Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
Add more filters

Publication year range
1.
BMC Biotechnol ; 24(1): 60, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227838

ABSTRACT

BACKGROUND: Glaesserella parasuis (G. parasuis) is the causative agent of Glässer's disease, which causes significant economic losses in the swine industry. However, research on the pathogenesis of G. parasuis has been hampered by the lack of a simple and efficient marker-free knockout system. RESULTS: In this study, a marker-free knockout system was developed for G. parasuis using a temperature-sensitive vector. By alternating the incubation of transformants at 30°C and 37°C, we optimized the screening process for this system. The system was successfully applied to knockout the KanR cassette from JS0135ΔnanH::KanR, achieving a knockout efficiency of 90% in the final round of screening. To confirm that temperature variation was a key factor, we proceeded with knocking out the nanH and apd genes in the CF7066 strain. The knockout efficiency reached up to 100%, with the shortest screening time being only four days. The knockout of the nanH gene resulted in a significant reduction in the growth vitality of the strains, while the knockout of the apd gene led to an approximate 56% improvement in the adhesion rate. Additionally, we observed that the expression of recombinant genes in transformants was higher at 30℃ than at 37℃, with the recC gene being upregulated approximately 7-fold. In contrast, there was almost no difference in the expression of recombinant genes between 30℃ and 37℃ in the wild-type strains. This discrepancy was likely due to an elevated copy number of target plasmids at 30℃, which may have resulted in the enhanced expression of recombinant genes. CONCLUSIONS: In conclusion, this newly developed gene knockout system for G. parasuis presents a valuable tool for advancing research on this organism.


Subject(s)
Gene Knockout Techniques , Haemophilus parasuis , Temperature , Haemophilus parasuis/genetics , Gene Knockout Techniques/methods , Animals , Swine , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
Microb Pathog ; 192: 106685, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750774

ABSTRACT

QseC is a membrane sensor kinase that enables bacteria to perceive autoinducers -3, adrenaline, and norepinephrine to initiate downstream gene transcription. In this study, we found that the QseC protein of Glaesserella parasuis can serve as an effective antigen to activate the host's immune response. Therefore, we investigated the immunogenicity and host protective effect of this protein. ELISA and indirect immunofluorescence results showed that QseC protein can induce high titer levels of humoral immunity in mice and regularly generate specific serum antibodies. We used MTS reagents to detect lymphocyte proliferation levels and found that QseC protein can cause splenic lymphocyte proliferation with memory and specificity. Further immunological analysis of the spleen cell supernatant revealed significant upregulation of levels of IL-1ß, IL-4 and IFN-γ in the QseC + adjuvant group. In the mouse challenge experiment, it was found that QseC + adjuvant can provide effective protection. The results of this study demonstrate that QseC protein provides effective protection in a mouse model and has the potential to serve as a candidate antigen for a novel subunit vaccine for further research.


Subject(s)
Antibodies, Bacterial , Haemophilus Infections , Interferon-gamma , Interleukin-4 , Animals , Mice , Interleukin-4/metabolism , Interleukin-4/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Haemophilus Infections/immunology , Haemophilus Infections/prevention & control , Haemophilus Infections/microbiology , Interferon-gamma/metabolism , Histidine Kinase/genetics , Histidine Kinase/metabolism , Histidine Kinase/immunology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Immunity, Humoral , Mice, Inbred BALB C , Spleen/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Cell Proliferation , Female , Adjuvants, Immunologic , Haemophilus parasuis/immunology , Haemophilus parasuis/genetics , Cytokines/metabolism , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Disease Models, Animal , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Lymphocytes/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics
3.
Vet Res ; 55(1): 130, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39375812

ABSTRACT

Capsular polysaccharide is an important virulence factor of Glaesserella parasuis. An acapsular mutant displays multiple phenotype variations, while the underlying mechanism for these variations is unknown. In this study, we created an acapsular mutant by deleting the wza gene in the capsule locus. We then used transcriptome analysis to compare the gene expression profiles of the wza deletion mutant with those of the parental strain to understand the possible reasons for the phenotypic differences. The mutant Δwza, which has a deleted wza gene, secreted less polysaccharide and lost its capsule structure. The Δwza exhibited increased autoagglutination, biofilm formation and adherence to eukaryotic cells, while the complementary strain C-Δwza partially restored the phenotype. Transcriptome analysis revealed several differentially expressed genes (DEGs) in Δwza, including up-regulated outer membrane proteins and proteins involved in peptidoglycan biosynthesis, suggesting that wza deletion affects the cell wall homeostasis of G. parasuis. Transcriptome analysis revealed the contribution of non-coding RNAs in the regulation of DEGs. Moreover, a new virulence-associated trimeric autotransporter, VtaA31 is upregulated in Δwza. It is responsible for enhanced autoagglutination but not for enhanced biofilm formation and adherence to eukaryotic cells in Δwza. In conclusion, these data indicate that wza affects the expression of multiple genes, especially those related to cell wall synthesis. Furthermore, they provide evidence that vtaA31 is involved in the autoagglutination of G. parasuis.


Subject(s)
Gene Expression Profiling , Haemophilus parasuis , Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/physiology , Virulence , Gene Expression Profiling/veterinary , Animals , Biofilms , Virulence Factors/genetics , Virulence Factors/metabolism , Transcriptome , Swine Diseases/microbiology , Type V Secretion Systems/genetics , Type V Secretion Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
4.
Int J Mol Sci ; 25(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38674155

ABSTRACT

Different levels of EspP2 expression are seen in strains of Glaesserella parasuis with high and low pathogenicity. As a potential virulence factor for G. parasuis, the pathogenic mechanism of EspP2 in infection of host cells is not clear. To begin to elucidate the effect of EspP2 on virulence, we used G. parasuis SC1401 in its wild-type form and SC1401, which was made EspP2-deficient. We demonstrated that EspP2 causes up-regulation of claudin-1 and occludin expression, thereby promoting the adhesion of G. parasuis to host cells; EspP2-deficiency resulted in significantly reduced adhesion of G. parasuis to cells. Transcriptome sequencing analysis of EspP2-treated PK15 cells revealed that the Rap1 signaling pathway is stimulated by EspP2. Blocking this pathway diminished occludin expression and adhesion. These results indicated that EspP2 regulates the adhesion of Glaesserella parasuis via Rap1 signaling pathway.


Subject(s)
Haemophilus parasuis , Signal Transduction , rap1 GTP-Binding Proteins , Animals , Haemophilus parasuis/pathogenicity , Haemophilus parasuis/genetics , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics , Bacterial Adhesion , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Occludin/metabolism , Occludin/genetics , Claudin-1/metabolism , Claudin-1/genetics , Cell Line , Swine
5.
Infect Immun ; 91(12): e0035123, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37930004

ABSTRACT

Virulent Glaesserella parasuis may engender systemic infection characterized by fibrinous polyserositis and pneumonia. G. parasuis causes systemic disease through upper respiratory tract infection, but the mechanism has not been fully characterized. Tight junction (TJ) proteins maintain the integrity and impermeability of the epithelial barriers. In this work, we applied the recombinant cytolethal distending toxin (CDT) holotoxin and cdt-deficient mutants to assess whether CDT interacted with TJ proteins of airway tract cells. Our results indicated that CDT induced the TJ occludin (OCLN) expression in newborn pig tracheal epithelial cells within the first 3 hours of bacterial infection, followed by a significant decrease. Overexpression of OCLN in target cells made them more susceptible to G. parasuis adhesion, whereas ablation of OCLN expression by CRISPR/Cas 9 gene editing technology in target cells decreased their susceptibility to bacterial adhesion. In addition, CDT treatment could upregulate the OCLN levels in the lung tissue of C57/BL6 mice. In summary, highly virulent G. parasuis strain SC1401 stimulated the tight junction expression, resulting in higher bacterial adhesion to respiratory tract cells, and this process is closely related to CDT. Our results may provide novel insights into G. parasuis infection and CDT-mediated pathogenesis.


Subject(s)
Bacterial Adhesion , Haemophilus Infections , Haemophilus parasuis , Lung , Occludin , Animals , Mice , Epithelial Cells/microbiology , Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Occludin/genetics , Occludin/metabolism , Swine , Up-Regulation , Haemophilus Infections/metabolism , Haemophilus Infections/microbiology , Lung/microbiology , Mice, Inbred C57BL
6.
BMC Vet Res ; 19(1): 135, 2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37641044

ABSTRACT

BACKGROUND: Glaesserella parasuis is the causative agent of Glässer's disease in pigs. Serotyping is the most common method used to type G. parasuis isolates. However, the high number of non-typables (NT) and low discriminatory power make serotyping problematic. In this study, 218 field clinical isolates and 15 G. parasuis reference strains were whole-genome sequenced (WGS). Multilocus sequence types (MLST), serotypes, core-genome phylogeny, antimicrobial resistance (AMR) genes, and putative virulence gene information was extracted. RESULTS: In silico WGS serotyping identified 11 of 15 serotypes. The most frequently detected serotypes were 7, 13, 4, and 2. MLST identified 72 sequence types (STs), of which 66 were novel. The most predominant ST was ST454. Core-genome phylogeny depicted 3 primary lineages (LI, LII, and LIII), with LIIIA sublineage isolates lacking all vtaA genes, based on the structure of the phylogenetic tree and the number of virulence genes. At least one group 1 vtaA virulence genes were observed in most isolates (97.2%), except for serotype 8 (ST299 and ST406), 15 (ST408 and ST552) and NT (ST448). A few group 1 vtaA genes were significantly associated with certain serotypes or STs. The putative virulence gene lsgB, was detected in 8.3% of the isolates which were predominantly of serotype 5/12. While most isolates carried the bcr, ksgA, and bacA genes, the following antimicrobial resistant genes were detected in lower frequency;  blaZ (6.9%), tetM (3.7%), spc (3.7%), tetB (2.8%), bla-ROB-1 (1.8%), ermA (1.8%), strA (1.4%), qnrB (0.5%), and aph3''Ia (0.5%).   CONCLUSION: This study showed the use of WGS to type G. parasuis isolates and can be considered an alternative to the more labor-intensive and traditional serotyping and standard MLST. Core-genome phylogeny provided the best strain discrimination. These findings will lead to a better understanding of the molecular epidemiology and virulence in G. parasuis that can be applied to the future development of diagnostic tools, autogenous vaccines, evaluation of antibiotic use, prevention, and disease control.


Subject(s)
Haemophilus parasuis , Animals , Swine , Multilocus Sequence Typing/veterinary , Phylogeny , Serogroup , Serotyping/veterinary , Haemophilus parasuis/genetics , North America
7.
Microb Pathog ; 169: 105617, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35680006

ABSTRACT

Glaesserella parasuis (G. parasuis) is the pathogen of Glässer's disease in pig herds, which can cause severe inflammatory responses. However, at present, the pathogenic mechanism of G. parasuis is not very clear. LncRNAs can regulate the expression of mRNA in a variety of ways, thereby causing host cells to produce a variety of functional changes in response to bacterial infection. Here, we detected the changes in lncRNAs and mRNAs of 3D4/21 cells after G. parasuis CY1201 strain (serotype 13) infection. A total of 876 lncRNAs and 2166 mRNAs were differentially expression in 3D4/21 cells after G. parasuis infection. GO and KEGG enrichment analysis showed that the differentially up-regulated lncRNA target genes were mainly involved in the response to extracellular stimuli, cell receptor signaling pathways and chemokine signaling pathways. The differentially down-regulated lncRNA target genes were mainly involved in ERK1/ERK2 cascade reaction and adhesion junctions. 44 lncRNAs were screened that might be related in inflammation. CeRNA regulatory network of the top five difference inflammation-related lncRNAs showed that the up-regulated lncRNA group involved 5 lncRNAs, 50 miRNAs and 49 mRNAs. Meanwhile, there were 26 miRNAs and 36 mRNAs in the top five down-regulated lncRNA group. Our results contribute to understand the basic role of lncRNAs in 3D4/21 cells during G. parasuis infection, and lay the foundation for following research.


Subject(s)
Haemophilus parasuis , MicroRNAs , RNA, Long Noncoding , Animals , Gene Regulatory Networks , Haemophilus parasuis/genetics , Inflammation/metabolism , Lung , Macrophages, Alveolar/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Transcriptome
8.
Appl Microbiol Biotechnol ; 106(13-16): 5167-5178, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35851417

ABSTRACT

Glaesserella parasuis is an important bacterial pathogen that affects the swine industry worldwide. Research on the pathogenic mechanism and genetically engineered vaccine remains undeveloped because an effective markerless and multiple-gene knockout system is unavailable for G. parasuis yet. To establish a markerless knockout, deleted allelic genes with kanamycin resistance (KanR) cassettes were introduced into the genome of G. parasuis by using natural transformation with suicide plasmids. Then, the KanR cassette was excised with a thermosensitive plasmid pGF conferring a constitutive Flp expression. To realize the markerless and multiple-gene knockout, plasmid pGAF was constructed by placing the Flp gene under the control of an arabinose-inducible promoter. Firstly, pGAF was introduced into G. parasuis by electroporation, and the marked mutants were produced following natural transformation. Finally, the KanR cassette was excised from the genome by the inducible expression of Flp upon arabinose action. Based on the natural transformation and the inducible expression of Flp, the markerless single-gene knockout mutants of ΔhsdR, ΔneuA2, ΔespP2, Δapd, and ΔnanH were constructed. In addition, a five-gene knockout mutant of ΔhsdRΔneuA2ΔespP2ΔapdΔnanH was generated by successive natural transformation with five suicide plasmids. Taken together, a markerless and multiple-gene deletion system was established for G. parasuis in the present study for the first time. This system is simple, efficient, and easy to manipulate for G. parasuis; thus, our technique will substantially aid the understanding of the etiology, pathogenesis, and genetic engineering of G. parasuis and other bacteria that can be naturally transformed in laboratory conditions. KEY POINTS: • Flp recombinase excised the KanR gene flanked by FRT sites in Glaesserella parasuis. • The regulatory expression of Flp enabled a multiple-gene knockout forG. parasuis. • The technique will promote the understanding of Glässer's disease pathogens.


Subject(s)
Arabinose , Haemophilus parasuis , Animals , DNA Nucleotidyltransferases/genetics , Gene Knockout Techniques , Haemophilus parasuis/genetics , Haemophilus parasuis/metabolism , Humans , Swine
9.
Funct Integr Genomics ; 21(5-6): 695-707, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34676472

ABSTRACT

Haemophilus (Glaesserella) parasuis is a commensal bacterium that causes Glässer's disease (GD) in swine. As a global transcriptional factor, CheY regulates the expression of hundreds of genes in H. parasuis. In this study, we measured changes in gene expression at the whole transcriptome level using RNAseq. We identified 2058 co-expressed genes, and found 624 differentially expressed genes (q < 0.05) in ΔcheY and SC1401. Several important GO annotations and signaling pathways were identified. RNA-seq results were assembled according to the reference genome, compared with the annotated gene model, and 12 new transcriptional regions were found. Finally, q-PCR results validated the RNA-seq results with 8 randomly selected genes. The present study indicated that CheY is mainly involved in the regulation of ABC transport, oxidative phosphorylation, and ß-Lactam resistance. We draw the regulatory network of CheY, which offers greater insight into the regulatory mechanism of CheY in H.parasuis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/genetics , Gene Deletion , Haemophilus parasuis/genetics , Haemophilus parasuis/metabolism , Transcriptome , Animals , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Swine/microbiology
10.
Vet Res ; 52(1): 135, 2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34674760

ABSTRACT

Glaesserella parasuis (G. parasuis) is a commensal bacterium in the upper respiratory tract of pigs that can also cause the swine Glässer disease, which induces an intensive inflammatory response and results in significant economic losses to the swine industry worldwide. G. parasuis can cause disease through infection of the respiratory tract, resulting in systemic infection, but the mechanism is largely unknown. Recently we showed that Glaesserella parasuis serotype 4 (GPS4) increased swine tracheal epithelial barrier permeability, resulting in easier bacterial translocation. Tight junction proteins (TJ) play a crucial role in maintaining the integrity and impermeability of the epithelial barrier. GPS4 decreased the expression of the TJ ZO-1 and occludin in swine tracheal epithelial cells (STEC). Furthermore, the proinflammatory cytokines IL-6, IL-8 and TNF-α were significantly upregulated in GPS4-infected STEC, and both the MAPK and NF-κB signaling pathways were activated and contributed to the expression of TNF-α. We demonstrate that the production of proinflammatory cytokines, especially TNF-α, during GPS4 infection was involved in barrier dysfunction. Additionally, animal challenge experiments confirmed that GPS4 infection downregulated TJ in the lungs of piglets and induced a severe inflammatory response. In general, G. parasuis infection downregulated the expression of TJ and induced massive secretion of proinflammatory cytokines, resulting in epithelial barrier disruption and favoring bacterial infection. This study allowed us to better understand the mechanism by which G. parasuis crosses the respiratory tract of pigs.


Subject(s)
Bacterial Translocation , Haemophilus parasuis/physiology , Pasteurellaceae Infections/veterinary , Signal Transduction , Swine Diseases/microbiology , Animals , Epithelial Cells , Haemophilus Infections/microbiology , Haemophilus Infections/physiopathology , Haemophilus Infections/veterinary , Haemophilus parasuis/genetics , Pasteurellaceae Infections/microbiology , Pasteurellaceae Infections/physiopathology , Serogroup , Sus scrofa , Swine , Swine Diseases/physiopathology
11.
Vet Res ; 52(1): 68, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33980312

ABSTRACT

Glaesserella parasuis strains were characterized by serotyping PCR, vtaA virulence marker Leader Sequence (LS)-PCR, clinical significance, and geographic region. Overall, the serovars 4, 5/12, 7, 1, and 13 were the most commonly detected. Serovars of greatest clinical relevance were systemic isolates that had a higher probability of being serovar 5/12, 13, or 7. In comparison, pulmonary isolates had a higher likelihood of being serovars 2, 4, 7, or 14. Serovars 5/12 and 13 have previously been considered disease-associated, but this study agrees with other recent studies showing that serovar 7 is indeed associated with systemic G. parasuis disease. Serovar 4 strains illustrated how isolates can have varying degrees of virulence and be obtained from pulmonary, systemic, or nasal sites. Serovars 8, 9, 15, and 10 were predominantly obtained from nasal samples, which indicates a limited clinical significance of these serovars. Additionally, most internal G. parasuis isolates were classified as virulent by LS-PCR and were disease-associated isolates, including serovars 1, 2, 4, 5/12, 7, 13, and 14. Isolates from the nasal cavity, including serovars 6, 9, 10, 11, and 15, were classified as non-virulent by LS-PCR. In conclusion, the distribution of G. parasuis serovars remains constant, with few serovars representing most of the strains isolated from affected pigs. Moreover, it was confirmed that the LS-PCR can be used for G. parasuis virulence prediction of field strains worldwide.


Subject(s)
Haemophilus Infections/veterinary , Haemophilus parasuis/genetics , Swine Diseases/epidemiology , Animals , Asia/epidemiology , Europe/epidemiology , Haemophilus Infections/epidemiology , Haemophilus Infections/microbiology , North America/epidemiology , Polymerase Chain Reaction/veterinary , Prevalence , Seroepidemiologic Studies , Serotyping/veterinary , Sus scrofa , Swine , Swine Diseases/microbiology
12.
Curr Microbiol ; 78(4): 1566-1576, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33674900

ABSTRACT

Haemophilus parasuis is commonly found in the upper respiratory tract of the pigs. Some isolates of H. parasuis can lead to both pneumonia and Glässer's disease of pigs with severe clinical symptoms. The virulence-associated genes for the various degrees of virulence observed in H. parasuis remains poorly understood. In the present study, we identified the differentially expressed genes between YK1603 (non-virulent strain) and XM1602 (moderately virulent strain) or CY1201 (highly virulent strain) of H. parasuis using Illumina sequencing technique. In comparison to YK1603, a total of 195 genes were significantly changed in CY1201, of which 71 genes were up-regulated and 124 genes were down-regulated, whereas 705 genes were significantly changed in XM1602, of which 415 genes were up-regulated and 290 genes were down-regulated. The enriched analysis of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways on the differentially expressed genes showed that both enriched main GO terms and KEGG pathways appear to be different between the two kinds of comparision: CY1201 versus YK1603, and XM1602 versus YK1603. Based on real-time PCR technique, on the whole, it was confirmed that the expression of ten genes: lpxL, tbpB, kdtA, waaQ, oapA, napA, ptsH, mmsA, lpxM, and lpxB were agreement with the findings in Illumina sequencing analysis. These identified genes might participate in the regulation of a wide range of biological process involved in virulence of H. parasuis, such as phosphotransferase system and ABC transporters. Our results from this study provide a new way to gain insight into the virulent mechanisms of H. parasuis.


Subject(s)
Haemophilus Infections , Haemophilus parasuis , Swine Diseases , Animals , Haemophilus Infections/veterinary , Haemophilus parasuis/genetics , Swine , Transcriptome , Virulence/genetics
13.
Curr Microbiol ; 78(8): 3152-3164, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34191053

ABSTRACT

Haemophilus parasuis (H. parasuis, HPS) can elicit serious inflammatory responses and cause enormous economic loss to swine industry worldwide. However, the factors responsible for systemic infection and inflammatory responses of HPS have not yet been fully clarified. In this study, we found that lncRNA-MEG3 was significantly up-regulated in porcine alveolar macrophages (PAMs) infected with HPS. The gain- and loss-of-function analysis confirmed that lncRNA-MEG3 participated in the inflammatory responses and apoptosis in HPS-infected PAMs, which was assessed via several inflammatory cytokine genes (TNF-α, IL-1ß, and IL-6) and apoptotic factors (Bcl-2, Bax, and C-caspase-3). Based on biotin-labeled RNA pull-down assay, we found that lncRNA-MEG3 bound with miR-210 in HPS-infected PAMs. Based on both overexpression and knockdown analysis of lncRNA-MEG3, our results indicated that lncRNA-MEG3 promoted the expression of TLR4 in HPS-infected PAMs. Using dual-luciferase reporter assays, we showed that lncRNA-MEG3 positively regulated the expression of TLR4 gene in HPS-infected PAMs through miR-210 pathway. Taken together, our results indicated that lncRNA-MEG3 participated in the inflammatory responses and apoptosis in HPS-infected PAMs through modulating the miR-210/TLR4 axis. The results from this investigation provided significant information for a novel target to control HPS infection in swine.


Subject(s)
Haemophilus parasuis , MicroRNAs , RNA, Long Noncoding , Animals , Apoptosis , Haemophilus parasuis/genetics , Macrophages, Alveolar , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Swine , Toll-Like Receptor 4/genetics
14.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299288

ABSTRACT

(1) Background: Short-read sequencing allows for the rapid and accurate analysis of the whole bacterial genome but does not usually enable complete genome assembly. Long-read sequencing greatly assists with the resolution of complex bacterial genomes, particularly when combined with short-read Illumina data. However, it is not clear how different assembly strategies affect genomic accuracy, completeness, and protein prediction. (2) Methods: we compare different assembly strategies for Haemophilus parasuis, which causes Glässer's disease, characterized by fibrinous polyserositis and arthritis, in swine by using Illumina sequencing and long reads from the sequencing platforms of either Oxford Nanopore Technologies (ONT) or SMRT Pacific Biosciences (PacBio). (3) Results: Assembly with either PacBio or ONT reads, followed by polishing with Illumina reads, facilitated high-quality genome reconstruction and was superior to the long-read-only assembly and hybrid-assembly strategies when evaluated in terms of accuracy and completeness. An equally excellent method was correction with Homopolish after the ONT-only assembly, which had the advantage of avoiding hybrid sequencing with Illumina. Furthermore, by aligning transcripts to assembled genomes and their predicted CDSs, the sequencing errors of the ONT assembly were mainly indels that were generated when homopolymer regions were sequenced, thus critically affecting protein prediction. Polishing can fill indels and correct mistakes. (4) Conclusions: The assembly of bacterial genomes can be directly achieved by using long-read sequencing techniques. To maximize assembly accuracy, it is essential to polish the assembly with homologous sequences of related genomes or sequencing data from short-read technology.


Subject(s)
Haemophilus parasuis/genetics , High-Throughput Nucleotide Sequencing/methods , Nanopore Sequencing/methods , Sequence Analysis, DNA/methods , Animals , Genome, Bacterial , Haemophilus parasuis/isolation & purification , Phylogeny , Sequence Alignment , Swine
15.
Infect Immun ; 88(5)2020 04 20.
Article in English | MEDLINE | ID: mdl-32094250

ABSTRACT

Glaesserella (Haemophilus) parasuis is a commensal bacterium of the upper respiratory tract in pigs and also the causative agent of Glässer's disease, which causes significant morbidity and mortality in pigs worldwide. Isolates are characterized into 15 serovars by their capsular polysaccharide, which has shown a correlation with isolate pathogenicity. To investigate the role the capsule plays in G. parasuis virulence and host interaction, a capsule mutant of the serovar 5 strain HS069 was generated (HS069Δcap) through allelic exchange following natural transformation. HS069Δcap was unable to cause signs of systemic disease during a pig challenge study and had increased sensitivity to complement killing and phagocytosis by alveolar macrophages. Compared with the parent strain, HS069Δcap produced more robust biofilm and adhered equivalently to 3D4/31 cells; however, it was unable to persistently colonize the nasal cavity of inoculated pigs, with all pigs clearing HS069Δcap by 5 days postchallenge. Our results indicate the importance of the capsular polysaccharide to G. parasuis virulence as well as nasal colonization in pigs.


Subject(s)
Haemophilus parasuis/genetics , Animals , Biofilms , Haemophilus Infections/microbiology , Macrophages, Alveolar/microbiology , Phagocytosis/physiology , Swine , Swine Diseases/microbiology , Virulence/genetics
16.
Arch Microbiol ; 202(9): 2499-2508, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32638056

ABSTRACT

Haemophilus parasuis (H. parasuis), the cause of the Glasser's disease, is a potentially pathogenic gram-negative organism that colonizes the upper respiratory tract of pigs. The extraction of Blumea balsamifera DC., as a traditional Chinese herb, has shown great bacteriostatic effect against several common bacteria. To study the antibacterial effect on H. parasuis in vitro, this study evaluated the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of Blumea balsamifera DC. essential oil (BBO) as well as morphological changes in H. parasuis treated with it. Furthermore, changes in expression of total protein and key virulence factors were also assessed. Results showed that the MIC and MBC were 0.625 and 1.25 µg/mL, respectively. As the concentration of BBO increased, the growth curve inhibition became stronger. H. parasuis cells were damaged severely after treatment with BBO for 4 h, demonstrating plasmolysis and enlarged vacuoles, along with broken cell walls and membranes. Total protein and virulence factor expression in H. parasuis was significantly downregulated by BBO. Taken together, these results indicated a substantial antibacterial effect of BBO on H. parasuis.


Subject(s)
Asteraceae/chemistry , Haemophilus parasuis/drug effects , Oils, Volatile/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Haemophilus Infections/drug therapy , Haemophilus parasuis/genetics , Microbial Sensitivity Tests , Swine , Swine Diseases/drug therapy , Swine Diseases/microbiology , Virulence Factors/genetics
17.
Vet Res ; 51(1): 137, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33203465

ABSTRACT

Glaesserella parasuis is an important pathogen in swine production. It acts as a primary pathogen in systemic Glässer´s disease and as a secondary pathogen in Porcine Respiratory Disease Complex. In this study, a collection of 308 isolates from carrier animals and individuals with respiratory or Glässer´s disease isolated 2012-2019 in Germany was analysed. Isolates were characterized for serovar implementing two different PCR methods. Additionally, two different PCR methods for pathotyping isolates were applied to the collection and results compared. Serovar 6 (p < 0.0001) and 9 (p = 0.0007) were correlated with carrier isolates and serovar 4 was associated with isolates from animals with respiratory disease (p = 0.015). In systemic isolates, serovar 13 was most frequently detected (18.9%). Various other serovars were isolated from all sites and the ratio of serovar 5 to serovar 12 was approximately 1:2. These two serovars together represented 14.3% of the isolates; only serovar 4 was isolated more frequently (24.7%). The pathotyping method based on the leader sequence (LS = ESPR of vta) was easy to perform and corresponded well to the clinical background information. Of the carrier isolates 72% were identified as non-virulent while 91% of the systemic isolates were classified as virulent (p < 0.0001). Results of the pathotyping PCR based on 10 different marker genes overall were in good agreement with clinical metadata as well as with results of the LS-PCR. However, the pathotyping PCR was more complicated to perform and analyze. In conclusion, a combination of the serotyping multiplex-PCR and the LS-PCR could improve identification of clinically relevant G. parasuis isolates, especially from respiratory samples.


Subject(s)
Haemophilus Infections/veterinary , Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Polymerase Chain Reaction/veterinary , Swine Diseases/microbiology , Virulence/genetics , Animals , Germany , Haemophilus Infections/microbiology , Polymerase Chain Reaction/methods , Serogroup , Serotyping/veterinary , Sus scrofa , Swine
18.
Vet Res ; 51(1): 97, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32736655

ABSTRACT

Lipooligosaccharides (LOSs) are virulence determinants of Glaesserella parasuis, a pathogen of the respiratory tract of pigs. We previously reported that disruption of the galU or galE gene in G. parasuis results in increased sensitivity to porcine serum, indicating that the galactose catabolism pathway is required for polysaccharide formation in G. parasuis. Here, we evaluated the role of the HAPS_0849 gene in LOS synthesis. The G. parasuis SC096 HAPS_0849 mutant produced a highly truncated LOS molecule, although a small fraction of intact LOS was still observed, and this mutant was found to be more sensitive to serum than the parental strain. HAPS_0849 was overexpressed and purified for biochemical assays, and this protein exhibited phosphoglucomutase (PGM) activity. Heterologous expression of a pgm gene from Escherichia coli in the HAPS_0849 mutant led to restoration of the wild-type LOS glycoform, further demonstrating the PGM function of HAPS_0849 in G. parasuis. The autoagglutination and biofilm formation ability of this strain were also investigated. Disruption of HAPS_0849 led to an increased tendency to autoagglutinate and form more biofilms, and these enhanced phenotypes were observed in the absence of glucose. In addition, LOSs from HAPS_0849, galU and lgtB mutants had similar truncated glycoforms, while LOSs from the galE and lex-1 mutants exhibited another type of defective LOS pattern. These findings imply that HAPS_0849 may function upstream of GalU in the generation of glucose 1-phosphate. In conclusion, our results preliminarily described the functions of HAPS_0849 in G. parasuis, and this gene was partially required for LOS synthesis.


Subject(s)
Bacterial Proteins/genetics , Haemophilus parasuis/genetics , Lipopolysaccharides/biosynthesis , Phosphoglucomutase/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Haemophilus parasuis/enzymology , Microorganisms, Genetically-Modified/genetics , Phosphoglucomutase/metabolism
19.
BMC Vet Res ; 16(1): 167, 2020 May 27.
Article in English | MEDLINE | ID: mdl-32460764

ABSTRACT

BACKGROUND: Glaesserella parasuis, the causative agent of Glӓsser's disease, is widespread in swine globally resulting in significant economic losses to the swine industry. Prevention of Glӓsser's disease in pigs has been plagued with an inability to design broadly protective vaccines, as many bacterin based platforms generate serovar or strain specific immunity. Subunit vaccines are of interest to provide protective immunity to multiple strains of G. parasuis. Selected proteins for subunit vaccination should be widespread, highly conserved, and surface exposed. RESULTS: Two candidate proteins for subunit vaccination (RlpB and VacJ) against G. parasuis were identified using random mutagenesis and an in vitro organ culture system. Pigs were vaccinated with recombinant RlpB and VacJ, outer membrane proteins with important contributions to cellular function and viability. Though high antibody titers to the recombinant proteins and increased interferon-γ producing cells were found in subunit vaccinated animals, the pigs were not protected from developing systemic disease. CONCLUSIONS: It appears there may be insufficient RlpB and VacJ exposed on the bacterial surface for antibody to bind, preventing high RlpB and VacJ specific antibody titers from protecting animals from G. parasuis. Additionally, this work confirms the importance of utilizing the natural host species when assessing the efficacy of vaccine candidates.


Subject(s)
Haemophilus Infections/veterinary , Haemophilus parasuis/immunology , Recombinant Proteins/immunology , Swine Diseases/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Haemophilus Infections/immunology , Haemophilus Infections/prevention & control , Haemophilus Vaccines/immunology , Haemophilus parasuis/genetics , Serogroup , Sus scrofa , Swine , Swine Diseases/immunology , Swine Diseases/microbiology , Tissue Culture Techniques/veterinary , Vaccination/veterinary , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
20.
Microb Pathog ; 134: 103565, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31158493

ABSTRACT

Haemophilus parasuis (H. parasuis) is rather difficult to manipulate genetically due to the diversity of restriction-modification systems and other mechanisms harbored by various isolates. This prevents exogenous plasmids from replicating in this species and hinders research efforts focused on transcriptional regulators in this bacterium. In this study, we generated a convenient promoter reporter system based on gene knock-in method using natural transformation in H. parasuis. Gene knock-in has proven useful as a powerful tool facilitating identification and studying the transcription activities of regulators under a variety of conditions that favor gene transcription or expression from an incorporated promoter. The vectors, pDK-K and pDK-G, carrying promoterless reporter lacZ gene and two homologous sequences flanking a knock-in site, may have some advantages over the extensively used plasmid-bearing reporter system in other bacteria in stability and ease of genetic manipulation in H. parasuis. The knock-in site was positioned at a site occupied by flanking genes that were both hypothetical and had the same transcription orientation, thus the expression of the reversely cloned promoter-lacZ fusion wouldn't be affected by the upstream promoter on the chromosome. The expression activity of lacZ gene under the transcriptional activation of a 300 bp promoter-proximal segment of cyaA, crp or comA genes in H. parasuis was separately validated using X-gal and o-nitrophenyl-ß-d-galactoside(ONPG) as substrates. The derivatives harboring promoter-lacZ fusion segments showed significantly higher ß-galactosidase activity levels than the promoterlessones both in TSB++ broth and on TSA++ plate as screened either by X-gal method or the standard Miller method. We also used pDK vector to further certify that the cyaA promoter is inducible and whose transcriptional levels were in correlation with the growth kinetics of the bacteria in TSB++. With this system, gene knock-in method based on natural transformation in H. parasuis proved to be useful in identifying transcriptional regulation of a certain promoter.


Subject(s)
Gene Expression , Genes, Reporter/genetics , Haemophilus parasuis/genetics , Plasmids/genetics , Promoter Regions, Genetic/genetics , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Genetic Vectors , Genome, Bacterial , Genomic Instability , Kinetics , Lac Operon , Regulatory Elements, Transcriptional/genetics , Transformation, Bacterial , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL