Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Anim Sci ; 1022024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38563410

RESUMO

We conducted two experiments to evaluate the effects of a novel bacterial-based direct-fed microbial (DFM) on intestinal barrier integrity using the in vitro transepithelial electrical resistance (TEER) assay. In experiment 1, human-derived Caco-2 cells received or not (CON) a DFM containing Ligilactobacillus (formerly Lactobacillus) animalis 506, Propionibacterium freudenreichii 507, Bacillus paralicheniformis 809, and B. subtilis 597 (BDP; BOVAMINE DEFEND® Plus) at a rate of 1 × 108 CFU/transwell. Concurrently with treatment application (CON or BDP), a pathogenic challenge of Clostridium perfringens type A was added alone (PAT) or with BDP (PAT + BDP) at a rate of 2.8 × 107 CFU/transwell in a 2 × 2 factorial arrangement. In experiment 2, Caco-2 cells were also assigned in a 2 × 2 factorial design to CON or BDP and then, 2 h post-treatment administration (CON and BDP), a mixture of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) was added alone (CYT) or with BDP (CYT + BDP) at a 10:1 ratio, respectively. In both experiments, TEER was measured for 18 h. In experiment 1, a DFM × pathogen × hour interaction was observed for TEER (P < 0.0001). Adding the PAT alone initially tended to increase TEER vs. CON from 1.1 to 2.2 h (P ≤ 0.09), increased TEER at 3.2 h (P < 0.01), but reduced TEER from 5.4 to the end of the experimental period at 18.4 h (P ≤ 0.01). On the other hand, adding DFM, with or without the pathogenic challenge, yielded greater TEER vs. CON-CON and CON-PAT for most of the experimental period (P ≤ 0.04). A similar interaction was detected and reported in experiment 2 (P < 0.0001). The CYT challenge reduced mean TEER compared with all other treatments from 3.2 h to the remainder of the study (P ≤ 0.03). On the other hand, BDP-CYT was able to maintain the integrity of the epithelial cells when compared with CON-CON throughout the experimental period (P ≤ 0.03), the exception being at 3.2 h (P = 0.20). Moreover, BDP-CON increased (P ≤ 0.04) TEER when compared with CON-CON from 3.2 to 18.4 h, but also in comparison with BDP-CYT from 4.3 to 18.4 h post-DFM and challenge administration into the cells. In summary, C. perfringens type A and a pro-inflammatory cytokine cocktail compromised the integrity of intestinal epithelial cell monolayers in vitro, whereas adding a multispecies bacteria-based DFM counteracted these damaging effects.


Two experiments were designed to evaluate the effects of adding a bacterial-based direct-fed microbial (DFM) containing Lactobacillus animalis 506, Propionibacterium freudenreichii 507, Bacillus paralicheniformis 809, and Bacillus subtilis 597 on the integrity of intestinal epithelial cells challenged with Clostridium perfringens type A or a pro-inflammatory cytokine cocktail. Regardless of the challenge, the addition of the DFM maintained the integrity of the intestinal epithelial cells in vitro. These results help to elucidate the potential beneficial effects that the bacterial-based DFM containing L. animalis 506, P. freudenreichii 507, B. paralicheniformis 809, and B. subtilis 597 may bring to livestock species.


Assuntos
Citocinas , Dieta , Humanos , Animais , Células CACO-2 , Lactobacillus , Clostridium perfringens , Ração Animal/análise
2.
Transl Anim Sci ; 8: txae061, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38685987

RESUMO

We designed and conducted two in vitro experiments to evaluate the effects of two Bacillus spp. probiotics on gut barrier integrity using the transepithelial electrical resistance (TEER) assay under two different challenge models. In Exp. 1, intestinal epithelial cells received or not (CON) B. paralicheniformis 809 (BLI) or B. subtilis 810 (BSU) at a rate of 1 × 108 colony forming units (CFU)/transwell. Two hours after treatment application (CON, BLI, or BSU), 5 mM of the reactive oxygen species hydrogen peroxide, mimicking mucosal oxidative stress, was added alone (HYP) or with each of the Bacillus spp. (HYP + BLI or HYP + BSU). In Exp. 2, cells were assigned to the same treatments as in Exp. 1 (CON, BLI, and BSU), or mycotoxin deoxynivalenol (DON), which was added alone or in combination with BLI or BSU, resulting in another two treatments (DON + BLI and DON + BSU). Transepithelial electrical resistance was measured for 14 h postchallenge. In Exp. 1, a treatment × hour interaction was observed for TEER (P < 0.0001). Adding BLI and BSU resulted in greater TEER values vs. CON for most of the experimental period (P < 0.02), whereas HYP reduced mean TEER and area under the curve (AUC), while increasing the amount of sugar that translocated through the monolayer cells (P < 0.001). A treatment × hour interaction was also observed in Exp. 2 (P < 0.0001), as DON led to an immediate and acute drop in TEER that lasted until the end of the experimental period (P < 0.0001). Both BLI and BSU alleviated the DON-induced damaging effects on the integrity of intestinal epithelial cells, whereas both Bacillus spp. alleviated the damage caused by DON alone and the proportion of sugar that translocated through the monolayer cells was not different between CON and DON + BLI (P = 0.14) and DON + BLI and DON + BSU (P = 0.62). In summary, both Bacillus spp. strains (B. paralicheniformis 809 and B. subtilis 810) were able to counteract the damaging effects of the challenge agents, hydrogen peroxide and deoxynivalenol, on gut barrier integrity.

3.
JDS Commun ; 4(4): 284-287, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37521053

RESUMO

Optimization and support of health and performance of preweaning dairy calves is paramount to any dairy operation, and natural solutions, such as probiotics, may help to achieve such a goal. Two experiments were designed to evaluate the effects of direct-fed microbial (DFM) Enterococcus faecium 669 on performance of preweaning dairy calves. In experiment 1, twenty 4-d-old Holstein calves [initial body weight (BW) 41 ± 2.1 kg] were randomly assigned to either (1) no probiotic supplementation (CON; n = 10) or (2) supplementation with probiotic strain E. faecium 669 during the preweaning period (DFM; n = 10) at 2.0 × 1010 cfu/kg of whole milk. Full individual BW was analyzed every 20 d for average daily gain (ADG) and feed efficiency (FE) determination. In experiment 2, thirty 4-d-old Holstein calves (initial BW 40 ± 1.9 kg) were assigned to the same treatments as in experiment 1 (CON and DFM). The DFM supplementation period was divided into period I (from d 0 to 21) and II (from d 22 to 63), with weaning occurring when animals were 67 d of age. During the entire experimental period, DFM was mixed into the whole milk at a rate of 1.5 × 1010 and 2.5 × 109 cfu/kg of whole milk/calf per day for periods I and II, respectively (6-time reduction). Full individual BW was taken every 21 d. As a routine of the experiment, calves were monitored daily, and diarrhea cases were evaluated using a daily 3-point fecal score. For both experiments, all data were analyzed using calf as the experimental unit. In experiment 1, DFM-supplemented calves were heavier on d 40 (+ 4.5 kg) and 60 (+ 6.5 kg) and had a greater ADG (+ 118 g) versus CON. In experiment 2, supplementation with DFM significantly tended to reduce diarrhea occurrence. Treatment × day and treatment × week interactions were observed for BW, ADG, and gain-to-feed ratio. Dairy calves supplemented with DFM were 1.8 and 3.5 kg heavier on d 42 and at weaning, respectively, and had a greater ADG from d 21 to 42 (+ 52 g) and 42 to 63 (+ 77 g) and gain-to-feed ratio from d 42 to 63 (+ 8.6%). In summary, supplementation of E. faecium 669 to dairy calves improved preweaning performance, even when the dose of the DFM was reduced by 6- to 8-times. Additionally, initial promising results were observed on diarrhea occurrence, but further studies are warranted.

4.
mBio ; 11(3)2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32430467

RESUMO

Escherichia coli sequence type 131 (ST131) is a major cause of urinary and bloodstream infections. Its association with extended-spectrum ß-lactamases (ESBLs) significantly complicates treatment. Its best-described component is the rapidly expanding H30Rx clade, containing allele 30 of the type 1 fimbrial adhesin gene fimH This lineage appears to have emerged in the United States and spread around the world in part due to the acquisition of the ESBL-encoding blaCTX-M-15 gene and resistance to fluoroquinolones. However, non-H30 ST131 sublineages with other acquired CTX-M-type resistance genes are also emerging. Based on whole-genome analyses, we describe here the presence of an (fimH) H27 E. coli ST131 sublineage that has recently caused an outbreak of community-acquired bacteremia and recurrent urinary tract infections (UTIs) in Denmark. This sublineage has acquired both a virulence plasmid (pAA) that defines the enteroaggregative E. coli (EAEC) diarrheagenic pathotype and multiple genes associated with extraintestinal E. coli (ExPEC); combined, these traits have made this particular ST131 sublineage successful at colonizing its human host and causing recurrent UTI. Moreover, using a historic World Health Organization (WHO) E. coli collection and publicly available genome sequences, we identified a global H27 EAEC ST131 sublineage that dates back as far as 1998. Most H27 EAEC ST131 isolates harbor pAA or pAA-like plasmids, and our analysis strongly implies a single ancestral acquisition among these isolates. These findings illustrate both the profound plasticity of this important pathogenic E. coli ST131 H27 sublineage and genetic acquisitions of EAEC-specific virulence traits that likely confer an enhanced ability to cause intestinal colonization.IMPORTANCEE. coli ST131 is an important extraintestinal pathogenic lineage. A signature characteristic of ST131 is its ability to asymptomatically colonize the gastrointestinal tract and then opportunistically cause extraintestinal infections, such as cystitis, pyelonephritis, and urosepsis. In this study, we identified an ST131 H27 sublineage that has acquired the enteroaggregative diarrheagenic phenotype, spread across multiple continents, and caused multiple outbreaks of community-acquired ESBL-associated bloodstream infections in Denmark. The strain's ability to both cause diarrhea and innocuously colonize the human gastrointestinal tract may facilitate its dissemination and establishment in the community.


Assuntos
Bacteriemia/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Infecções Urinárias/microbiologia , Antibacterianos/farmacologia , Bancos de Espécimes Biológicos , Infecções Comunitárias Adquiridas/microbiologia , Dinamarca , Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/efeitos dos fármacos , Genoma Bacteriano , Humanos , Tipagem de Sequências Multilocus , Filogenia , Plasmídeos/genética , Análise de Sequência de DNA , Virulência/genética , Sequenciamento Completo do Genoma , Organização Mundial da Saúde
5.
mBio ; 8(3)2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28588132

RESUMO

Enteroaggregative Escherichia coli (EAEC) causes diarrhea and intestinal inflammation worldwide. EAEC strains are characterized by the presence of aggregative adherence fimbriae (AAF), which play a key role in pathogenesis by mediating attachment to the intestinal mucosa and by triggering host inflammatory responses. Here, we identify the epithelial transmembrane mucin MUC1 as an intestinal host cell receptor for EAEC, demonstrating that AAF-mediated interactions between EAEC and MUC1 facilitate enhanced bacterial adhesion. We further demonstrate that EAEC infection also causes elevated expression of MUC1 in inflamed human intestinal tissues. Moreover, we find that MUC1 facilitates AAF-dependent migration of neutrophils across the epithelium in response to EAEC infection. Thus, we show for the first time a proinflammatory role for MUC1 in the host response to an intestinal pathogen.IMPORTANCE EAEC is a clinically important intestinal pathogen that triggers intestinal inflammation and diarrheal illness via mechanisms that are not yet fully understood. Our findings provide new insight into how EAEC triggers host inflammation and underscores the pivotal role of AAFs-the principal adhesins of EAEC-in driving EAEC-associated disease. Most importantly, our findings add a new dimension to the signaling properties of the transmembrane mucin MUC1. Mostly studied for its role in various forms of cancer, MUC1 is widely regarded as playing an anti-inflammatory role in response to infection with bacterial pathogens in various tissues. However, the role of MUC1 during intestinal infections has not been previously explored, and our results describe the first report of MUC1 as a proinflammatory factor following intestinal infection.


Assuntos
Aderência Bacteriana , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Fímbrias Bacterianas/imunologia , Mucina-1/metabolismo , Infiltração de Neutrófilos , Movimento Celular , Diarreia/microbiologia , Escherichia coli/imunologia , Escherichia coli/patogenicidade , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/fisiologia , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/fisiopatologia , Mucina-1/genética , Neutrófilos/fisiologia , Transdução de Sinais/imunologia
6.
Front Microbiol ; 8: 579, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28439262

RESUMO

Heat treatment is a widely used process to reduce bacterial loads in the food industry or to decontaminate surfaces, e.g., in hospital settings. However, there are situations where lower temperatures must be employed, for instance in case of food production such as raw milk cheese or for decontamination of medical devices such as thermo-labile flexible endoscopes. A recently identified locus of heat resistance (LHR) has been shown to be present in and confer heat resistance to a variety of Enterobacteriaceae, including Escherichia coli isolates from food production settings and clinical ESBL-producing E. coli isolates. Here, we describe the presence of two distinct LHR variants within a particularly heat resistant E. coli raw milk cheese isolate. We demonstrate for the first time in this species the presence of one of these LHRs on a plasmid, designated pFAM21805, also encoding type 3 fimbriae and three bacteriocins and corresponding self-immunity proteins. The plasmid was highly transferable to other E. coli strains, including Shiga-toxin-producing strains, and conferred LHR-dependent heat resistance as well as type 3 fimbriae-dependent biofilm formation capabilities. Selection for and acquisition of this "survival" plasmid by pathogenic organisms, e.g., in food production environments, may pose great concern and emphasizes the need to screen for the presence of LHR genes in isolates.

7.
Front Microbiol ; 8: 263, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28275371

RESUMO

Enteroaggregative Escherichia coli (EAEC) is an increasingly recognized pathogen associated with acute and persistent diarrhea worldwide. While EAEC strains are considered highly heterogeneous, aggregative adherence fimbriae (AAFs) are thought to play a pivotal role in pathogenicity by facilitating adherence to the intestinal mucosa. In this study, we optimized an existing multiplex PCR to target all known AAF variants, which are distinguished by differences in their pilin subunits. We applied the assay on a collection of 162 clinical Danish EAEC strains and interestingly found six, by SNP analysis phylogenetically distinct, strains harboring the major pilin subunits from both AAF/III and AAF/V. Whole-genome and plasmid sequencing revealed that in these six strains the agg3A and agg5A genes were located on a novel pAA plasmid variant. Moreover, the plasmid also encoded several other virulence genes including some not previously found on pAA plasmids. Thus, this plasmid endows the host strains with a remarkably high number of EAEC associated virulence genes hereby likely promoting strain pathogenicity.

8.
Res Microbiol ; 167(5): 345-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26946311

RESUMO

Nosocomial infections caused by extended-spectrum ß-lactamase (ESBL)-producing Escherichia coli are a major concern worldwide. There is an urgent need to identify bacterial factors promoting survival and persistence of these organisms in the nosocomial environment. Here, we describe the presence of a gene cluster, containing the Clp ATPase ClpK, within a collection of Danish ESBL-producing E. coli isolates. The cluster conferred thermoprotection upon the isolates, and thus might facilitate survival on medical devices exposed to semi-high temperatures in a hospital setting.


Assuntos
Infecção Hospitalar/microbiologia , Microbiologia Ambiental , Escherichia coli/fisiologia , Escherichia coli/efeitos da radiação , Viabilidade Microbiana/efeitos da radiação , beta-Lactamases/metabolismo , Dinamarca , Endopeptidase Clp/genética , Endopeptidase Clp/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Hospitais , Temperatura Alta , Família Multigênica
9.
Cell Microbiol ; 17(6): 843-59, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25486861

RESUMO

Salmonella enterica Typhimurium induces intestinal inflammation through the activity of type III secreted effector (T3SE) proteins. Our prior results indicate that the secretion of the T3SE SipA and the ability of SipA to induce epithelial cell responses that lead to induction of polymorphonuclear transepithelial migration are not coupled to its direct delivery into epithelial cells from Salmonella. We therefore tested the hypothesis that SipA interacts with a membrane protein located at the apical surface of intestinal epithelial cells. Employing a split ubiquitin yeast-two-hybrid screen, we identified the tetraspanning membrane protein, p53 effector related to PMP-22 (PERP), as a SipA binding partner. SipA and PERP appear to have intersecting activities as we found PERP to be involved in proinflammatory pathways shown to be regulated by SipA. In sum, our studies reveal a critical role for PERP in the pathogenesis of S. Typhimurium, and for the first time demonstrate that SipA, a T3SE protein, can engage a host protein at the epithelial surface.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Inflamação/microbiologia , Inflamação/patologia , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Salmonella typhimurium/imunologia , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Genes Supressores de Tumor , Humanos , Ligação Proteica , Mapeamento de Interação de Proteínas , Migração Transendotelial e Transepitelial , Técnicas do Sistema de Duplo-Híbrido
10.
Cell Microbiol ; 16(9): 1339-53, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24617613

RESUMO

Neutrophil (polymorphonuclear leucocytes; PMN) transmigration across mucosal surfaces contributes to dysfunction of epithelial barrier properties, a characteristic underlying many mucosal inflammatory diseases. Using Salmonella enterica serovar Typhimurium (S. Typhimurium) as a prototypic proinflammatory insult, we have previously reported that the eicosanoid hepoxilin A3 (HXA3 ), an endogenous product of 12-lipoxygenase (12-LOX) activity, is secreted from the apical surface of the intestinal epithelium to establish a chemotactic gradient that guides PMN across the epithelial surface. Since little is known regarding the molecular mechanisms that regulate 12-LOX during S. Typhimurium infection, we investigated this pathway. We found that expression of phospholipid glutathione peroxidase (GPX4), which is known to have an inhibitory effect on 12-LOX activity, is significantly decreased at both the mRNA and protein level during infection with S. Typhimurium. Moreover, employing intestinal epithelial cell monolayers expressing siRNA against GPX4 mRNA, S. Typhimurium-induced PMN migration was significantly increased compared with the non-specific siRNA control cells. Conversely, in cells engineered to overexpress GPX4, S. Typhimurium-induced PMN migration was significantly decreased, which is consistent with the finding that partial depletion of GPX4 by RNAi resulted in a significant increase in HXA3 secretion during S. Typhimurium infection. Mechanistically, although we found Salmonella entry not to be required for the induced decrease in GPX4, the secreted effector, SipA, which is known to induce epithelial responses leading to stimulation of HXA3 , governed the decrease in GPX4 in a process that does not lead to an overall increase in the levels of ROS. Taken together, these results suggest that S. Typhimurium induces apical secretion of HXA3 by decreasing the expression of phospholipid GPX, which in turn leads to an increase in 12-LOX activity, and hence HXA3 synthesis.


Assuntos
Glutationa Peroxidase/metabolismo , Mucosa Intestinal/enzimologia , Neutrófilos/citologia , Neutrófilos/metabolismo , Salmonella typhimurium/fisiologia , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Mucosa Intestinal/citologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Espécies Reativas de Oxigênio/metabolismo , Migração Transendotelial e Transepitelial/genética , Migração Transendotelial e Transepitelial/fisiologia
11.
Infect Dis Clin North Am ; 27(3): 631-49, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24011834

RESUMO

Pathogenic Escherichia coli are genetically diverse and encompass a broad variety of pathotypes, such as enteroaggregative E. coli (EAEC) or enterohemorrhagic E. coli (EHEC), which cause distinct clinical syndromes. The historically large 2011 German outbreak of hemolytic uremic syndrome (HUS), caused by a Shiga-toxin producing E. coli (STEC) of the serotype O104:H4, illustrated the emerging importance of non-O157 STEC. STEC O104:H4, with features characteristic of both enteroaggregative E. coli and enterohemorrhagic E. coli, represents a unique and highly virulent pathotype. The German outbreak both allowed for the evaluation of several potential therapeutic approaches to STEC-induced HUS and emphasizes the importance of early and specific detection of both O157 and non-O157 STEC.


Assuntos
Doenças Transmissíveis Emergentes/microbiologia , Infecções por Escherichia coli/microbiologia , Escherichia coli Shiga Toxigênica/patogenicidade , Surtos de Doenças , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/genética , Europa (Continente)/epidemiologia , Genoma Bacteriano , Humanos , Escherichia coli Shiga Toxigênica/genética , Virulência/genética
12.
Infect Immun ; 81(4): 1164-71, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23357383

RESUMO

A multiresistant clonal Escherichia coli O78:H10 strain qualifying molecularly as enteroaggregative Escherichia coli (EAEC) was recently shown to be the cause of a community-acquired outbreak of urinary tract infection (UTI) in greater Copenhagen, Denmark, in 1991. This marks the first time EAEC has been associated with an extraintestinal disease outbreak. Importantly, the outbreak isolates were recovered from the urine of patients with symptomatic UTI, strongly implying urovirulence. Here, we sought to determine the uropathogenic properties of the Copenhagen outbreak strain and whether these properties are conferred by the EAEC-specific virulence factors. We demonstrated that through expression of aggregative adherence fimbriae, the principal adhesins of EAEC, the outbreak strain exhibited pronouncedly increased adherence to human bladder epithelial cells compared to prototype uropathogenic strains. Moreover, the strain was able to produce distinct biofilms on abiotic surfaces, including urethral catheters. These findings suggest that EAEC-specific virulence factors increase uropathogenicity and may have played a significant role in the ability of the strain to cause a community-acquired outbreak of UTI. Thus, inclusion of EAEC-specific virulence factors is warranted in future detection and characterization of uropathogenic E. coli.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/patogenicidade , Infecções Urinárias/microbiologia , Fatores de Virulência/metabolismo , Animais , Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Dinamarca/epidemiologia , Células Epiteliais/microbiologia , Escherichia coli/isolamento & purificação , Escherichia coli/fisiologia , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/metabolismo , Fímbrias Bacterianas/metabolismo , Humanos , Camundongos , Infecções Urinárias/epidemiologia , Infecções Urinárias/patologia
13.
Cell Adh Migr ; 6(5): 413-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23076052

RESUMO

Enteroaggregative Escherichia coli (EAEC) is an important cause of endemic and epidemic diarrheal disease worldwide. Although not classically considered an inflammatory pathogen in the style of Shigella and Salmonella species, clinical data from patients suggests that inflammatory responses may play an important role during EAEC disease. However, the specific role of inflammation during EAEC pathogenesis has not been investigated in detail. To better understand how EAEC may induce inflammation, we have focused our attention on the intimate interactions between EAEC and the host epithelium and the subsequent induction of host cell signaling events leading to innate immune responses. Here, we discuss our recent findings on the signaling pathway by which EAEC promotes transepithelial migration of polymorphonuclear leukocytes (PMNs), the role of aggregative adherence fimbriae in triggering this event and the implementation of human intestinal xenografts in immunodeficient mice for studying EAEC pathogenesis in vivo. Our findings suggest that EAEC shares conserved mechanisms of inducing PMN recruitment with other intestinal pathogens, providing new insight into the potential pathological consequences of EAEC-induced inflammation.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/patogenicidade , Interações Hospedeiro-Patógeno , Inflamação/microbiologia , Adesinas de Escherichia coli/imunologia , Adesinas de Escherichia coli/metabolismo , Araquidonato 12-Lipoxigenase/metabolismo , Ácido Araquidônico/metabolismo , Aderência Bacteriana , Membrana Celular/metabolismo , Movimento Celular , Escherichia coli/imunologia , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/imunologia , Humanos , Imunidade Inata , Inflamação/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Proteína Quinase C-delta/imunologia , Proteína Quinase C-delta/metabolismo , Transdução de Sinais , Migração Transendotelial e Transepitelial
14.
BMC Microbiol ; 12: 201, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22967317

RESUMO

BACKGROUND: Klebsiella pneumoniae is an important opportunistic pathogen causing pneumonia, sepsis and urinary tract infections. Colonisation of the gastrointestinal (GI) tract is a key step in the development of infections; yet the specific factors important for K. pneumoniae to colonize and reside in the GI tract of the host are largely unknown. To identify K. pneumoniae genes promoting GI colonisation, a novel genomic-library-based approach was employed. RESULTS: Screening of a K. pneumoniae C3091 genomic library, expressed in E. coli strain EPI100, in a mouse model of GI colonisation led to the positive selection of five clones containing genes promoting persistent colonisation of the mouse GI tract. These included genes encoding the global response regulator ArcA; GalET of the galactose operon; and a cluster of two putative membrane-associated proteins of unknown function. Both ArcA and GalET are known to be involved in metabolic pathways in Klebsiella but may have additional biological actions beneficial to the pathogen. In support of this, GalET was found to confer decreased bile salt sensitivity to EPI100. CONCLUSIONS: The present work establishes the use of genomic-library-based in vivo screening assays as a valuable tool for identification and characterization of virulence factors in K. pneumoniae and other bacterial pathogens.


Assuntos
Trato Gastrointestinal/microbiologia , Genética Microbiana/métodos , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/patogenicidade , Biologia Molecular/métodos , Seleção Genética , Fatores de Virulência/metabolismo , Animais , Portador Sadio/microbiologia , Modelos Animais de Doenças , Escherichia coli/genética , Feminino , Biblioteca Gênica , Testes Genéticos/métodos , Humanos , Klebsiella pneumoniae/genética , Camundongos , Fatores de Virulência/genética
15.
J Infect Dis ; 206(5): 714-22, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22723643

RESUMO

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) are increasingly recognized as an important agent of inflammatory and often persistent diarrhea. Although previous studies report on the inflammatory aspects of EAEC pathogenesis, the mechanisms by which EAEC trigger these events are not well understood. METHODS: EAEC strains harboring mutations in known EAEC virulence determinants were tested in an in vitro model of transepithelial migration of polymorphonuclear neutrophils (PMNs) and in human intestinal xenografts in severe-combined immunodeficient (SCID-HU-INT) mice, a novel model for studying EAEC disease in vivo. RESULTS: Expression of aggregative adherence fimbriae (AAFs), the principal adhesins of EAEC, was required for EAEC-induced PMN transepithelial migration in vitro. Moreover, constructed plasmids encoding AAF gene clusters demonstrated that the AAF adhesins are sufficient for triggering this event in a nonpathogenic E. coli background. Furthermore, with use of the SCID-HU-INT mouse model, severe tissue damage and infiltration of inflammatory cells was observed in the human tissue after EAEC infection. These pathological marks were strongly related to AAF expression, thus clearly confirming our in vitro findings. CONCLUSIONS: The present work establishes EAEC as an important inflammatory pathogen and the AAF adhesins as inducers of potentially detrimental immune responses.


Assuntos
Adesinas de Escherichia coli/imunologia , Diarreia/microbiologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Fímbrias Bacterianas/imunologia , Adesinas de Escherichia coli/genética , Animais , Aderência Bacteriana/imunologia , Movimento Celular/imunologia , Clonagem Molecular , Diarreia/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/microbiologia , Histocitoquímica , Humanos , Imunidade Inata/imunologia , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos SCID , Neutrófilos/imunologia , Transplante Heterólogo
16.
Cell Microbiol ; 14(1): 120-32, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21951973

RESUMO

Enteroaggregative Escherichia coli (EAEC) induces release of pro-inflammatory markers and disruption of intestinal epithelial barriers in vitro, suggesting an inflammatory aspect to EAEC infection. However, the mechanisms underlying EAEC-induced mucosal inflammatory responses and the extent to which these events contribute to pathogenesis is not well characterized. Employing an established in vitro model we demonstrated that EAEC prototype strain 042 induces migration of polymorphonuclear neutrophils (PMNs) across polarized T84 cell monolayers. This event was mediated through a conserved host cell signalling cascade involving the 12/15-LOX pathway and led to apical secretion of an arachidonic acid-derived lipid PMN chemoattractant, guiding PMNs across the epithelia to the site of infection. Moreover, supporting the hypothesis that inflammatory responses may contribute to EAEC pathogenesis, we found that PMN transepithelial migration promoted enhanced attachment of EAEC 042 to T84 cells. These findings suggest that EAEC-induced PMN infiltration may favour colonization and thus pathogenesis of EAEC.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Quimiotaxia de Leucócito , Escherichia coli/imunologia , Neutrófilos/metabolismo , Migração Transendotelial e Transepitelial/imunologia , Ácidos Araquidônicos , Aderência Bacteriana , Linhagem Celular Tumoral , Escherichia coli/patogenicidade , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Proteína Quinase C/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA