Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Microbiol ; 13: 980989, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36246229

RESUMO

Certain lactic acid bacteria (LAB) are associated with immune modulatory activities including down-regulation of pro-inflammatory gene transcription and expression. While host antigen-presenting cells (APCs) and intestinal epithelial cells (IEC) can interact directly with both pathogenic and commensal bacteria through innate immune pattern recognition receptors, recent evidence indicates indirect communication through secreted molecules is an important inter-domain communication mechanism. This communication route may be especially important in the context of IEC and APC interactions which shape host immune responses within the gut environment. We have previously shown that the Lacticaseibacillus rhamnosus R0011 secretome (LrS) dampens pro-inflammatory gene transcription and mediator production from Tumor Necrosis Factor-α and Salmonella enterica serovar Typhimurium secretome (STS)-challenged HT-29 IECs through the induction of negative regulators of innate immunity. However, many questions remain about interactions mediated through these bacterial-derived soluble components and the resulting host immune outcomes in the context of IEC and APC interactions. In the present study, we examined the ability of the LrS to down-regulate pro-inflammatory gene transcription and cytokine production from STS-challenged T84 human IEC and THP-1 human monocyte co-cultures. Cytokine and chemokine profiling revealed that apically delivered LrS induces apical secretion of macrophage inhibitory factor (MIF) and down-regulates STS-induced pro-inflammatory mediator secretion into the apical and basolateral chambers of the T84/THP-1 co-culture. Transcriptional profiling confirmed these results, as the LrS attenuated STS challenge-induced CXCL8 and NFκB1 expression in T84 IECs and THP-1 APCs. Interestingly, the LrS also reversed STS-induced damage to monolayer transepithelial resistance (TER) and permeability, results which were confirmed by ZO-1 gene expression and immunofluorescence visualization of ZO-1 expression in T84 IEC monolayers. The addition of a MIF-neutralizing antibody abrogated the ability of the LrS to reverse STS-induced damage to T84 IEC monolayer integrity, suggesting a novel role for MIF in maintaining IEC barrier function and integrity in response to soluble components derived from LAB. The results presented here provide mechanistic evidence for indirect communication mechanisms used by LAB to modulate immune responses to pathogen challenge, using in vitro approaches which allow for IEC and APC cell communication in a context which more closely mimics that which occurs in vivo.

2.
Sci Rep ; 12(1): 16517, 2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-36192578

RESUMO

Increasing evidence suggests that the intestinal microbiota composition could play a role in specific pathologies such as hypertension, obesity and diabetes. This study aims to demonstrate that the intestinal microbiota modulated by a diet creating dysbiosis increased the size of the myocardial infarction and that probiotics could attenuate this effect. To do this, microbiota transplants from rats fed a dysbiotic or non-dysbiotic diet in the presence or absence of probiotics were performed for 10 days on rats whose microbiota had been previously suppressed by antibiotic therapy. Then, the anterior coronary artery of the transplanted rats was occluded for 30 min. Infarct size was measured after 24 h of reperfusion, while signaling pathways were evaluated after 15 min of reperfusion. Intestinal resistance, plasma concentration of LPS (lipopolysaccharides), activation of NF-κB and Akt and composition of the microbiota were also measured. Our results demonstrate a larger infarct size in animals transplanted with the dysbiotic microbiota without probiotics compared to the other groups, including those that received the dysbiotic microbiota with probiotics. This increase in infarct size correlates with a higher firmicutes/bacteroidetes ratio, NF-kB phosphorylation and plasma LPS concentration, and a decrease in intestinal barrier resistance and Akt. These results indicate that dysbiotic microbiota promotes an increase in infarct size, an effect that probiotics can attenuate.


Assuntos
Microbiota , Infarto do Miocárdio , Probióticos , Animais , Antibacterianos , Disbiose , Lipopolissacarídeos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos
3.
Int J Mol Sci ; 22(11)2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-34073352

RESUMO

Helicobacter pylori is a prevalent bacterium that can cause gastric ulcers and cancers. Lactic acid bacteria (LAB) ameliorate treatment outcomes against H. pylori, suggesting that they could be a source of bioactive molecules usable as alternatives to current antibiotics for which resistance is mounting. We developed an in vitro framework to compare the anti-H. pylori properties of 25 LAB and their secretions against H. pylori. All studies were done at acidic and neutralized pH, with or without urea to mimic various gastric compartments. Eighteen LAB strains secreted molecules that curtailed the growth of H. pylori and the activity was urea-resistant in five LAB. Several LAB supernatants also reduced the urease activity of H. pylori. Pre-treatment of H. pylori with acidic LAB supernatants abrogated its flagella-mediated motility and decreased its ability to elicit pro-inflammatory IL-8 cytokine from human gastric cells, without reverting the H. pylori-induced repression of other pro-inflammatory cytokines. This study identified the LAB that have the most anti-H. pylori effects, decreasing its viability, its production of virulence factors, its motility and/or its ability to elicit pro-inflammatory IL-8 from gastric cells. Once identified, these molecules can be used as alternatives or complements to current antibiotics to fight H. pylori infections.


Assuntos
Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter , Helicobacter pylori/crescimento & desenvolvimento , Interleucina-8/metabolismo , Lactobacillales , Antibacterianos , Linhagem Celular , Mucosa Gástrica/patologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/terapia , Humanos , Concentração de Íons de Hidrogênio
4.
Front Microbiol ; 12: 698638, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35154018

RESUMO

Clostridioides difficile infection (CDI) is frequently associated with intestinal injury and mucosal barrier dysfunction, leading to an inflammatory response involving neutrophil localization and upregulation of pro-inflammatory cytokines. The severity of clinical manifestations is associated with the extent of the immune response, which requires mitigation for better clinical management. Probiotics could play a protective role in this disorder due to their immunomodulatory ability in gastrointestinal disorders. We assessed five single-strain and three multi-strain probiotics for their ability to modulate CDI fecal water (FW)-induced effects on T84 cells. The CDI-FW significantly (p < 0.05) decreased T84 cell viability. The CDI-FW-exposed cells also exhibited increased pro-inflammatory cytokine production as characterized by interleukin (IL)-8, C-X-C motif chemokine 5, macrophage inhibitory factor (MIF), IL-32, and tumor necrosis factor (TNF) ligand superfamily member 8. Probiotics were associated with strain-specific attenuation of the CDI-FW mediated effects, whereby Saccharomyces boulardii CNCM I-1079 and Lacticaseibacillus rhamnosus R0011 were most effective in reducing pro-inflammatory cytokine production and in increasing T84 cell viability. ProtecFlor™, Lactobacillus helveticus R0052, and Bifidobacterium longum R0175 showed moderate effectiveness, and L. rhamnosus GG R0343 along with the two other multi-strain combinations were the least effective. Overall, the findings showed that probiotic strains possess the capability to modulate the CDI-mediated inflammatory response in the gut lumen.

5.
J Immunol ; 204(9): 2523-2534, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238458

RESUMO

Recent evidence suggests that lactic acid bacteria communicate with host cells via secretome components to influence immune responses but less is known about gut-pathogen secretomes, impact of lactic acid bacteria secretomes on host-pathogen interactions, and the mechanisms underlying these interactions. Genome-wide microarrays and cytokine profiling were used to interrogate the impact of the Lactobacillus rhamnosus R0011 secretome (LrS) on TNF-α and Salmonella enterica subsp. enterica serovar Typhimurium secretome (STS)-induced outcomes in human intestinal epithelial cells. The LrS attenuated both TNF-α- and STS-induced gene expression involved in NF-κB and MAPK activation, as well as expression of genes involved in other immune-related signaling pathways. Specifically, the LrS induced the expression of dual specificity phosphatase 1 (DUSP1), activating transcription factor 3 (ATF3), and tribbles pseudokinase 3 (TRIB3), negative regulators of innate immune signaling, in HT-29 intestinal epithelial cells challenged with TNF-α or STS. TNF-α- and STS-induced acetylation of H3 and H4 histones was attenuated by the LrS, as was the production of TNF-α- and STS-induced proinflammatory cytokines and chemokines. Interestingly, the LrS induced production of macrophage migration inhibitory factor (MIF), a cytokine involved in host-microbe interactions at the gut interface. We propose that the LrS attenuates proinflammatory mediator expression through increased transcription of negative regulators of innate immune activity and changes in global H3 and H4 histone acetylation. To our knowledge, these findings provide novel insights into the complex multifaceted mechanisms of action behind secretome-mediated interdomain communication at the gut-mucosal interface.


Assuntos
Células Epiteliais/imunologia , Inflamação/imunologia , Intestinos/imunologia , Lacticaseibacillus rhamnosus/imunologia , Salmonelose Animal/imunologia , Salmonella typhimurium/imunologia , Fator de Necrose Tumoral alfa/imunologia , Acetilação , Animais , Linhagem Celular Tumoral , Citocinas/imunologia , Células Epiteliais/microbiologia , Expressão Gênica/imunologia , Células HT29 , Histonas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/imunologia , Inflamação/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Salmonelose Animal/microbiologia , Sorogrupo , Transdução de Sinais/fisiologia , Transcrição Gênica/imunologia
6.
PLoS One ; 12(1): e0169847, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28099447

RESUMO

Genome-wide transcriptional analysis in intestinal epithelial cells (IEC) can aid in elucidating the impact of single versus multi-strain probiotic combinations on immunological and cellular mechanisms of action. In this study we used human expression microarray chips in an in vitro intestinal epithelial cell model to investigate the impact of three probiotic bacteria, Lactobacillus helveticus R0052 (Lh-R0052), Bifidobacterium longum subsp. infantis R0033 (Bl-R0033) and Bifidobacterium bifidum R0071 (Bb-R0071) individually and in combination, and of a surface-layer protein (SLP) purified from Lh-R0052, on HT-29 cells' transcriptional profile to poly(I:C)-induced inflammation. Hierarchical heat map clustering, Set Distiller and String analyses revealed that the effects of Lh-R0052 and Bb-R0071 diverged from those of Bl-R0033 and Lh-R0052-SLP. It was evident from the global analyses with respect to the immune, cellular and homeostasis related pathways that the co-challenge with probiotic combination (PC) vastly differed in its effect from the single strains and Lh-R0052-SLP treatments. The multi-strain PC resulted in a greater reduction of modulated genes, found through functional connections between immune and cellular pathways. Cytokine and chemokine analyses based on specific outcomes from the TNF-α and NF-κB signaling pathways revealed single, multi-strain and Lh-R0052-SLP specific attenuation of the majority of proteins measured (TNF-α, IL-8, CXCL1, CXCL2 and CXCL10), indicating potentially different mechanisms. These findings indicate a synergistic effect of the bacterial combinations relative to the single strain and Lh-R0052-SLP treatments in resolving toll-like receptor 3 (TLR3)-induced inflammation in IEC and maintaining cellular homeostasis, reinforcing the rationale for using multi-strain formulations as a probiotic.


Assuntos
Gastroenterite/metabolismo , Redes Reguladoras de Genes/imunologia , Probióticos/farmacologia , Receptor 3 Toll-Like/metabolismo , Bifidobacterium longum subspecies infantis , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Epiteliais , Gastroenterite/tratamento farmacológico , Gastroenterite/imunologia , Perfilação da Expressão Gênica , Genoma Humano , Células HT29 , Humanos , Mediadores da Inflamação/metabolismo , Lactobacillus helveticus , Análise de Sequência com Séries de Oligonucleotídeos , Receptor 3 Toll-Like/imunologia
7.
Clin Infect Dis ; 62(12): 1479-1486, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27025836

RESUMO

BACKGROUND: Recurrent Clostridium difficile infection (RCDI) is associated with repeated antibiotic treatment and the enhanced growth of antibiotic-resistant microbes. This study tested the hypothesis that patients with RCDI would harbor large numbers of antibiotic-resistant microbes and that fecal microbiota transplantation (FMT) would reduce the number of antibiotic-resistant genes. METHODS: In a single center study, patients with RCDI (n = 20) received FMT from universal donors via colonoscopy. Stool samples were collected from donors (n = 3) and patients prior to and following FMT. DNA was extracted and shotgun metagenomics performed. Results as well as assembled libraries from a healthy cohort (n = 87) obtained from the Human Microbiome Project were aligned against the NCBI bacterial taxonomy database and the Comprehensive Antibiotic Resistance Database. Results were corroborated through a DNA microarray containing 354 antibiotic resistance (ABR) genes. RESULTS: RCDI patients had a greater number and diversity of ABR genes compared with donors and healthy controls. Beta-lactam, multidrug efflux pumps, fluoroquinolone, and antibiotic inactivation ABR genes were increased in RCDI patients, although donors primarily had tetracycline resistance. RCDI patients were dominated by Proteobacteria with Escherichia coli and Klebsiella most prevalent. FMT resulted in a resolution of symptoms that correlated directly with a decreased number and diversity of ABR genes and increased Bacteroidetes and Firmicutes with reduced Proteobacteria. ABR gene profiles were maintained in recipients for up to a year following FMT. CONCLUSIONS: RCDI patients have increased numbers of antibiotic-resistant organisms. FMT is effective in the eradication of pathogenic antibiotic-resistant organisms and elimination of ABR genes.


Assuntos
Clostridioides difficile/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Enterocolite Pseudomembranosa/terapia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/farmacologia , Clostridioides difficile/genética , Enterocolite Pseudomembranosa/microbiologia , Fezes/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
8.
Appl Environ Microbiol ; 80(5): 1692-700, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24375132

RESUMO

A commercially available product containing three probiotic bacterial strains (Lactobacillus helveticus R0052, Bifidobacterium longum subsp. infantis R0033, and Bifidobacterium bifidum R0071) was previously shown in animal trials to modulate both TH1 and TH2 immune responses. Clinical studies on this combination of bacteria have also shown positive health effects against seasonal winter diseases and rotavirus infection. The goal of this study was to use a well-established in vitro intestinal epithelial (HT-29) cell model that has been shown to constitutively express double-stranded RNA (dsRNA) sensors (Toll-like receptor 3 [TLR3], retinoic acid-inducible gene I, melanoma differentiation-associated gene 5, and dsRNA-activated protein kinase). By using the HT-29 cell model, we wanted to evaluate whether or not this combination of three bacteria had the capacity to immune modulate the host cell response to a dsRNA ligand, poly(I·C). Using a custom-designed, two-color expression microarray targeting genes of the human immune system, we investigated the response of HT-29 cells challenged with poly(I·C) both in the presence and in the absence of the three probiotic bacteria. We observed that the combination of the three bacteria had a major impact on attenuating the expression of genes connected to proinflammatory TH1 and antiviral innate immune responses compared to that obtained by the poly(I·C)-only challenge. Major pathways through which the multistrain combination may be eliciting its immune-modulatory effect include the TLR3 domain-containing adapter-inducing beta interferon (TRIF), mitogen-activated protein kinase, and NF-κB signaling pathways. Such a model may be useful for selecting potential biomarkers for the design of future clinical trials.


Assuntos
Bifidobacterium/imunologia , Células Epiteliais/imunologia , Fatores Imunológicos/farmacologia , Lactobacillus helveticus/imunologia , Poli I-C/imunologia , Probióticos/farmacologia , Linhagem Celular , Perfilação da Expressão Gênica , Humanos
9.
Ther Adv Respir Dis ; 4(5): 259-70, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20929951

RESUMO

BACKGROUND AND METHODS: The immunomodulatory activity of a synbiotic combination containing three bacterial strains (Lactobacillus helveticus R0052, Bifidobacterium longum subsp. infantis R0033 and Bifidobacterium bifidum R0071) and short-chain fructooligosaccharide was examined in two distinct infectious rat models. In the T(h)1 model, Wistar rats were administered the synbiotic combination for 2 weeks prior to challenge with a single oral dose of enterotoxigenic Escherichia coli or vehicle. In the T(h)2 model, pretreated rats were challenged with a single subcutaneous dose of hook worm, Nippostrongylus brasiliensis. Blood samples were collected 3 hours or 4 days postchallenge and serum levels of pro- and anti-inflammatory cytokines were measured. RESULTS: Significant reductions in pro-inflammatory cytokines interleukin (IL)-1α, IL-1ß, IL-6, and tumour necrosis factor (TNF)-α were observed in both models suggesting a single, unifying mode of action on an upstream regulator. The N. brasiliensis study also compared the effect of the individual strains to synbiotic. For most of cytokines the combination appeared to average the effect of the individual strains with the exception of IL-4 and IL-10 where there was apparent synergy for the combination. Furthermore, the cytokine response varied by strain. CONCLUSIONS: It was concluded that this synbiotic combination of these three microbes could be beneficial in both T(h)1 and T(h)2 diseases.


Assuntos
Infecções por Escherichia coli/terapia , Infecções por Strongylida/terapia , Simbióticos , Animais , Bifidobacterium , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Interleucinas/imunologia , Lactobacillus helveticus , Masculino , Nippostrongylus/imunologia , Oligossacarídeos/administração & dosagem , Ratos , Ratos Wistar , Infecções por Strongylida/imunologia , Células Th1/imunologia , Células Th1/microbiologia , Células Th2/imunologia , Células Th2/parasitologia , Fator de Necrose Tumoral alfa/imunologia
10.
Br J Nutr ; 102(10): 1420-5, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19563693

RESUMO

Myocardial infarction (MI) stimulates the release of pro-inflammatory substances that induce apoptosis in the limbic system. Pro-inflammatory cytokines are considered as the root cause of apoptosis, although the mechanism is not fully explained and/or understood at this time. In addition, depression may induce gastrointestinal perturbations that maintain the elevated levels of pro-inflammatory cytokines. It has been shown that some specific probiotic formulations may reduce gastrointestinal problems induced by stress and the pro/anti-inflammatory cytokine ratio. Therefore, we hypothesised that probiotics, when given prophylactically, may diminish the apoptosis propensity in the limbic system following a MI. Male adult Sprague-Dawley rats were given probiotics (Lactobacillus helveticus and Bifidobacterium longum in combination) or placebo in their drinking-water for four consecutive weeks. A MI was then induced in the rats by occluding the left anterior coronary artery for 40 min. Rats were killed following a 72 h reperfusion period. Infarct size was not different in the two groups. Bax/Bcl-2 (pro-apoptotic/anti-apoptotic) ratio and caspase-3 (pro-apoptotic) activity were reduced in the amygdala (lateral and medial), as well as in the dentate gyrus in the probiotics group when compared with the placebo. Akt activity (anti-apoptotic) was increased in these same three regions. No significant difference was observed in Ca1 and Ca3 for the different markers measured. In conclusion, the probiotics L. helveticus and B. longum, given in combination as preventive therapy, reduced the predisposition of apoptosis found in different cerebral regions following a MI.


Assuntos
Apoptose/efeitos dos fármacos , Bifidobacterium/fisiologia , Lactobacillus helveticus/fisiologia , Sistema Límbico/efeitos dos fármacos , Infarto do Miocárdio/fisiopatologia , Animais , Caspase 3/metabolismo , Dieta , Ativação Enzimática , Sistema Límbico/citologia , Sistema Límbico/patologia , Masculino , Fenômenos Fisiológicos da Nutrição , Probióticos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
11.
Can J Microbiol ; 54(5): 391-400, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18449224

RESUMO

Probiotics, known for their prophylactic and therapeutic properties, are routinely used by the medical community in various regions of the world. In some Asian countries, these products are controlled as pharmaceutical substances and must adhere to strict regulatory guidelines. However, outside of Europe where the European Food Safety Authority has recently adopted a Qualified Presumption of Safety approach for probiotics used in food and feed, current safety requirements do not necessitate screening for the presence of virulence and other risk factors, which may result in the inadvertent use of probiotic strains harboring harmful genes. A safety evaluation was conducted on Enterococcus faecium R0026 and Bacillus subtilis R0179 used in several commercial probiotic products marketed in Asia. Molecular techniques were used to verify the identity of each strain and antibiotic resistance profiles were determined towards clinically relevant antibiotics. Strains were subsequently screened for the presence of enterotoxins and virulence factors and were subjected to 28 days of repeated high-dose oral toxicity testing in rats. No risk factors or aberrant activities were identified using such a detailed approach. Thus, both microbes were deemed to pose low risk to the consumer and, therefore, safe for use as probiotics.


Assuntos
Bacillus subtilis/isolamento & purificação , Qualidade de Produtos para o Consumidor/normas , Enterococcus faecium/isolamento & purificação , Probióticos/química , Animais , Antibacterianos/farmacologia , Ásia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/patogenicidade , Bacillus subtilis/fisiologia , Aderência Bacteriana , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , DNA Bacteriano/genética , DNA Ribossômico/genética , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/patogenicidade , Enterococcus faecium/fisiologia , Células Epiteliais/microbiologia , Feminino , Microbiologia de Alimentos , Infecções por Bactérias Gram-Positivas/microbiologia , Células HT29 , Humanos , Masculino , Dados de Sequência Molecular , Plasmídeos/genética , Probióticos/metabolismo , Probióticos/toxicidade , RNA Ribossômico 16S/genética , Ratos , Ratos Sprague-Dawley , Fatores de Virulência/genética
12.
J Agric Food Chem ; 55(21): 8731-6, 2007 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-17880156

RESUMO

Selenium has a central role in antioxidant pathways as a cofactor to glutathione peroxidase. The present study evaluated the effects of four different preparations of inactivated yeast containing various concentrations of selenium and glutathione on a combined atherosclerosis and diabetes hamster model. The hamsters were supplemented with the yeast products for three months. The enriched yeast with the highest selenium and glutathione levels reduced the weight loss induced by diabetes, inhibited an increase in plasma cholesterol and triglyceride caused by a high-cholesterol and high-fat diet, increased the time taken for oxidation of lower density lipoproteins (lag time), and inhibited the formation of atherosclerosis better than low selenium/glutathione yeast supplementation. It was concluded that the yeast prepared to provide high selenium and high glutathione was the best for effecting beneficial changes in glutathione, cholesterol, atherosclerosis, and for demonstrating an antioxidant effect. The high selenium and low glutathione yeast was the best for improving selenium and glucose levels.


Assuntos
Aterosclerose/terapia , Diabetes Mellitus Experimental/terapia , Glutationa/administração & dosagem , Saccharomyces cerevisiae , Selênio/administração & dosagem , Animais , Aterosclerose/sangue , Cricetinae , Diabetes Mellitus Experimental/sangue , Suplementos Nutricionais , Glutationa/sangue , Lipídeos/sangue , Masculino , Mesocricetus , Probióticos , Saccharomyces cerevisiae/química , Selênio/sangue
13.
Cell Microbiol ; 9(2): 356-67, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16925785

RESUMO

Adherence of intestinal pathogens, including Escherichia coli O157:H7, to human intestinal epithelial cells is a key step in pathogenesis. Probiotic bacteria, including Lactobacillus helveticus R0052 inhibit the adhesion of E. coli O157:H7 to epithelial cells, a process which may be related to specific components of the bacterial surface. Surface-layer proteins (Slps) are located in a paracrystalline layer outside the bacterial cell wall and are thought to play a role in tissue adherence. However, the ability of S-layer protein extract derived from probiotic bacteria to block adherence of enteric pathogens has not been investigated. Human epithelial (HEp-2 and T84) cells were treated with S-layer protein extract alone, infected with E. coli O157:H7, or pretreated with S-layer protein extract prior to infection to determine their importance in the inhibition of pathogen adherence. The effects of S-layer protein extracts were characterized by phase-contrast and immunofluorescence microscopy and measurement of the transepithelial electrical resistance of polarized monolayers. Pre-treatment of host epithelial cells with S-layer protein extracts prior to E. coli O157:H7 infection decreased pathogen adherence and attaching-effacing lesions in addition to preserving the barrier function of monolayers. These in vitro studies indicate that a non-viable constituent derived from a probiotic strain may prove effective in interrupting the infectious process of an intestinal pathogen.


Assuntos
Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/farmacologia , Células Epiteliais/efeitos dos fármacos , Escherichia coli O157/efeitos dos fármacos , Lactobacillus helveticus/química , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Proteínas de Bactérias/farmacologia , Linhagem Celular , Células Epiteliais/microbiologia , Escherichia coli O157/metabolismo
14.
Infect Immun ; 73(8): 5183-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16041036

RESUMO

The aim of this study was to determine if probiotics reduce epithelial injury following exposure to Escherichia coli O157:H7 and E. coli O127:H6. The pretreatment of intestinal (T84) cells with lactic acid-producing bacteria reduced the pathogen-induced drop in transepithelial electrical resistance. These findings demonstrate that probiotics prevent epithelial injury induced by attaching-effacing bacteria.


Assuntos
Aderência Bacteriana/fisiologia , Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Escherichia coli O157/metabolismo , Probióticos/metabolismo , Actinina/metabolismo , Células Epiteliais/microbiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/terapia , Humanos , Lactobacillus acidophilus/metabolismo
15.
Biochemistry ; 44(2): 726-33, 2005 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-15641799

RESUMO

The natural resistance-associated macrophage protein (Nramp) family is functionally conserved in bacteria and eukarya; Nramp homologues function as proton-dependent membrane transporters of divalent metals. Sequence analyses indicate that five phylogenetic groups comprise the Nramp family, three bacterial and two eukaryotic, which are distinct from a more distantly related group of microbial sequences (Nramp outgroup). The Nramp family and outgroup share many conserved residues, suggesting they derived from a common ancestor and raising the possibility that the residues invariant in the Nramp family that correspond to residues which are different but also conserved in the outgroup represent candidate sites of functional divergence of the Nramp family. Four Nramp family-specific residues were identified within transmembrane domains 1, 6, and 11, and replaced by the corresponding invariant outgroup residues in the Escherichia coli Nramp ortholog (the proton-dependent manganese transporter, MntH of group A, EcoliA). The resulting mutants (Asp(34)Gly, Asn(37)Thr, His(211)Tyr, and Asn(401)Gly) were tested for both divalent metal uptake and proton transport; quasi-simultaneous analyses of uptake of metals and protons revealed for the first time protons and metals cotransport by a bacterial Nramp homologue. Additional mutations were studied for comparison (Asp(34)Asn, Asn(37)Asp and Asn(37)Val, Asn(401)Thr, His(211)Ala, His(216)Ala, and His(216)Arg). EcoliA activity was impaired after each of the Nramp/outgroup substitutions, as well as after more conservative replacements, showing that the tested sites are all important for metal uptake and metal-dependent H(+) transport. It is proposed that co-occurrence of these four Nramp-specific transmembrane residues may have contributed to the emergence of this family of metal and proton cotransporters.


Assuntos
Aminoácidos/química , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Evolução Molecular , Metais Pesados/metabolismo , Prótons , Aminoácidos/genética , Asparagina/química , Asparagina/genética , Ácido Aspártico/química , Ácido Aspártico/genética , Cádmio/metabolismo , Proteínas de Transporte de Cátions/genética , Permeabilidade da Membrana Celular/genética , Cobalto/metabolismo , Proteínas de Escherichia coli/genética , Compostos Ferrosos/metabolismo , Histidina/química , Histidina/genética , Manganês/metabolismo , Mutagênese Sítio-Dirigida , Transporte Proteico/genética , Proteínas Proto-Oncogênicas c-myc/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA