Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Front Vet Sci ; 10: 1137967, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37065255

RESUMEN

Novel Duck reovirus (NDRV) is an ongoing non-enveloped virus with ten double-stranded RNA genome segments that belong to the genus Orthoreovirus, in the family Reoviridae. NDRV-associated spleen swelling, and necrosis disease have caused considerable economic losses to the waterfowl industry worldwide. Since 2017, a significant number of NDRV outbreaks have emerged in China. Herein, we described two cases of duck spleen necrosis disease among ducklings on duck farms in Henan province, central China. Other potential causative agent, including Muscovy duck reovirus (MDRV), Duck hepatitis A virus type 1 (DHAV-1), Duck hepatitis A virus type 3 (DHAV-3), Newcastle disease virus (NDV), and Duck tembusu virus (DTMUV), were excluded by reverse transcription-polymerase chain reaction (RT-PCR), and two NDRV strains, HeNXX-1/2021 and HNJZ-2/2021, were isolated. Sequencing and phylogenetic analysis of the σC genes revealed that both newly identified NDRV isolates were closely related to DRV/SDHZ17/Shandong/2017. The results further showed that Chinese NDRVs had formed two distinct clades, with late 2017 as the turning point, suggesting that Chinese NDRVs have been evolving in different directions. This study identified and genetic characteristics of two NDRV strains in Henan province, China, indicating NDRVs have evolved in different directions in China. This study provides an insight into the ongoing emerged duck spleen necrosis disease and enriches our understanding of the genetic diversity and evolution of NDRVs.

2.
Virulence ; 12(1): 2296-2313, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34482810

RESUMEN

Airway epithelial cells are the first line of defense against respiratory pathogens. Porcine bacterial pathogens, such as Bordetella bronchiseptica, Actinobacillus pleuropneumoniae, Glaesserella (Haemophilus) parasuis, and Pasteurella multocida, breach this barrier to lead to local or systematic infections. Here, we demonstrated that respiratory bacterial pathogen infection disrupted the airway epithelial intercellular junction protein, E-cadherin, thus contributing to impaired epithelial cell integrity. E-cadherin knocking-out in newborn pig tracheal cells via CRISPR/Cas9 editing technology confirmed that E-cadherin was sufficient to suppress the paracellular transmigration of these porcine respiratory bacterial pathogens, including G. parasuis, A. pleuropneumoniae, P. multocida, and B. bronchiseptica. The E-cadherin ectodomain cleavage by these pathogens was probably attributed to bacterial HtrA/DegQ protease, but not host HtrA1, MMP7 and ADAM10, and the prominent proteolytic activity was further confirmed by a serine-to-alanine substitution mutation in the active center of HtrA/DegQ protein. Moreover, deletion of the htrA gene in G. parasuis led to severe defects in E-cadherin ectodomain cleavage, cell adherence and paracellular transmigration in vitro, as well as bacterial breaking through the tracheal epithelial cells, systemic invasion and dissemination in vivo. This common pathogenic mechanism shared by other porcine respiratory bacterial pathogens explains how these bacterial pathogens destroy the airway epithelial cell barriers and proliferate in respiratory mucosal surface or other systemic tissues.


Asunto(s)
Infecciones Bacterianas , Cadherinas , Infecciones del Sistema Respiratorio , Enfermedades de los Porcinos , Actinobacillus pleuropneumoniae , Animales , Infecciones Bacterianas/veterinaria , Bordetella bronchiseptica , Cadherinas/genética , Células Epiteliales/microbiología , Haemophilus parasuis , Pasteurella multocida , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/veterinaria , Porcinos , Enfermedades de los Porcinos/microbiología
3.
Foodborne Pathog Dis ; 18(10): 733-743, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34143653

RESUMEN

Escherichia coli is an important foodborne pathogen and also plays key roles in dissemination of antimicrobial resistance genes (ARGs). However, current data on the prevalence of antimicrobial-resistant E. coli at different nodes of the pork supplying chain are still limited. Herein, we investigated drug-resistant phenotypes and molecular characteristics of E. coli strains isolated from different pig farms, slaughterhouses, and terminal markets in the Henan Province of China. A total of 191 (70.74%), 140 (35.09%), and 77 (30.20%) E. coli strains were isolated from 270, 399, and 255 samples collected from pig farms, slaughterhouses, and retailing markets, respectively. Antimicrobial susceptibility testing revealed that these 408 strains showed severe antimicrobial resistance profiles. Approximately 93.19% (178/191), 66.43% (93/140), and 67.53% (52/77) of the isolates from farms, slaughterhouses, and terminal markets were resistant to three of the nine antibiotic classes tested, respectively. Multilocus sequence typing showed that sequence types (STs) 10 and ST101 were commonly identified among the isolates from farms, slaughterhouses, and terminal markets. Isolates belonging to these two STs carried multiple ARGs, conferring resistance to the antibiotics tested. Two important ARGs with great public health concerns (mcr-1 and blaNDM-1) were found from these two STs. Isolates belonging to these two STs also carried several virulence factor-encoding genes, including astA, tsh, and traT, which might contribute to the pathogenesis of these isolates. The wide prevalence and distribution of these two STs in different nodes of pork supplying chain might represent a big public health threat and should receive more attention.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Mataderos , Animales , Antibacterianos/farmacología , China/epidemiología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Granjas , Pruebas de Sensibilidad Microbiana , Prevalencia , Porcinos
5.
ACS Infect Dis ; 7(3): 661-671, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33645216

RESUMEN

Pathogens utilize various mechanisms to escape host immunological surveillance, break down different tissue barriers, and cause infection. Sialylation is an important surface modification of bacterial outer membrane components, especially the lipooligosaccharide of Gram-negative bacteria. It is widely involved in multiple microbe-host interactions, such as bacterial virulence regulation, host recognition, and immune evasion. There are some sialylation modifications on the lipooligosaccharide structure of Glaesserella parasuis (G. parasuis) virulent strains. However, the role of lipooligosaccharide sialylation modification in the process of G. parasuis infection and penetration of the porcine respiratory epithelial barrier is still unclear. In this study, we investigated the role and mechanism of lsgB-mediated lipooligosaccharide sialylation in G. parasuis invasion of the host respiratory epithelial barrier. Specifically, G. parasuis lsgB-mediated lipooligosaccharide sialylation and sialylated-lipooligosaccharide interacted with Siglec1 on porcine alveolar macrophages 3D4/21 and triggered the subsequent generation of TGFß1 through Siglec1/Dap12/Syk/p38 signaling cascade. TGFß1 decreased the tracheal epithelial tight junctions and the expression of extracellular adhesion molecule fibronectin, thus assisting G. parasuis invasion and entry to the respiratory epithelial barrier. Characterizing the potential effects and mechanisms of lipooligosaccharide sialylation-mediated TGFß1 production would further expand our current knowledge on the pathogenesis of G. parasuis which will contribute to better prevention and control of G. parasuis infection in piglets.


Asunto(s)
Haemophilus parasuis , Animales , Lipopolisacáridos , Transducción de Señal , Porcinos
6.
ACS Infect Dis ; 7(4): 777-789, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33723986

RESUMEN

Meningitic Escherichia coli can infiltrate the central nervous system (CNS), consequently increasing the levels of proinflammatory cytokines and chemokines and deteriorating the integrity of the blood-brain barrier (BBB). Resveratrol has emerged in recent years as a compound with antioxidant and anti-inflammatory properties. However, it is still unknown how resveratrol affects meningitic E. coli-induced CNS dysfunction. Here, by using in vivo and in vitro BBB models, we demonstrated that resveratrol treatment significantly inhibited meningitic E. coli invasion of the BBB, protected the integrity of the BBB, and reduced neuroinflammation and lethality. In mechanism, resveratrol inhibited bacterial penetration of the BBB by attenuating the upregulation of caveolin-1 (CAV-1), a class of lipid rafts maintaining endothelial cell function. Resveratrol treatment also maintained BBB permeability by suppressing the ERK1/2-VEGFA signaling cascade. In vivo treatment of resveratrol decreased the production of inflammatory cytokines and improved the survival rate in mice challenged with meningitic E. coli. These findings collectively indicated that resveratrol could attenuate meningitic E. coli-induced CNS injury, which might constitute a new approach for future prevention and treatment of E. coli meningitis.


Asunto(s)
Barrera Hematoencefálica , Meningitis por Escherichia coli , Animales , Células Endoteliales , Escherichia coli , Ratones , Resveratrol/farmacología
7.
Animals (Basel) ; 10(10)2020 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-33020441

RESUMEN

The outbreak of pseudorabies in China, caused by more virulent pseudorabies virus (PRV) than the classical strains, has led to considerable economic losses. In this study, PRV strain HNXY was isolated from the Henan province of China in 2015 from the pig farm with severe reproductive failure in sows and a high mortality in piglets. The 50% tissue culture infectious doses (TCID50) of HNXY in Vero cells were examined to be 106.5/mL, and the neutralisation titer against Bartha-K61 was significantly higher than against HNXY when tested with the serum from Bartha-K61 vaccinated pigs. The 50% lethal doses (LD50) of HNXY to six-week-old BALB/c mice and two-month-old PRV-free pigs were both 102.3 TCID50. HNXY was classified as genotype II, and numerous amino acid variations were found in gB, gE, gC, gD, TK, and RR1 proteins, compared with PRV from other countries or those prevalent in China before 2012. The attenuated rHNXY-∆TK/∆gE was further constructed, which presented significantly smaller plaques than HNXY, as well as the similar growth kinetics. rHNXY-∆TK/∆gE was confirmed to be non-pathogenic to six-week-old BALB/c mice and zero-day-old piglets. This study isolated updated PRV promising to develop into a new vaccine candidate.

8.
Pathogens ; 8(4)2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766159

RESUMEN

Glaesserella parasuis is a habitual bacterium of pigs' upper respiratory tracts. Its infection initiates with the invasion and colonization of the lower respiratory tracts of pigs, and develops as the bacteria survive host pulmonary defenses and clearance by alveolar macrophages. Alveolar macrophage-derived nitric oxide (NO) is recognized as an important mediator that exerts antimicrobial activity as well as immunomodulatory effects. In this study, we investigated the effects and the signaling pathway of NO generation in porcine alveolar macrophages 3D4/21 during G. parasuis infection. We demonstrated a time and dose-dependent generation of NO in 3D4/21 cells by G. parasuis, and showed that NO production required bacterial viability and nitric oxide synthase 2 upregulation, which was largely contributed by G. parasuis-induced nuclear factor-κB signaling's activation. Moreover, the porcine alveolar macrophage-derived NO exhibited prominent bacteriostatic effects against G. parasuis and positive host immunomodulation effects by inducing the production of cytokines and chemokines during infection. G. parasuis in turn, selectively upregulated several nitrate reductase genes to better survive this NO stress, revealing a battle of wits during the bacteria-host interactions. To our knowledge, this is the first direct demonstration of NO production and its anti-infection effects in alveolar macrophages with G. parasuis infection.

9.
Microorganisms ; 7(11)2019 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-31652858

RESUMEN

The emergence of carbapenem-resistant and colistin-resistant Enterobacteriaceae represents a great risk for public health. In this study, the phenotypical and genetic characteristics of eight carbapenem-resistant and colistin-resistant isolates from pig farms in China were determined by the broth microdilution method and whole genome sequencing. Antimicrobial susceptibility testing showed that the eight carbapenem-resistant and colistin-resistant strains were resistant to three aminoglycosides, twelve ß-lactams, one of the phenicols, one of the tetracyclines, and one of the fluoroquinolones tested, simultaneously. The prediction of acquired resistant genes using the whole genome sequences revealed the co-existence of blaNDM-1 and mcr-1 as well as the other genes that were responsible for the multidrug-resistant phenotypes. Bioinformatics analysis also showed that the carbapenem-resistant gene blaNDM-1 was located on a putative IncFII-type plasmid, which also carried the other acquired resistant genes identified, including fosA3, blaTEM-1B and rmtB, while the colistin-resistant gene mcr-1 was carried by a putative IncX4-type plasmid. Finally, we found that these resistant genes/plasmids were conjugative, and they could be co-conjugated, conferring resistance to multiple types of antibiotics, including the carbapenems and colistin, to the recipient Escherichia coli strains.

10.
J Neuroinflammation ; 16(1): 101, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092253

RESUMEN

BACKGROUND: Blood-brain barrier (BBB) disruption and neuroinflammation are considered key mechanisms of pathogenic Escherichia coli invasion of the brain. However, the specific molecules involved in meningitic E. coli-induced BBB breakdown and neuroinflammatory response remain unclear. Our previous RNA-sequencing data from human brain microvascular endothelial cells (hBMECs) revealed two important host factors: platelet-derived growth factor-B (PDGF-B) and intercellular adhesion molecule-1 (ICAM-1), which were significantly upregulated in hBMECs after meningitic E. coli infection. Whether and how PDGF-B and ICAM-1 contribute to the development of E. coli meningitis are still unclear. METHODS: The western blot, real-time PCR, enzyme-linked immunosorbent assay, immunohistochemistry, and immunofluorescence were applied to verify the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in vivo and in vitro. Evan's blue assay and electric cell-substrate impedance sensing assay were combined to identify the effects of PDGF-B on BBB permeability. The CRISPR/Cas9 technology, cell-cell adhesion assay, and electrochemiluminescence assay were used to investigate the role of ICAM-1 in neuroinflammation subversion. RESULTS: We verified the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in mouse as well as monolayer hBMECs models. Functionally, we showed that the increase of PDGF-B may directly enhance the BBB permeability by decreasing the expression of tight junction proteins, and the upregulation of ICAM-1 contributed to neutrophils or monocytes recruitment as well as neuroinflammation subversion in response to meningitic E. coli infection. CONCLUSIONS: Our findings demonstrated the roles of PDGF-B and ICAM-1 in mediating bacterial-induced BBB damage as well as neuroinflammation, providing new concepts and potential targets for future prevention and treatment of bacterial meningitis.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Infecciones por Escherichia coli/metabolismo , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Linfocinas/biosíntesis , Meningitis Bacterianas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Células Cultivadas , Escherichia coli , Infecciones por Escherichia coli/patología , Femenino , Meningitis Bacterianas/patología , Ratones , Uniones Estrechas/metabolismo , Uniones Estrechas/microbiología , Regulación hacia Arriba/fisiología
11.
J Neuroinflammation ; 15(1): 291, 2018 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340642

RESUMEN

BACKGROUND: Bacterial meningitis remains a big threat to the integrity of the central nervous system (CNS), despite the advancements in antimicrobial reagents. Escherichia coli is a bacterial pathogen that can disrupt the CNS function, especially in neonates. E. coli meningitis occurs after bacteria invade the brain microvascular endothelial cells (BMECs) that form a direct and essential barrier restricting the entry of circulating microbes and toxins to the brain. Previous studies have reported on several cellular proteins that function during meningitic E. coli infections; however, more comprehensive investigations to elucidate the potential targets involved in E. coli meningitis are essential to better understand this disease and discover new treatments for it. METHODS: The isobaric tags for relative and absolute quantification (iTRAQ) approach coupled with LC-MS/MS were applied to compare and characterize the different proteomic profiles of BMECs in response to meningitic or non-meningitic E. coli strains. KEGG and gene ontology annotations, ingenuity pathways analysis, and functional experiments were combined to identify the key host molecules involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammatory responses. RESULTS: A total of 13 cellular proteins were found to be differentially expressed by meningitic E. coli strains PCN033 and RS218, including one that was also affected by HB101, a non-meningitic E. coli strain. Through bioinformatics analysis, we identified the macrophage migration inhibitory factor (MIF), granzyme A, NF-κB signaling, and mitogen-activated protein kinase (MAPK) pathways as being biologically involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammation. Functionally, we showed that MIF facilitated meningitic E. coli-induced production of cytokines and chemokines and also helped to disrupt the blood-brain barrier by decreasing the expression of tight junction proteins like ZO-1, occludin. Moreover, we demonstrated the significant activation of NF-κB and MAPK signaling in BMECs in response to meningitic E. coli strains, which dominantly determined the generation of the proinflammatory cytokines including IL-6, IL-8, TNF-α, and IL-1ß. CONCLUSIONS: Our work identified 12 host cellular targets that are affected by meningitic E. coli strains and revealed MIF to be an important contributor to meningitic E. coli-induced cytokine production and tight junction disruption, and also the NF-κB and MAPK signaling pathways that are mainly involved in the infection-induced cytokines production. Characterization of these distinct proteins and pathways in BMECs will facilitate further elucidation of meningitis-causing mechanisms in humans and animals, thereby enabling the development of novel preventative and therapeutic strategies against infection with meningitic E. coli.


Asunto(s)
Encéfalo/citología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Proteómica/métodos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Biología Computacional , Citocinas/genética , Citocinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/farmacología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/farmacología , Meningitis por Escherichia coli/metabolismo , Meningitis por Escherichia coli/patología , FN-kappa B/metabolismo , Transducción de Señal/fisiología
12.
Virulence ; 9(1): 1247-1262, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30036124

RESUMEN

Bacterial lipooligosaccharide (LOS) is an important virulence-associated factor, and its sialylation largely confers its ability to mediate cell adhesion, invasion, inflammation, and immune evasion. Here, we investigated the function of the Haemophilus parasuis α-2,3-sialyltransferase gene, lsgB, which determines the terminal sialylation of LOS, by generating a lsgB deletion mutant as well as a complementation strain. Our data indicate a direct effect of lsgB on LOS sialylation and reveal important roles of lsgB in promoting the pathogenicity of H. parasuis, including adhesion to and invasion of porcine cells in vitro, bacterial load and survival in vivo, as well as a contribution to serum resistance. These observations highlight the function of lsgB in mediating LOS sialylation and more importantly its role in H. parasuis infection. These findings provide a more profound understanding of the pathogenic mechanism of this disease-causing bacterium.


Asunto(s)
Haemophilus parasuis/genética , Haemophilus parasuis/patogenicidad , Lipopolisacáridos/química , Sialiltransferasas/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Eliminación de Gen , Prueba de Complementación Genética , Haemophilus parasuis/enzimología , Mutación , Porcinos , Virulencia , Factores de Virulencia/genética
13.
Sci Rep ; 5: 10864, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242322

RESUMEN

The response regulator OtpR is critical for the growth, morphology and virulence of Brucella melitensis. Compared to its wild type strain 16 M, B. melitensis 16 MΔotpR mutant has decreased tolerance to acid stress. To analyze the genes regulated by OtpR under acid stress, we performed RNA-seq whole transcriptome analysis of 16 MΔotpR and 16 M. In total, 501 differentially expressed genes were identified, including 390 down-regulated and 111 up-regulated genes. Among these genes, 209 were associated with bacterial metabolism, including 54 genes involving carbohydrate metabolism, 13 genes associated with nitrogen metabolism, and seven genes associated with iron metabolism. The 16 MΔotpR also decreased capacity to utilize different carbon sources and to tolerate iron limitation in culture experiments. Notably, OtpR regulated many Brucella virulence factors essential for B. melitensis intracellular survival. For instance, the virB operon encoding type IV secretion system was significantly down-regulated, and 36 known transcriptional regulators (e.g., vjbR and blxR) were differentially expressed in 16 MΔotpR. Selected RNA-seq results were experimentally confirmed by RT-PCR and RT-qPCR. Overall, these results deciphered differential phenomena associated with virulence, environmental stresses and cell morphology in 16 MΔotpR and 16 M, which provided important information for understanding the detailed OtpR-regulated interaction networks and Brucella pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella melitensis/genética , ARN Bacteriano/química , Ácidos/farmacología , Proteínas Bacterianas/genética , Secuencia de Bases , Brucella melitensis/metabolismo , Brucella melitensis/patogenicidad , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético , ARN Bacteriano/aislamiento & purificación , ARN Bacteriano/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia , Análisis de Secuencia de ARN , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA