analyzed, and a total of ten clades could be established. All of them showed a wide diversity with respect to capsular type, anatomical isolation site and geographical origin, with minor exceptions. Regardless of the serovars, the in silico analysis of TbpB sequences revealed that a vaccine based on a TbpB recombinant protein could potentially prevent Glässer's disease outbreaks in Spain.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Animals , Swine , Transferrin-Binding Protein B/chemistry , Transferrin-Binding Protein B/genetics , Transferrin-Binding Protein B/metabolism , Phylogeny , Haemophilus parasuis/genetics , Haemophilus Infections/veterinary , Iron/metabolism , Swine Diseases/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL