Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Virol ; 95(18): e0092021, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34191582

RESUMEN

K1 capsule-specific phages of Escherichia coli have been reported in recent years, but the molecular mechanism involved in host recognition of these phages remains unknown. In this study, the interactions between PNJ1809-36, a new K1-specific phage, and its host bacterium, E. coli DE058, were investigated. A transposon mutation library was used to screen for receptor-related genes. Gene deletion, lysis curve determination, plaque formation test, adsorption assay, and inhibition assay of phage by lipopolysaccharide (LPS) showed that capsular polysaccharide (CPS) was the first receptor for the initial adsorption of PNJ1809-36 to E. coli DE058 and that LPS was a secondary receptor for the irreversible binding of the phage. The penultimate galactose in the outer core was identified as the specific binding region on LPS. Through antibody blocking assay, fluorescence labeling and high-performance gel permeation chromatography, the tail protein ORF261 of phage PNJ1809-36 was identified as the receptor-binding protein on CPS. Given these findings, we propose a model for the recognition process of phage PNJ1809-36 on E. coli DE058: the phage PNJ1809-36 tail protein ORF261 recognizes and adsorbs to the K1 capsule, and then the K1 capsule is partially degraded, exposing the active site of LPS which is recognized by phage PNJ1809-36. This model provides insight into the molecular mechanisms between K1-specific phages and their host bacteria. IMPORTANCE It has been speculated that CPS is the main receptor of K1-specific phages belonging to Siphoviridae. In recent years, a new type of K1-specific phage belonging to Myoviridae has been reported, but its host recognition mechanisms remain unknown. Here, we studied the interactions between PNJ1809-36, a new type of K1 phage, and its host bacterium, E. coli DE058. Our research showed that the phage initially adsorbed to the K1 capsule mediated by ORF261 and then bound to the penultimate galactose of LPS to begin the infection process.


Asunto(s)
Antígenos Bacterianos/metabolismo , Cápsulas Bacterianas/metabolismo , Bacteriófago T7/fisiología , Escherichia coli/metabolismo , Lipopolisacáridos/metabolismo , Polisacáridos Bacterianos/metabolismo , Secuencia de Aminoácidos , Escherichia coli/virología , Homología de Secuencia de Aminoácido
2.
Vet Res ; 52(1): 118, 2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521472

RESUMEN

Escherichia coli (E. coli) O157:H7 bacterial infection causes severe disease in mammals and results in substantial economic losses worldwide. Due to the development of antibiotic resistance, bacteriophage (phage) therapy has become an alternative to control O157:H7 infection. However, the therapeutic effects of phages are frequently disappointing because of their low resistance to the gastrointestinal environment. In this study, to improve the stability of phages in the gastrointestinal tract, E. coli O157:H7 phages were microencapsulated and their in vitro stability and in vivo therapeutic efficiency were investigated. The results showed that compared to free phages, the resistance of microencapsulated phages to simulated gastric fluid and bile salts significantly increased. The microencapsulated phages were efficiently released into simulated intestinal fluid, leading to a better therapeutic effect in rats infected with E. coli O157:H7 compared to the effects of the free phages. In addition, the microencapsulated phages were more stable during storage than the free phages, showing how phage microencapsulation can play an essential role in phage therapy.


Asunto(s)
Colifagos/fisiología , Infecciones por Escherichia coli/prevención & control , Escherichia coli O157/fisiología , Enfermedades Gastrointestinales/prevención & control , Tracto Gastrointestinal/microbiología , Animales , Infecciones por Escherichia coli/microbiología , Femenino , Enfermedades Gastrointestinales/microbiología , Ratas , Ratas Sprague-Dawley
3.
Vet Res ; 51(1): 5, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973724

RESUMEN

Avian pathogenic Escherichia coli (APEC), a pathotype of extraintestinal pathogenic E. coli, causes one of the most serious infectious diseases of poultry and shares some common virulence genes with neonatal meningitis-associated E. coli. TonB-dependent receptors (TBDRs) are ubiquitous outer membrane ß-barrel proteins; they play an important role in the recognition of siderophores during iron uptake. Here, in the APEC strain DE205B, we investigated the role of four putative TBDRs-ireA, 0007, 0008, and 2235-in iron uptake. Glutathione-S-transferase pulldown assays indicated that the proteins encoded by these genes directly interact with TonB. Moreover, the expression levels of all four genes were significantly upregulated under iron-depleted conditions compared with iron-rich conditions. The expression levels of several iron uptake-related genes were significantly increased in the ireA, 0007, 0008, and 2235 deletion strains, with the upregulation being the most prominent in the ireA deletion mutant. Furthermore, iron uptake by the ireA deletion strain was significantly increased compared to that by the wild-type strain. Moreover, a tonB mutant strain was constructed to study the effect of tonB deletion on the TBDRs. We found that regardless of the presence of tonB, the expression levels of the genes encoding the four TBDRs were regulated by fur. In conclusion, our findings indicated that ireA, 0007, 0008, and 2235 indeed encode TBDRs, with ireA having the most important role in iron uptake. These results should help future studies explore the mechanisms underlying the TonB-dependent iron uptake pathway.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Enfermedades de las Aves de Corral/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/metabolismo , Pollos , Escherichia coli/metabolismo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Enfermedades de las Aves de Corral/microbiología
4.
Analyst ; 145(8): 3106-3115, 2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32159201

RESUMEN

Escherichia coli O157:H7 is a severe foodborne pathogen that causes lots of life-threatening diseases. In the search for a rapid, sensitive, portable and low-cost method to detect this pathogen, we developed a wax-printed paper-based enzyme-linked immunosorbent assay (P-ELISA) based on microfluidic paper-based analytical devices (µPADs), with the whole operation time being less than 3 h and only needing 5 µl samples for detection. The limit of detection (LOD) of E. coli O157:H7 reached 104 CFU ml-1, which is an order of magnitude higher than that of conventional ELISA (C-ELISA). The LOD in artificially contaminated beef samples is 1 CFU per 25 g after enriching the culture for 8 h. This method is superior to the molecular biology method in detection sensitivity and superior to C-ELISA and the national standard method in detection time and cost. Thus, the established P-ELISA method has good sensitivity, specificity and repeatability. It can be suitable for point-of-care testing without expensive and bulky instruments and can also provide a platform for detecting other pathogens, especially in areas that lack advanced clinical equipment.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Escherichia coli O157/aislamiento & purificación , Contaminación de Alimentos/análisis , Papel , Carne Roja/microbiología , Ceras , Animales , Carga Bacteriana/instrumentación , Carga Bacteriana/métodos , Bovinos , Ensayo de Inmunoadsorción Enzimática/instrumentación , Límite de Detección , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Sensibilidad y Especificidad , Teléfono Inteligente
5.
Mikrochim Acta ; 187(12): 642, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33155077

RESUMEN

A molecularly imprinted electrochemical sensor for the detection of serum amyloid A (MAA) in milk was established for early diagnosis of subclinical mastitis in dairy cows. The electrochemical sensor was initially constructed using a nanocomposite material (reduced graphene oxide/gold nanoparticles, AuNPs@rGO) to modify the working electrode. The template protein, MAA, was then immobilized using pyrrole as the functional monomer to carry out the electropolymerization. Finally, the template protein was removed to form a molecular imprint film with the capability to qualitatively and quantitatively signaling of MAA. Cyclic voltammetry (CV), differential pulse voltammetry (DPV), and scanning electron microscopy (SEM) were used to characterize the modification process of the molecularly imprinted electrochemical sensors. Under optimized conditions, the sensor shows two well-behaved linear relationships in the MAA concentration range 0.01 to 200 ng/mL. A lower detection limit was estimated to be 5 pg/mL (S/N = 3). Other parameters including the selectivity, reproducibility (RSD 3.2%), and recovery rate (96.1-103%) are all satisfactory. Compared with the traditional methods, detection of MAA to determine the subclinical mastitis of dairy cows can efficiently be diagnosed and hence prevent an outbreak of dairy cow mastitis. The electrochemical sensor can detect MAA more rapidly, sensitively, and inexpensively than the ELISA-based MAA detection. These advantages indicate that the method is promising for early diagnosis of dairy cows.


Asunto(s)
Técnicas Electroquímicas/instrumentación , Leche/química , Polímeros Impresos Molecularmente/química , Proteína Amiloide A Sérica/análisis , Animales , Bovinos , Industria Lechera , Diagnóstico Precoz , Femenino , Oro/química , Grafito/química , Límite de Detección , Mastitis Bovina/diagnóstico , Nanopartículas del Metal/química , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Reproducibilidad de los Resultados , Espectrofotometría Ultravioleta
6.
BMC Genomics ; 20(1): 212, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30866824

RESUMEN

BACKGROUND: Escherichia coli is an important pathogen that causes diarrhea in both humans and animals. To determine the relationships between putative virulence factors and pathotypes or host taxa, many molecular studies on diarrhea-associated E. coli have been reported. However, little is known regarding genome-wide variation of E. coli from animal hosts. In this study, we performed whole genome sequencing of 127 E. coli isolates from sheep and swine with diarrhea in China. We compared isolates to explore the phylogenomic relatedness based on host origin. We explored the relationships of putative virulence factors across host taxa and pathotypes. Antimicrobial resistance was also tested. RESULTS: The E. coli genomes in this study were diverse with clear differences in the SNP, MLST, and O serotypes. Seven putative virulence factors (VFs) were prevalent (> 95%) across the isolates, including Hcp, csgC, dsdA, feoB, fepA, guaA, and malX. Sixteen putative VFs showed significantly different distributions (P < 0.05) in strains from sheep and swine and were primarily adhesion- and toxin-related genes. Some putative VFs were co-occurrent in some specific pathotypes and O serotypes. The distribution of 4525 accessory genes of the 127 strains significantly differed (P < 0.05) between isolates obtained from the two animal species. The 127 animal isolates sequenced in this study were each classified into one of five pathotypes: EAEC, ETEC, STEC, DAEC, and EPEC, with 66.9% of isolates belonging to EAEC. Analysis of stx subtypes and a minimum spanning tree based on MLST revealed that STEC isolates from sheep and EAEC isolates from sheep and swine have low potential to infect humans. Antibiotic resistance analysis showed that the E. coli isolates were highly resistant to ampicillin and doxycycline. Isolates from southeast China were more resistant to antibiotics than isolates from northwest China. Additionally, the plasmid-mediated colist in resistance gene mcr-1 was detected in 15 isolates, including 4 from sheep in Qinghai and 11 from swine in Jiangsu. CONCLUSIONS: Our study provides insight into the genomes of E. coli isolated from animal sources. Distinguishable differences between swine and sheep isolates at the genomic level provides a baseline for future investigations of animal E. coli pathogens.


Asunto(s)
Animales Domésticos/microbiología , Diarrea/microbiología , Infecciones por Escherichia coli/veterinaria , Escherichia coli/clasificación , Genómica/métodos , Animales , Técnicas de Tipificación Bacteriana , China , Diarrea/veterinaria , Farmacorresistencia Bacteriana , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/genética , Tipificación de Secuencias Multilocus , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos , Serogrupo , Ovinos/microbiología , Porcinos/microbiología , Factores de Virulencia/genética
7.
Microb Pathog ; 126: 193-198, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30408490

RESUMEN

Salmonella are causes of livestock, poultry, and other animal diseases but they also have the potential to infect people. Currently, antibiotics are the first choice for treatment of Salmonella infections. Thus, the utility of phage has become the research focus for scientists for several reasons. There are efficient, non-toxic, ubiquitous, easy to prepare and can result in the lysis of host bacteria. In this study, a broad spectrum bacteriophage of Salmonella was isolated from the fecal samples of a poultryfarm and we studied the morphological aspects, thermal stability, pH stability, optimal multiplicity of infection (MOI), and one-step growth curve of this phage. This phage was named Salmonella phage SaFB14 and lysed 54.12%(105/194)Salmonella spp. SaFB14 belongs to the Siphoviridae and has a polyhedron head with a diameter approximating 60 × 60 nm and a tail approximating 140 nm. The optimum growth temperature was 37 °C and maintained high activity over a widepH range(pH3-10) with an optimum of pH 7.0. The optimal MOI was 0.1. A one-step growth curve showed that its latency time was 10 min, burst time was 70 min, and burst was 23 particles. In order to study the therapeutic effect of phage SaFB14 in infected mice, mice were challenged with 2 × 109 CFU/mouse Salmonella (cs20130523-001-1). Each mouse was injected to 2 × 1010 PFU SaFB14 1 h later. SaFB14 protected 40% of mice from infection. Then, the same dose of phage was given to mice for 3 days continuously. After 3 days treatment, the survival rate increased to 60%.In conclusion, phage SaFB14 showed wide host range and good activity in vivo, it is promising against diseases caused by Salmonella.


Asunto(s)
Bacteriófagos/clasificación , Bacteriófagos/aislamiento & purificación , Farmacorresistencia Bacteriana Múltiple , Terapia de Fagos , Infecciones por Salmonella/terapia , Salmonella/virología , Animales , Bacteriófagos/genética , Bacteriófagos/crecimiento & desarrollo , ADN Viral/genética , Modelos Animales de Enfermedad , Granjas , Heces/microbiología , Heces/virología , Femenino , Genoma Viral , Especificidad del Huésped , Concentración de Iones de Hidrógeno , Ratones , Viabilidad Microbiana , Aves de Corral/microbiología , Aves de Corral/virología , Salmonella/patogenicidad , Siphoviridae/clasificación , Tasa de Supervivencia , Temperatura
8.
Vet Res ; 50(1): 31, 2019 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046828

RESUMEN

Avian pathogenic Escherichia coli (APEC) is a facultative intracellular pathogen, and intracellular persistence in macrophages is essential for APEC extraintestinal dissemination. Until now, there is still no systematic interpretation of APEC intracellular proliferation. Intracellular survival factors, especially involved in pathometabolism, need to be further revealed. Acetate plays critical roles in supporting energy homeostasis and acts as a metabolic signal in the inflammatory response of eukaryotes. In this study, we identified that APEC acs-yjcH-actP operon, encoding acetate assimilation system, presented the host-induced transcription during its proliferation in macrophages. Our result showed that this acetate assimilation system acted as a novel intracellular survival factor to promote APEC replication within macrophages. Furthermore, deletion of acs-yjcH-actP operon in APEC decreased its cytotoxic level to macrophages. qRT-PCR results showed that the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8, IL-12ß, and TNF-α) and iNOS in FY26∆acs-yjcH-actP infected macrophages were obviously down-regulated compared to that in wild-type FY26 infected cells. Deletion of actP/yjcH/acs genes attenuated APEC virulence and colonization capability in avian lungs in vivo for colibacillosis infection models. And acetate assimilation system acted as a virulence factor and conferred a fitness advantage during APEC early colonization. Taken together, our research unravelled the metabolic requirement of APEC intracellular survival/replication within macrophages, and acetate metabolic requirement acted as an important strategy of APEC pathometabolism. The intracellular acetate consumption during facultative intracellular bacteria replication within macrophages promoted immunomodulatory disorders, resulting in excessively pro-inflammatory responses of host macrophages.


Asunto(s)
Acetatos/metabolismo , Enfermedades de las Aves/microbiología , Infecciones por Escherichia coli/veterinaria , Escherichia coli/metabolismo , Macrófagos/microbiología , Animales , Línea Celular , Pollos , Citocinas/metabolismo , Patos/microbiología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/crecimiento & desarrollo , Escherichia coli/fisiología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Enfermedades de las Aves de Corral/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Microb Pathog ; 100: 56-61, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27616446

RESUMEN

Chaperone-usher (CU) fimbriae, which are adhesive surface organelles found in many Gram-negative bacteria, mediate tissue tropism through the interaction of fimbrial adhesins with specific receptors expressed on the host cell surface. A CU fimbrial gene yfcO, was identified in avian pathogenic E. coli (APEC) strain DE205B via gene functional analysis. In this study, yfcO was found in 13.41% (11/82) of E. coli strains, including phylogenetic groups A, B1, B2 and D, with the highest percentage in group B2. The expression of yfcO in biofilm forming bacteria was significantly higher (P < 0.05) than that in the planktonic bacteria. A yfcO deletion mutant was constructed, and adherence to DF-1 chicken embryo fibroblast cells was analyzed in vitro. Compared to the wild-type (WT), adherence of the mutant to DF-1 cells was significantly decreased (P < 0.01). The mutant bacterial loads in the heart, brain and liver were significantly lower (P < 0.05) than those of the WT strain. Resistance of the mutant to acidic (acetic, pH 4.0, 20 min) and high osmolarity (2.5 M NaCl, 1 h) stress conditions decreased by 51.28% (P < 0.001) and 80.34% (P < 0.01), respectively. These results suggest that yfcO contributes to APEC virulence through bacterial adherence to host tissues.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Infecciones por Escherichia coli/patología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiología , Escherichia coli/patogenicidad , Fimbrias Bacterianas/fisiología , Adhesinas Bacterianas/genética , Estructuras Animales/microbiología , Animales , Carga Bacteriana , Proteínas de la Membrana Bacteriana Externa/genética , Biopelículas/crecimiento & desarrollo , Línea Celular , Pollos , Modelos Animales de Enfermedad , Escherichia coli/clasificación , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Fibroblastos/microbiología , Eliminación de Gen , Genotipo , Cloruro de Sodio/metabolismo , Virulencia
10.
BMC Vet Res ; 12(1): 167, 2016 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-27531140

RESUMEN

BACKGROUND: Avian pathogenic Escherichia coli (APEC) causes avian colibacillosis, which results in economic and welfare costs in the poultry industry worldwide. The pathogenesis of avian pathogenic E. coli strains is not well defined. Here, the function of an outer membrane protein encoded by the ireA gene of avian pathogenic E. coli strain DE205B was investigated. RESULTS: The ireA gene was distributed in 32.9 % (46/140) of tested E. coli strains, with high percentages in the phylogenetic ECOR groups B2 (58.8 %, 10/17) and D (55.9 %, 19/34). The gene expression level of ireA of APEC strain DE205B in high Fe M9 media was 1.8 times higher (P < 0.05) than that in low Fe M9 media. An ireA deletion mutant and complementary strain were constructed. Compared with the wild-type strain DE205B, the expression of most ferric uptake genes in the ireA deletion mutant were significantly upregulated (P < 0.05). The adhesion ability of the ireA deletion mutant to DF-1 cells was significantly decreased. The survival rate of ireA deletion mutant was reduced 21.17 % (P < 0.01), 25.42 (P < 0.05) and 70.0 % (P < 0.01) under alkali, high osmolarity, and low temperature (4 °C) conditions, respectively, compared with the wild-type strain. CONCLUSIONS: The results suggested that the protein encoded by the iron-regulated gene ireA has roles in adhesion and stress resistance in avian pathogenic E. coli.


Asunto(s)
Adhesión Bacteriana/fisiología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/clasificación , Regulación Bacteriana de la Expresión Génica/fisiología , Enfermedades de las Aves de Corral/microbiología , Estrés Fisiológico/fisiología , Animales , Adhesión Bacteriana/genética , Patos , Escherichia coli/genética , Escherichia coli/fisiología , Proteínas de Escherichia coli/genética , Mutación , Enfermedades de las Aves de Corral/patología , Estrés Fisiológico/genética
11.
Nat Commun ; 15(1): 4764, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834561

RESUMEN

Bacteriophage are sophisticated cellular parasites that can not only parasitize bacteria but are increasingly recognized for their direct interactions with mammalian hosts. Phage adherence to mucus is known to mediate enhanced antimicrobial effects in vitro. However, little is known about the therapeutic efficacy of mucus-adherent phages in vivo. Here, using a combination of in vitro gastrointestinal cell lines, a gut-on-a-chip microfluidic model, and an in vivo murine gut model, we demonstrated that a E. coli phage, øPNJ-6, provided enhanced gastrointestinal persistence and antimicrobial effects. øPNJ-6 bound fucose residues, of the gut secreted glycoprotein MUC2, through domain 1 of its Hoc protein, which led to increased intestinal mucus production that was suggestive of a positive feedback loop mediated by the mucus-adherent phage. These findings extend the Bacteriophage Adherence to Mucus model into phage therapy, demonstrating that øPNJ-6 displays enhanced persistence within the murine gut, leading to targeted depletion of intestinal pathogenic bacteria.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Mucosa Intestinal , Mucina 2 , Animales , Escherichia coli/virología , Ratones , Mucosa Intestinal/microbiología , Mucosa Intestinal/virología , Mucina 2/metabolismo , Humanos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/terapia , Terapia de Fagos/métodos , Adhesión Bacteriana , Femenino , Moco/metabolismo , Moco/virología , Colifagos/fisiología , Fucosa/metabolismo , Ratones Endogámicos C57BL
12.
Infect Immun ; 81(7): 2437-47, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23630958

RESUMEN

Autotransporter (AT) proteins constitute a large family of extracellular proteins that contribute to bacterial virulence. A novel AT adhesin gene, aatB, was identified in avian pathogenic Escherichia coli (APEC) DE205B via genomic analyses. The open reading frame of aatB was 1,017 bp, encoding a putative 36.3-kDa protein which contained structural motifs characteristic for AT proteins: a signal peptide, a passenger domain, and a translocator domain. The predicted three-dimensional structure of AatB consisted of two distinct domains, the C-terminal ß-barrel translocator domain and an N-terminal passenger domain. The prevalence analyses of aatB in APEC indicated that aatB was detected in 26.4% (72/273) of APEC strains and was strongly associated with phylogenetic groups D and B2. Quantitative real-time reverse transcription-PCR analyses revealed that AatB expression was increased during infection in vitro and in vivo. Moreover, AatB could elicit antibodies in infected ducks, suggesting that AatB is involved in APEC pathogenicity. Thus, APEC DE205B strains with a mutated aatB gene and mutated strains complemented with the aatB gene were constructed. Inactivation of aatB resulted in a reduced capacity to adhere to DF-1 cells, defective virulence capacity in vivo, and decreased colonization capacity in lung during systemic infection compared with the capacities of the wild-type strain. Furthermore, these capacities were restored in the complementation strains. These results indicated that AatB makes a significant contribution to APEC virulence through bacterial adherence to host tissues in vivo and in vitro. In addition, biofilm formation assays with strain AAEC189 expressing AatB indicated that AatB mediates biofilm formation.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Regulación Bacteriana de la Expresión Génica , Factores de Virulencia/metabolismo , Adhesinas de Escherichia coli/genética , Animales , Adhesión Bacteriana , Carga Bacteriana , Biopelículas , Patos/microbiología , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/veterinaria , Genes Bacterianos , Prueba de Complementación Genética , Pulmón/microbiología , Sistemas de Lectura Abierta , Filogenia , Estructura Terciaria de Proteína , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de Proteína , Eliminación de Secuencia , Factores de Virulencia/genética
13.
Res Vet Sci ; 150: 72-78, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-35809415

RESUMEN

Shiga toxin-producing Escherichia coli (STEC) is a group of pathogen that can cause various diseases in both humans and animals, such as watery diarrhea, hemorrhagic colitis, and uremia syndrome. Due to the serious situation of antibiotic resistance, phage therapy is considered to have a great potential in combating bacterial diseases. In this study, three phages (NJ-10, NJ-20, and NJ-38) with strong abilities to lyse virulent STEC strain CVCC193 cells in vitro were isolated. Subsequently, the therapeutic effects of the three phages were investigated in mice infected with CVCC193 cells. The results showed that the survival rates of mice injected with the phages at 3 h after challenge with CVCC193 cells were 40%-50%, while the survival rates of mice injected with the phages at 24 h before challenge were 80%-100%, indicating that pre-treatment with phages had better therapeutic effects than post-treatment. Pathological changes, bacterial loads in different organs, and serum levels of inflammatory factors of the infected mice were also detected. The results showed that the mice injected with the phages at 3 h after or 24 h before challenge with CVCC193 cells had significantly decreased organ lesions, bacterial loads, and serum levels of inflammatory factors as compared to infected mice without phage treatment. These results suggested that phages NJ-10, NJ-20, and NJ-38 can potentially protect against STEC infections.


Asunto(s)
Bacteriófagos , Infecciones por Escherichia coli , Enfermedades de los Roedores , Escherichia coli Shiga-Toxigénica , Animales , Carga Bacteriana/veterinaria , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/veterinaria , Humanos , Ratones
14.
Braz J Microbiol ; 53(1): 289-301, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34652743

RESUMEN

Vibrio parahaemolyticus is an important foodborne pathogenic bacterium that harbors the type III secretion system 1 (T3SS1) as an essential virulence factor. However, the pathogenesis and infection mechanism mediated by T3SS1 are not entirely clarified. Similar to previous studies on other T3SS-positive bacteria, the T3SS1 needle is a major extracellular component in V. parahaemolyticus. We recently showed that the needle gene-deletion mutant (ΔvscF) exhibited markedly decreased cytotoxicity and effector translocation during interaction with HeLa cells. To further elucidate the pathogenesis of T3SS1 during host cell infection, bacterial RNA was extracted from wild-type POR-1 and ΔvscF mutants under infected condition for comparative RNA sequencing analysis in HeLa cell. The results showed that 120 differentially expressed genes (DEGs) were identified in the ΔvscF-infected group. These encoded proteins of DEGs, such as VP2088, VP2089, and VP2091, were annotated as ABC transporter system, whereas VP0757, VP1123, and VP1289 may be new transcriptional regulators. In addition, the downregulation of T3SS1 had a positive influence on the expression of T3SS2. Moreover, the transcription of the basal body is unaffected by the needle, and there was a close relation among the tip, translocon, and needle, because bacterial adenylate cyclase two-hybrid system (BACTH system) assay indicated the interaction of VP1656, VP1670, VP1693, and VP1694 (VscF). This study provides insights into transcription mechanism of T3SS1 upon infecting HeLa cell, which is expected to better clarify the T3SS1 virulent mechanism.


Asunto(s)
Vibriosis , Vibrio parahaemolyticus , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células HeLa , Humanos , Transcriptoma , Vibriosis/microbiología , Vibriosis/patología , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo
15.
Life (Basel) ; 12(11)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36362895

RESUMEN

BACKGROUND: With the discovery of more and more drug-resistant bacterial strains, there is an urgent need for safer and more effective alternative treatments. In this study, antibacterial peptides and probiotic microcapsules were combined to treat gastrointestinal inflammation caused by Vibrio parahaemolyticus infection. METHODS: To improve the stability of probiotics in the gastrointestinal tract, two types of mixed natural anionic polysaccharides and chitosan were used as carriers to embed the probiotics. Taking Lacticaseibacillus casei CGMCC1.8727 microcapsules with good performance as the research object, the in vitro characteristics of the microcapsules were studied via acid resistance test and intestinal release test. The microcapsules were then tested for in vivo treatment in combination with the antibacterial peptide, bomidin, and the therapeutic effects were compared among microencapsulated probiotics, free probiotics, and probiotics in combination with bomidin. RESULTS: Microencapsulation was successfully manufactured under suitable processing parameters, with the product particle size being 2.04 ± 0.2743 mm. Compared with free probiotics, microencapsulation significantly improved the activity and preservation stability of the probiotics under simulated gastrointestinal conditions. Microencapsulated probiotics showed better therapeutic effects than free probiotics in vivo. Microcapsules combined with antimicrobial peptides accelerated the elimination of bacteria in vivo. This study provides a reference for anti-inflammatory treatment, especially for the treatment of gastrointestinal diseases.

16.
Front Microbiol ; 13: 1049341, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36452923

RESUMEN

Prophage sequences are present in most bacterial genomes and account for up to 20% of its host genome. Integration of temperate phages may have an impact on the expression of host genes, while some prophages could turn into the lytic cycle and affect bacterial host biological characteristics. We investigated the role of spontaneous induction prophages in avian pathogenic Escherichia coli (APEC), which is the causative agent of avian colibacillosis in poultry, and considered a potential zoonotic bacterium related to the fact it serves as an armory of extraintestinal pathogenic E. coli. We found that APEC strain DE458 had a high spontaneous induction rate in vivo and in vitro. The released phage particles, phi458, were isolated, purified, and sequenced, and the deletion mutant, DE458Δphi458, was constructed and characterized. Biofilm formation of DE458Δphi458 was strongly decreased compared to that of the wild-type strain (p < 0.01). In addition, while the addition of DNase (100 µg/ml) did not affect prophage release but could digest eDNA, it significantly reduced the biofilm production of DE458 biofilm to a level close to that of DE458Δphi458. Compared to DE458, the adhesion and invasion abilities of DE458Δphi458 increased by approximately 6-20 times (p < 0.05). The virulence of DE458Δphi458 was enhanced by approximately 10-fold in chickens based on a 50% lethal dose. Furthermore, avian infection assays showed that the bacterial loads of DE458Δphi458 in the lung and liver were increased by 16.5- and 10-fold (p < 0.05), respectively, compared with those of the WT strain. The qRT-PCR revealed that deletion of phi458 led to upregulation of type I fimbriate-related gene fimH and curli-related gene csgC by 3- and 2.8-fold, respectively (p < 0.01). Our study revealed that phi458 promoted biofilm formation by spontaneously inducing and decreasing virulence by repressing virulence genes.

17.
Pathogens ; 11(12)2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36558824

RESUMEN

Extraintestinal pathogenic Escherichia coli (ExPEC) is a pathogen that causes host extraintestinal diseases. The ST95 E. coli lineage is one of the dominant ExPEC lineages in humans and poultry. In this study, we took advantage of extensive E. coli genomes available through public open-access databases to construct a detailed understanding of the phylogeny and evolution of ST95. We used a high variability of accessory genomes to highlight the diversity and dynamic traits of ST95. Isolates from diverse hosts and geographic sources were randomly located on the phylogenetic tree, which suggested that there is no host specificity for ST95. The time-scaled phylogeny showed that ST95 is an ancient and long-lasting lineage. The virulence genes, resistance genes, and pathogenicity islands (PAIs) were characterized in ST95 pan-genomes to provide novel insights into the pathogenicity and multidrug resistance (MDR) genotypes. We found that a pool of large plasmids drives virulence and MDR. Based on the unique genes in the ST95 pan-genome, we designed a novel multiplex PCR reaction to rapidly detect ST95. Overall, our study addressed a gap in the current understanding of ST95 ExPEC genomes, with significant implications for recognizing the success and spread of ST95.

18.
Life (Basel) ; 12(9)2022 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-36143390

RESUMEN

Avian pathogenic Escherichia coli (APEC) is an important extra-intestinal pathogenic E. coli (ExPEC), which often causes systemic infection in poultry and causes great economic loss to the breeding industry. In addition, as a major source of human ExPEC infection, the potential zoonotic risk of APEC has been an ongoing concern. Previous studies have pointed out that APEC is a potential zoonotic pathogen, which has high homology with human pathogenic E. coli such as uro-pathogenic E. coli (UPEC) and neonatal meningitis E. coli (NMEC), shares multiple virulence factors and can cause mammalian diseases. Previous studies have reported that O18 and O78 could cause different degrees of meningitis in neonatal rats, and different serotypes had different degrees of zoonotic risk. Here, we compared APEC DE205B (O2:K1) with NMEC RS218 (O18:K1:H7) by phylogenetic analysis and virulence gene identification to analyze the potential risk of DE205B in zoonotic diseases. We found that DE205B possessed a variety of virulence factors associated with meningitis and, through phylogenetic analysis, had high homology with RS218. DE205B could colonize the cerebrospinal fluid (CSF) of rats, and cause meningitis and nerve damage. Symptoms and pathological changes in the brain were similar to RS218. In addition, we found that DE205B had a complete T6SS, of which Hcp protein was its important structural protein. Hcp1 induced cytoskeleton rearrangement in human brain microvascular endothelial cells (HBMECs), and Hcp2 was mainly involved in the invasion of DE205B in vitro. In the meningitis model of rats, deletion of hcp2 gene reduced survival in the blood and the brain invasiveness of DE205B. Compared with WT group, Δhcp2 group induced lower inflammation and neutrophils infiltration in brain tissue, alleviating the process of meningitis. Together, these results suggested that APEC DE205B had close genetic similarities to NMEC RS218, and a similar mechanism in causing meningitis and being a risk for zoonosis. This APEC serotype provided a basis for zoonotic research.

19.
Microbiol Spectr ; 10(3): e0023222, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35536028

RESUMEN

O157:H7 is the most important Shiga toxin-producing Escherichia coli (STEC) serotype in relation to public health. Given that antibiotics may contribute to the exacerbation of STEC-related disease and an increased frequency of antibiotic-resistant strains, bacteriophage (phage) therapy is considered a promising alternative. However, phage therapy targeting enteric pathogens is still underdeveloped with many confounding effects from the microbiota. Here we comprehensively compared the therapeutic efficacy of a phage cocktail with the antibiotic enrofloxacin in a mouse model of STEC O157:H7 EDL933 infection. Enrofloxacin treatment provided 100% survival and the phage cocktail treatment provided 90% survival. However, in terms of mouse recovery, the phage cocktail outperformed enrofloxacin in all measured outcomes. Compared with enrofloxacin treatment, phage treatment led to a faster elimination of enteric pathogens, decreased expression levels of inflammatory markers, increased weight gain, maintenance of a stable relative organ weight, and improved homeostasis of the gut microbiota. These results provide support for the potential of phage therapy to combat enteric pathogens and suggest that phage treatment leads to enhanced recovery of infected mice compared with antibiotics. IMPORTANCE With the increasing severity of antibiotic resistance and other adverse consequences, animal experiments and clinical trials investigating the use of phages for the control and prevention of enteric bacterial infections are growing. However, the effects of phages and antibiotics on organisms when treating intestinal infections have not been precisely studied. Here, we comprehensively compared the therapeutic efficacy of a phage cocktail to the antibiotic enrofloxacin in a mouse model of STEC O157:H7 EDL933 infection. We found that, despite a slightly lower protection rate, phage treatment contributed to a faster recovery of infected mice compared with enrofloxacin. These results highlight the potential benefits of phage therapy to combat enteric infections.


Asunto(s)
Bacteriófagos , Infecciones por Escherichia coli , Escherichia coli O157 , Escherichia coli Shiga-Toxigénica , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Modelos Animales de Enfermedad , Enrofloxacina/farmacología , Enrofloxacina/uso terapéutico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/terapia , Ratones
20.
Probiotics Antimicrob Proteins ; 14(1): 169-179, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34642879

RESUMEN

Macrobrachium rosenbergii is an economically important source of crustacean seafood worldwide. Vibrio parahaemolyticus is an important aquatic pathogen that causes epidemics of acute hepatopancreatic necrosis in shrimp populations, which results in significant economic losses to aquaculture farmers. To prevent the antibiotics abuse, which has become a serious threat to human health, novel anti-infective strategies are urgently required to control V. parahaemolyticus. Antimicrobial peptides, which exhibit favourable germicidal activity compared to traditional antibiotics, can be used as a key method to prevent and treat bacterial diseases. Herein, an antimicrobial peptide, bomidin, was expressed through genetic engineering technology. The minimum inhibitory concentration (MIC) of bomidin showed a significant inhibitory effect on V. parahaemolyticus that was equivalent to that of ampicillin. Subsequently, the mechanism of action of recombinant bomidin was explored using PNP and ONPG assays to investigate the effects on membrane permeability. These assays indicated that bomidin penetrated the germ membrane and induced the release of cytoplasmic contents and ultimately interacted with DNA to form a bomidin-DNA complex that inhibits bacterial survival. Transmission electron microscopy and scanning electron microscopy revealed that bomidin could cause damage and dysfunction to the cell wall and membrane. Bomidin was nontoxic to mouse red blood cells within a concentration range that was much larger than the MIC. Toxicity assays revealed that 0.02 mg/mL bomidin was safe for use with juvenile freshwater prawns of M. rosenbergii and significantly inhibited the growth of V. parahaemolyticus in cultured water. These results demonstrated that synthetic peptide bomidin had great antibacterial effect against V. parahaemolyticus and therefore a therapeutic potential in aquaculture.


Asunto(s)
Palaemonidae , Vibrio parahaemolyticus , Animales , Péptidos Antimicrobianos , Acuicultura , Ratones , Pruebas de Sensibilidad Microbiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA