Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 400
Filtrar
1.
BMC Vet Res ; 20(1): 141, 2024 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-38582846

RESUMO

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.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Suínos , Proteínas Proto-Oncogênicas c-akt , Receptor de Morte Celular Programada 1 , Fosfatidilinositol 3-Quinases , Antígeno B7-H1 , Serina-Treonina Quinases TOR , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/veterinária , Terapia de Imunossupressão/veterinária , Doenças dos Suínos/microbiologia
2.
Anal Chim Acta ; 1287: 342101, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38182383

RESUMO

BACKGROUND: Haemophilus parasuis (H. parasuis) is a gram-negative bacterial pathogen that causes severe infections in swine, resulting in substantial economic losses. Currently, the majority of H. parasuis detection methods are impractical for on-site application due to their reliance on large instruments or complex procedures. Thus, there is an urgent need to develop a rapid, visually detectable, and highly sensitive detection method, especially under resource-limited environments and field conditions. RESULTS: In this study, we established a naked eye assay for highly sensitive detection by combining recombinase polymerase amplification (RPA) with CRISPR/Cas12a technology. Positive samples exhibited a clear red color visible to the naked eye, while negative samples appeared blue. We achieved a remarkable sensitivity, detecting H. parasuis down to a single copy, with no cross-reactivity with other bacteria. In a mouse model, our assay detected H. parasuis infection nearly 8 h earlier than traditional PCR. Compared to qPCR, our detection results were 100 % accurate. To enhance point-of-care applicability and mitigate the risk of aerosol contamination from uncapping, we consolidated RPA and CRISPR/Cas12a cleavage into a single-tube reaction system. This integrated approach was validated with 20 clinical lung samples, yielding results consistent with those obtained from qPCR. The entire procedure, from DNA extraction to detection, was completed in 35 min. SIGNIFICANCE: We present an RPA-CRISPR/Cas12a assay suitable for the early and resource-efficient diagnosis of H. parasuis infections. Its simplicity and visual detection are advantageous for field diagnostics, representing a substantial develpoment in the diagnosis of H. parasuis.


Assuntos
Haemophilus parasuis , Recombinases , Camundongos , Animais , Suínos , Haemophilus parasuis/genética , Sistemas CRISPR-Cas , Bioensaio , Reações Cruzadas
3.
J Med Microbiol ; 72(12)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38112519

RESUMO

Introduction. Various plasmid-mediated resistance genes have been reported in Glaesserella parasuis, but little is known about their global distribution features, evolution pattern and spread.Gap Statement. The potential mobilization mechanisms of resistance plasmids in G. parasuis have been poorly explored.Aim. The aim of the study was to investigate the prevalence and diversity of plasmid-mediated resistance genes among G. parasuis isolates, and focus on the analysis of the features of the resistance plasmids from G. parasuis.Method. The plasmids tested were sequenced using the Illumina HiSeq platform in conjunction with PCR and inverted PCR. The susceptibility of the host strains was determined by broth microdilution. The transfer of plasmids tested was conducted by electroporation. The sequence data were compared using bioinformatics tools and the data from our laboratory and the National Center for Biotechnology Information (NCBI) database.Results. Nineteen plasmids were identified from our laboratory and these resistance plasmids were functional and transferable. Moreover, we clustered five types of genetic backbones of plasmids from G. parasuis and revealed the global distribution features of the plasmid-mediated resistance genes.Conclusions. This is the first report of the coexistence of tet(H)-bearing type I plasmid and lnu(C)-bearing type II plasmid in one G. parasuis clinical isolate. In addition, this study provides the first view of the global distribution of plasmid-mediated resistance genes and classifies the plasmids in G. parasuis according to their backbone regions.


Assuntos
Haemophilus parasuis , Plasmídeos/genética , Haemophilus parasuis/genética , Sequência de Bases
4.
Infect Immun ; 91(12): e0035123, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37930004

RESUMO

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.


Assuntos
Aderência Bacteriana , Infecções por Haemophilus , Haemophilus parasuis , Pulmão , Ocludina , Animais , Camundongos , Células Epiteliais/microbiologia , Haemophilus parasuis/genética , Haemophilus parasuis/patogenicidade , Ocludina/genética , Ocludina/metabolismo , Suínos , Regulação para Cima , Infecções por Haemophilus/metabolismo , Infecções por Haemophilus/microbiologia , Pulmão/microbiologia , Camundongos Endogâmicos C57BL
5.
Microbiol Spectr ; 11(6): e0150823, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37882555

RESUMO

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.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Suínos , Fatores de Virulência , Haemophilus parasuis/genética , Infecções por Haemophilus/veterinária , Infecções por Haemophilus/microbiologia , Doenças dos Suínos/microbiologia
6.
PeerJ ; 11: e15823, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37790626

RESUMO

Background: Glaesserella parasuis (G. parasuis) belongs to the normal microbiota of the upper respiratory tract in the swine, but virulent strains can cause systemic infections commonly known as Glässer's disease that leads to significant economic loss in the swine industry. Fifteen serotypes of G. parasuis have been classified by gel immunodiffusion test while the molecular serotyping based on variation within the capsule loci have further improved the serotype determination of unidentified field strains. Serovar has been commonly used as an indicator of virulence; however, virulence can be significantly differ in the field isolates with the same serotype. To date, investigations of G. parasuis isolated in Taiwan regarding antimicrobial resistance, serotypes, genotypes and virulence factors remain unclear. Methods: A total of 276 G.parasuis field isolates were collected from 263 diseased pigs at the Animal Disease Diagnostic Center of National Chiayi University in Taiwan from January 2013 to July 2021. Putative virulence factors and serotypes of the isolates were identified by polymerase chain reaction (PCR) and antimicrobial susceptibility testing was performed by microbroth dilution assay. Additionally, the epidemiology of G. parasuis was characterized by multilocus sequence typing (MLST). Results: Serotype 4 (33.3%) and 5 (21.4%) were the most prevalent, followed by nontypable isolates (15.9%), serotype 13 (9.4%), 12 (6.5%), 14 (6.2%), 7 (3.3%), 1 (1.8%), 9 (1.1%), 11 (0.7%) and 6 (0.4%). Nine out of 10 putative virulence factors showed high positive rates, including group 1 vtaA (100%), fhuA (80.4%), hhdA (98.6%), hhdB (96.0%), sclB7 (99.6%), sclB11 (94.9%), nhaC (98.2%), HAPS_0254 (85.9%), and cirA (99.3%). According to the results of antimicrobial susceptibility testing, ceftiofur and florfenicol were highly susceptible (>90%). Notably, 68.8% isolates showed multidrug resistance. MLST revealed 16 new alleles and 67 new sequence types (STs). STs of these isolated G. parasuis strains were classified into three clonal complexes and 45 singletons by Based Upon Related Sequence Types (BURST) analysis. All the G. parasuis strains in PubMLST database, including strains from the diseased pigs in the study, were defined into two main clusters by Unweighted Pair Group Method with Arithmetic Mean (UPGMA). Most isolates in this study and virulent isolates from the database were mainly located in cluster 2, while cluster 1 included a high percentage of nasal isolates from asymptomatic carriers. In conclusion, this study provides current prevalence and antimicrobial susceptibility of G. parasuis in Taiwan, which can be used in clinical diagnosis and treatment of Glässer's disease.


Assuntos
Anti-Infecciosos , Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Humanos , Suínos , Animais , Fatores de Virulência/genética , Sorogrupo , Tipagem de Sequências Multilocus , Taiwan/epidemiologia , Doenças dos Suínos/epidemiologia , Haemophilus parasuis/genética , Infecções por Haemophilus/epidemiologia
7.
BMC Vet Res ; 19(1): 135, 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37641044

RESUMO

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.


Assuntos
Haemophilus parasuis , Animais , Suínos , Tipagem de Sequências Multilocus/veterinária , Filogenia , Sorogrupo , Sorotipagem/veterinária , Haemophilus parasuis/genética , América do Norte
8.
Front Cell Infect Microbiol ; 13: 1192651, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37207184

RESUMO

Haemophilus parasuis is a commensal organism of the upper respiratory tract of pigs, but virulent strains can cause Glässer's disease, resulting in significant economic losses to the swine industry. OmpP2 is an outer membrane protein of this organism that shows considerable heterogeneity between virulent and non-virulent strains, with classification into genotypes I and II. It also acts as a dominant antigen and is involved in the inflammatory response. In this study, 32 monoclonal antibodies (mAbs) against recombinant OmpP2 (rOmpP2) of different genotypes were tested for reactivity to a panel of OmpP2 peptides. Nine linear B cell epitopes were screened, including five common genotype epitopes (Pt1a, Pt7/Pt7a, Pt9a, Pt17, and Pt19/Pt19a) and two groups of genotype-specific epitopes (Pt5 and Pt5-II, Pt11/Pt11a, and Pt11a-II). In addition, we used positive sera from mice and pigs to screen for five linear B-cell epitopes (Pt4, Pt14, Pt15, Pt21, and Pt22). After porcine alveolar macrophages (PAMs) were stimulated with overlapping OmpP2 peptides, we found that the epitope peptides Pt1 and Pt9, and the loop peptide Pt20 which was adjacent epitopes could all significantly upregulated the mRNA expression levels of IL-1α, IL-1ß, IL-6, IL-8, and TNF-α. Additionally, we identified epitope peptides Pt7, Pt11/Pt11a, Pt17, Pt19, and Pt21 and loop peptides Pt13 and Pt18 which adjacent epitopes could also upregulate the mRNA expression levels of most proinflammatory cytokines. This suggested that these peptides may be the virulence-related sites of the OmpP2 protein, with proinflammatory activity. Further study revealed differences in the mRNA expression levels of proinflammatory cytokines, including IL-1ß and IL-6, between genotype-specific epitopes, which may be responsible for pathogenic differences between different genotype strains. Here, we profiled a linear B-cell epitope map of the OmpP2 protein and preliminarily analyzed the proinflammatory activities and effects of these epitopes on bacterial virulence, providing a reliable theoretical basis for establishing a method to distinguish strain pathogenicity and to screen candidate peptides for subunit vaccines.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Suínos , Animais , Camundongos , Epitopos de Linfócito B/genética , Interleucina-6/metabolismo , Citocinas/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Infecções por Haemophilus/microbiologia , Doenças dos Suínos/microbiologia , RNA Mensageiro/metabolismo
9.
Res Vet Sci ; 158: 226-234, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37031471

RESUMO

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.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Suínos , Animais , Virulência/genética , Infecções por Haemophilus/veterinária , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/patologia , Sorogrupo , Haemophilus parasuis/genética , China/epidemiologia
10.
Vet Microbiol ; 282: 109748, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37120968

RESUMO

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.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Suínos , Sorogrupo , Virulência/genética , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/veterinária , Doenças dos Suínos/microbiologia
11.
Appl Environ Microbiol ; 89(4): e0200222, 2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-36920203

RESUMO

Glaesserella parasuis (G. parasuis) is commonly located in the upper respiratory tract of pigs as an opportunistic pathogen. It can cause Glässer's disease, which leads to serious economic losses in the swine industry. The occurrence of the disease is often linked with the adhesion and colonization of the pathogen. The PilA pilus subunit is important for adhesion to the host, twitching motility, and biofilm formation in many bacteria. However, no research has focused on the function of PilA in G. parasuis. To further reveal the pathogenesis of G. parasuis and to search for subunit vaccine candidates, we investigated whether PilA could adhere to cells and provide immune protection. A bioinformatic analysis showed that the protein secondary structure of the G. parasuis PilA was similar to that of Haemophilus influenzae (HI). Cell adhesion, ELISA, and far-Western blotting showed that rPilA could bind porcine-derived, porcine kidney-15 (PK-15) cells, swine tracheal epithelial cells (STECs), and the extracellular matrix components fibronectin (FN) and laminin (LN). An immunogenicity analysis showed that recombinant PilA (rPilA) reacted specifically with convalescent and hyperimmune serum. Importantly, purified rPilA elicited a strong immune response and conferred robust protection against challenges with serovar 5 G. parasuis in mice. These results suggested that the PilA protein might help G. parasuis adhere to host cells by binding to FN and LN, and its immunogenicity establishes it as a promising, novel subunit vaccine candidate against infections with G. parasuis. IMPORTANCE G. parasuis is one of the most prevalent bacterial infections in swine production and can lead to huge economic losses around the world. A full understanding of colonization and immunity with G. parasuis infections will be essential in disease control. In this study, the PilA protein, which is a common virulence factor in other bacteria that mediates adherence to the host, was assessed. The results suggested that the PilA protein of G. parasuis can mediate adhesion to host cells through FN and LN, which provides a new idea for the study of the pathogenicity of G. parasuis. Furthermore, fimbriae usually have high immunogenicity. Immunogenicity and protective capacity results showed that the use of this recombinant PilA antigen might be a promising candidate vaccine antigen with which to prevent G. parasuis infections.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Suínos , Camundongos , Fímbrias Bacterianas , Antígenos de Bactérias , Nariz , Vacinas de Subunidades , Infecções por Haemophilus/prevenção & controle , Infecções por Haemophilus/veterinária , Doenças dos Suínos/microbiologia
12.
Res Vet Sci ; 157: 35-39, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36863230

RESUMO

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.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Animais , Suínos , Proteína B de Ligação a Transferrina/química , Proteína B de Ligação a Transferrina/genética , Proteína B de Ligação a Transferrina/metabolismo , Filogenia , Haemophilus parasuis/genética , Infecções por Haemophilus/veterinária , Ferro/metabolismo , Doenças dos Suínos/epidemiologia
13.
Microb Drug Resist ; 29(6): 263-270, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36897759

RESUMO

Glässer's disease is caused by Glaesserella parasuis, a common bacterium in the upper respiratory tract of pigs. Antibiotics are widely used to control this disease. A G. parasuis isolate with amoxicillin (AMX) resistance was identified in our previous study. Outer membrane vesicles (OMVs) are naturally released by G. parasuis and contain many compounds. To reveal the underlying mechanisms associated with AMX resistance delivery, OMVs from G. parasuis were successfully isolated and identified by transmission electron microscopy. In particular, we found that ß-lactamase existed in OMVs using label-free analysis, and further verified that OMVs carry ß-lactamase by Western blotting. The minimal inhibitory concentration and growth rate were determined to evaluate the ß-lactamase activity in G. parasuis OMVs. Moreover, the effect of different concentrations of OMVs from aHPS7 on the growth rate of AMX sensitive strains was examined. Our results further confirmed that OMVs isolated from aHPS7 contain ß-lactamase, which can prevent AMX-susceptible strains from killing by hydrolyzing AMX. Our results first showed that OMVs of G. parasuis play an important role in the spread of antibiotic resistance, which seriously hampered the prevention of this disease by the delivery of OMVs in different strains.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Animais , Suínos , Amoxicilina/farmacologia , Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana , beta-Lactamases , Infecções por Haemophilus/microbiologia
14.
Int J Antimicrob Agents ; 61(4): 106740, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36736498

RESUMO

ICEGpa1804 was identified in the genome of a serovar 2, ST279 isolate EHP1804 carrying eight different resistance genes from 200 Glaesserella parasuis strains isolated from swine with lower respiratory tract infection in seven provinces of China. Susceptibility testing for EHP1804 was determined by broth microdilution, and its genetic profile was determined by whole-genome sequencing. The complete ICEGpa1804 was analysed by polymerase chain reaction, conjugation assay and bioinformatics tools. The conjugation assay was performed using EHP1804 as the donor and G. parasuis V43 (rifampicin-resistant) as the recipient. ICEGpa1804 has a size of 71,880 bp and contains 83 genes, including eight resistance genes [tet(B), blaRob-1, aphA1, strA, strB, aac(3)-IId, catA3 and sul2]. The conjugation assay showed that ICEGpa1804 could be transferred to G. parasuis V43 with frequencies of 4.3 × 10-7. To the best of the authors' knowledge, this is the first study to identify a novel integrative and conjugative element (ICE) carrying eight resistance genes and seven insertion sequence (IS) elements from a G. parasuis isolate. Tn6743, a novel transposon carrying six resistance genes, was identified. Moreover, ISGpa1, a novel IS256 family insertion element, is the first characterized example of a G. parasuis insertion element. Multiple mobile genetic elements involved in resistance genes were located in chromosomal ICEGpa1804, which showed that ICEs may serve as a vital platform for the accumulation of resistance genes.


Assuntos
Haemophilus parasuis , Infecções Respiratórias , Animais , Suínos , Elementos de DNA Transponíveis , Sequenciamento Completo do Genoma , China , Haemophilus parasuis/genética , Conjugação Genética
15.
Virulence ; 14(1): 2171636, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36694280

RESUMO

Haemophilus parasuis (H. parasuis) causes exudative inflammation, implying endothelial dysfunction during pathogen infection. However, so far, the molecular mechanism of endothelial dysfunction caused by H. parasuis has not been clarified. By using the transwell-based cell co-culture system, we demonstrate that knocking out resistin in porcine alveolar macrophages (PAMs) dramatically attenuated endothelial monolayer damage caused by H. parasuis. The resistin secreted by PAMs inhibited the expression of the tight junction proteins claudin-5 and occludin rather than the adherens junction protein VE-cadherin in co-cultured porcine aortic endothelial cells (PAECs). Furthermore, we demonstrate that resistin regulated claudin-5 and occludin expression and monolayer PAEC permeability in an LKB1/AMPK/mTOR pathway-dependent manner. Additionally, we reveal that the outer membrane lipoprotein gene lppA in H. parasuis induced resistin expression in PAMs, as deleting lppA reduced resistin expression in H. parasuis-infected PAMs, causing a significant change in LKB1/AMPK/mTOR pathway activity in co-cultured PAECs, thereby restoring tight junction protein levels and endothelial monolayer permeability. Thus, we postulate that the H. parasuis lppA gene enhances resistin production in PAMs, disrupting tight junctions in PAECs and causing endothelial barrier dysfunction. These findings elucidate the pathogenic mechanism of exudative inflammation caused by H. parasuis for the first time and provide a more profound angle of acute exudative inflammation caused by bacteria.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Suínos , Animais , Macrófagos Alveolares/microbiologia , Haemophilus parasuis/genética , Células Endoteliais , Resistina/genética , Resistina/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Claudina-5/metabolismo , Ocludina/metabolismo , Infecções por Haemophilus/veterinária , Infecções por Haemophilus/microbiologia , Inflamação , Serina-Treonina Quinases TOR/metabolismo
16.
Vet Microbiol ; 276: 109630, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36525718

RESUMO

Glässer's disease is one of the main diseases affecting young piglets, particularly during the nursery phase, that can significantly impact pork production. Vaccination of sows has the potential to prevent Glaesserella parasuis infection during the first weeks of life that is to a substantial degree due to the transfer of maternal derived antibodies (MDA) in colostrum. In this study we compare the antibody response to two vaccines administered to pregnant sows. A subunit vaccine containing the mutant transferrin-binding protein, TbpBY167A, and an autogenous vaccine formulated with the LM96/20 strain of G. parasuis (SV4) administered on days 65 and 86 of the gestational period were safe and induced high titers of antibodies in sows. The IgG peak was reached on day 100 of gestation, and the translocation of IgG to the mammary gland was confirmed in colostrum at the time of delivery. Piglets born from vaccinated sows maintained positive IgG titers against TbpBY167A or G. parasuis SV4 for the duration of the experiment (35 days of life). Piglets born from sows vaccinated with the TbpBY167A-based vaccine had a significantly (p = 0.001) lower load of G. parasuis in the respiratory tract compared to those born from sows vaccinated with the autogenous vaccine. Finally, we demonstrate that the LM96/20 (SV4) strain is highly virulent and a primary agent of Glässer's disease.


Assuntos
Autovacinas , Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Gravidez , Animais , Suínos , Feminino , Vacinação/veterinária , Infecções por Haemophilus/prevenção & controle , Infecções por Haemophilus/veterinária , Vacinas Bacterianas , Doenças dos Suínos/prevenção & controle , Anticorpos Antibacterianos , Imunoglobulina G
17.
Can J Vet Res ; 86(4): 254-260, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36211213

RESUMO

Glässer's disease in pigs is associated with infection by Glaesserella parasuis and is characterized by pneumonia-like symptoms, fibrinous polyserositis, polyarthritis, and meningitis. Macleaya cordata, a commonly used traditional Chinese medication, has been shown to have anti-inflammatory, antiviral, antioxidative, antimicrobial, insecticidal, and antitumor properties. However, the anti-inflammatory effects of M. cordata on G. parasuis stimulation are still poorly understood. This study explored the anti-inflammatory effects and mechanisms of M. cordata extract on G. parasuis-induced inflammatory responses, via the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, in porcine alveolar macrophages (PAMs). Porcine alveolar macrophages, when stimulated with G. parasuis, initiated transcription of interleukin (IL)-1α, IL-1ß, IL-6, IL-8, and tumor necrosis factor alpha (TNF-α). Furthermore, p65, IκBα, p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) phosphorylation were upregulated via the NF-κB and MAPK signaling pathways. However, treatment with M. cordata extract inhibited transcription of IL-1α, IL-1ß, IL-6, IL-8, and TNF-α and reduced p65, IκBα, p38, ERK, and JNK phosphorylation, by inhibiting activation of the NF-κB and MAPK signaling pathways in PAMs induced by G. parasuis. These findings reveal that M. cordata extract can reverse the inflammatory effect initiated by G. parasuis in vitro and that it possesses significant immunosuppression activity; thus, it may offer a novel strategy for controlling and treating G. parasuis infection.


La maladie de Glässer chez les porcs est associée avec une infection par Glaesserella parasuis et est caractérisée par des symptômes similaires à une pneumonie, une polysérosite fibrineuse, une polyarthrite et une méningite. Macleaya cordata, un médicament utilisé couramment en médecine traditionnelle chinoise, a été montré comme ayant des propriétés anti-inflammatoire, antivirale, anti-oxydative, antimicrobienne, insecticide et anti-tumeur. Toutefois, les effets anti-inflammatoires de M. cordata sur une stimulation par G. parasuis sont toujours peu compris. La présente étude explore les effets et mécanismes anti-inflammatoires d'un extrait de M. cordata sur les réponses inflammatoires induites par G. parasuis, via le facteur nucléaire-kappa B (NF-κB) et la voie de signalisation de la protéine kinase activée par les mitogènes (MAPK), dans les macrophages alvéolaires porcins (PAMs). Les PAMs, lorsque stimulés par G. parasuis, ont initié la transcription des interleukines (IL)-1α, IL-1ß, IL-6, IL-8, et le facteur de nécrose des tumeurs alpha (TNF-α). Également, la phosphorylation de p65, IκBα, p38, de la kinase régulée par signal extracellulaire (ERK), et de la kinase c-Jun N-terminal (JNK) était régulée à la hausse via les voies de signalisation NF-κB and MAPK. Toutefois, le traitement avec l'extrait de M. cordata a inhibé la transcription d'IL-1α, IL-1ß, IL-6, IL-8, et TNF-α et a diminué la phosphorylation de p65, IκBα, p38, ERK, et JNK, en inhibant les voies de signalisation de NF-κB et MAPK dans les PAMs induits par G. parasuis. Ces trouvailles révèlent qu'un extrait de M. cordata peut renverser l'effet inflammatoire initié par G. parasuis in vitro et qu'il possède une activité immunosuppressive significative; ainsi, ceci pourrait offrir une nouvelle stratégie pour limiter et traiter l'infection par G. parasuis.(Traduit par Docteur Serge Messier).


Assuntos
Haemophilus parasuis , Doenças dos Suínos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antivirais/metabolismo , Antivirais/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/veterinária , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/farmacologia , Lipopolissacarídeos , Macrófagos Alveolares/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , NF-kappa B/farmacologia , Transdução de Sinais , Suínos , Doenças dos Suínos/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo
18.
J Vet Med Sci ; 84(11): 1527-1535, 2022 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-36216558

RESUMO

Glaesserella parasuis is a pathogen causing Glässer's disease characterized by fibrinous polyserositis, polyarthritis, and meningitis. Owing to the low cross-immunogenicity of different bacterial antigens in commercial vaccines, finding and identifying effective immunoprotective antigens will facilitate the development of novel subunit vaccines. In this study, LolA, identified by bioinformatics approaches, was cloned and successfully expressed as a recombinant protein in Escherichia coli, and its immunogenicity and protection were evaluated in a mouse model. The results showed that the recombinant protein LolA can stimulate mice to produce high levels of IgG antibodies and confer 50% protection against challenge with the highly virulent G. parasuis CY1201 strain (serotype 13). By testing the cytokine levels of interleukin 4 (IL-4), IL-10, and interferon-γ (IFN-γ), it was found that the recombinant protein LolA can induce both Th1 and Th2 immune responses in mice. These results suggest that the recombinant protein LolA has the potential to serve as an alternative antigen for a novel vaccine to prevent G. parasuis infection.


Assuntos
Proteínas de Escherichia coli , Infecções por Haemophilus , Haemophilus parasuis , Proteínas Periplásmicas de Ligação , Doenças dos Roedores , Doenças dos Suínos , Camundongos , Animais , Suínos , Sorogrupo , Anticorpos Antibacterianos , Infecções por Haemophilus/prevenção & controle , Infecções por Haemophilus/veterinária , Proteínas Recombinantes , Vacinação/veterinária , Vacinas de Subunidades , Escherichia coli , Doenças dos Suínos/prevenção & controle
19.
PLoS Pathog ; 18(10): e1010912, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36228044

RESUMO

Glaesserella parasuis (G. parasuis), the primary pathogen of Glässer's disease, colonizes the upper respiratory tract and can break through the epithelial barrier of the respiratory tract, leading to lung infection. However, the underlying mechanisms for this adverse effect remain unclear. The G. parasuis serotype 5 SQ strain (HPS5-SQ) infection decreased the integrity of piglets' lung Occludin and Claudin-1. Autophagy regulates the function of the epithelial barrier and tight junction proteins (TJs) expression. We tested the hypothesis that HPS5-SQ breaking through the porcine respiratory epithelial barrier was linked to autophagy and Claudin-1 degradation. When HPS5-SQ infected swine tracheal epithelial cells (STEC), autophagosomes encapsulated, and autolysosomes degraded oxidatively stressed mitochondria covered with Claudin-1. Furthermore, we found that autophagosomes encapsulating mitochondria resulted in cell membrane Claudin-1 being unable to be replenished after degradation and damaged the respiratory tract epithelial barrier. In conclusion, G. parasuis serotype 5 breaks through the porcine respiratory epithelial barrier by inducing autophagy and interrupting cell membrane Claudin-1 replenishment, clarifying the mechanism of the G. parasuis infection and providing a new potential target for drug design and vaccine development.


Assuntos
Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Suínos , Animais , Claudina-1/metabolismo , Ocludina/metabolismo , Sorogrupo , Haemophilus parasuis/metabolismo , Autofagia , Membrana Celular , Proteínas de Junções Íntimas/metabolismo , Traqueia
20.
Microbiol Spectr ; 10(5): e0230722, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36094311

RESUMO

Glaesserella parasuis is a common bacterium in the porcine upper respiratory tract that causes severe Glasser's disease, which is characterized by polyarthritis, meningitis, and fibrinous polyserositis. TurboID is an enzyme that mediates the biotinylation of endogenous proteins that can fuse with proteins of interest to label protein interactors and local proteomes. To reveal the host proteins that interact with outer membrane protein P2 (OmpP2) by TurboID-mediated proximity labeling in immortalized porcine alveolar macrophage iPAM cells, 0.1 and 2.58 mg/mL His-tagged TurboID-OmpP2 and TurboID recombinant proteins were expressed and purified. By mass spectrometry, we identified 948 and 758 iPAM cell proteins that interacted with His-TurboID-OmpP2 and His-TurboID, respectively. After removal of background proteins through comparison with the TurboID-treated group, 240 unique interacting proteins were identified in the TurboID-OmpP2-treated group. Ultimately, only four membrane proteins were identified, CAV1, ARF6, PPP2R1A, and AP2M1, from these 240 host proteins. Our data indicated that CAV1, ARF6, and PPP2R1A could interact with OmpP2 of G. parasuis, as confirmed by coimmunoprecipitation assay. Finally, we found that CAV1, ARF6, and PPP2R1A were involved in the recognition and phagocytosis of G. parasuis serotype 5 by iPAM cells by using overexpression and RNA interference assays. This study provides first-hand information regarding the interaction of the iPAM cell proteomes with G. parasuis OmpP2 protein by using the TurboID proximity labeling system and identifies three novel host membrane proteins involved in the recognition and phagocytosis of G. parasuis by iPAM cells. These results provide new insight for a better understanding of Glasser's disease pathogenesis. IMPORTANCE G. parasuis can cause serious Glasser's disease, which is characterized by polyarthritis, meningitis, and fibrinous polyserositis in pigs. It can cause high morbidity and mortality in swine herds and major economic losses to the global pig industry. Understanding the mechanism of interactions between alveolar macrophages and pathogenic G. parasuis is essential for developing effective vaccines and targeted drugs against G. parasuis. To reveal the host proteins interacting with OmpP2 by TurboID-mediated proximity labeling in immortalized porcine alveolar macrophage (iPAM) cells, we identified 240 unique proteins from iPAM cells that could interact with G. parasuis OmpP2. Among them, only four membrane proteins, CAV1, ARF6, PPP2R1A, and AP2M1, were identified, and further study showed that CAV1, ARF6, and PPP2R1A are involved in the recognition and phagocytosis of G. parasuis serotype 5 by iPAM cells. This study provides new insight into proteomic interactions between hosts and pathogenic microorganisms.


Assuntos
Artrite , Infecções por Haemophilus , Haemophilus parasuis , Doenças dos Suínos , Suínos , Animais , Macrófagos Alveolares/metabolismo , Proteômica , Proteoma/metabolismo , Haemophilus parasuis/genética , Haemophilus parasuis/metabolismo , Infecções por Haemophilus/veterinária , Infecções por Haemophilus/microbiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Fagocitose , Doenças dos Suínos/microbiologia , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...