Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Gut Pathog ; 14(1): 30, 2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794638

RESUMO

BACKGROUND: Obesity is a worldwide health problem and a significant risk factor for diabetes and cardiovascular diseases. Gut microbiota (GM) plays an essential role in obesity, and prebiotics such as polyphenols could be one way to improve microbial dysbiosis-induced obesity. OBJECTIVE: This study was designed to assess the effectiveness of grape seed and skin extract (GSSE), and/or orlistat on obese rats fed with high fat diet by targeting GM modulations. The impact of treatments was also studied in non-obese rats. MATERIAL AND METHODS: Rats were rendered obese or kept with a standard diet for three months. Then they were treated either with GSSE or orlistat or with the combined treatment (GSOR) during three months and then sacrificed. Adipose tissues, blood and faeces were collected and analyzed. RESULTS: In obese rats and to a lesser extent in non-obese rats, treatments decreased the weight of various adipose tissues and the serum levels of cholesterol, LDL, triglycerides, lipase, and CRP and increased HDL and adiponectin. GSOR treatment was even more efficient that orlistat. Obese rats had less GM diversity than non-obese rats and orlistat reduced it even more. However, diversity was restored with GSSE and GSOR treatments. Potential pathogenic Streptococcus alactolyticus/gallolyticus species were greatly increased in obese rats and drastically reduced with the treatments, as wells as other potential pathobionts. CONCLUSIONS: GSSE exerts beneficial effects in obese rats and restores, at least partially, the observed dysbiosis. GSOR induced the highest beneficial effect. Moreover, the various treatments could also enhance physiological and GM modifications in non obese rats.

2.
FASEB J ; 33(4): 4921-4935, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30596521

RESUMO

Given the growing evidence that gut dysfunction, including changes in gut microbiota composition, plays a critical role in the development of inflammation and metabolic diseases, the identification of novel probiotic bacteria with immunometabolic properties has recently attracted more attention. Herein, bacterial strains were first isolated from dairy products and human feces and then screened in vitro for their immunomodulatory activity. Five selected strains were further analyzed in vivo, using a mouse model of diet-induced obesity. C57BL/6 mice were fed a high-fat high-sucrose diet, in combination with 1 of 3 Lactobacillus strains (Lb38, L. plantarum; L79, L. paracasei/casei; Lb102, L. rhamnosus) or Bifidobacterium strains (Bf26, Bf141, 2 different strains of B. animalis ssp. lactis species) administered for 8 wk at 109 colony-forming units/d. Whereas 3 strains showed only modest (Lb38, Bf26) or no (L79) effects, Lb102 and Bf141 reduced diet-induced obesity, visceral fat accretion, and inflammation, concomitant with improvement of glucose tolerance and insulin sensitivity. Further analysis revealed that Lb102 and Bf141 enhanced intestinal integrity markers in association with selective changes in gut microbiota composition. We have thus identified 2 new potential probiotic bacterial strains with immunometabolic properties to alleviate obesity development and associated metabolic disturbances.-Le Barz, M., Daniel, N., Varin, T. V., Naimi, S., Demers-Mathieu, V., Pilon, G., Audy, J., Laurin, E., Roy, D., Urdaci, M. C., St-Gelais, D., Fliss, I, Marette, A. In vivo screening of multiple bacterial strains identifies Lactobacillus rhamnosus Lb102 and Bifidobacterium animalis ssp. lactis Bf141 as probiotics that improve metabolic disorders in a mouse model of obesity.


Assuntos
Bifidobacterium animalis/fisiologia , Lacticaseibacillus rhamnosus/fisiologia , Obesidade/dietoterapia , Obesidade/microbiologia , Probióticos/uso terapêutico , Tecido Adiposo/metabolismo , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/metabolismo , Microbioma Gastrointestinal/fisiologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , RNA Ribossômico 16S/genética
3.
Front Microbiol ; 9: 1537, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30042756

RESUMO

Preventive actions of probiotics as antidiarrheal agents are well documented, but their mechanisms are poorly understood. Two selected probiotics, Bacillus subtilis CU1 and Lactobacillus plantarum CNCM I-4547, were tested in mouse experimental models of diarrhea and the possible mechanisms of action were investigated. Diarrhea was induced in mice by oral castor oil administration or by i.v. injection of lipopolysaccharide (LPS) of Salmonella enteritis. The antidiarrheal drug loperamide was used as control. Fecal water excretion was quantified for 2 h and paracellular permeability and electrical parameters of the colon were assessed in Ussing chambers. The expression of colonic exchangers or channels and of Toll-like receptor 4 (TLR4) was assessed by immunohistochemistry. Prophylactic treatment with B. subtilis CU1 or with L. plantarum CNCM I-4547 reduced LPS-induced diarrhea. The reduction of water excretion was in the same range as those induced by loperamide. In the castor oil model, this effect was only observed with B. subtilis CU1. The two probiotic treatments abolished the increase in paracellular permeability induced by LPS, but not by castor oil. However, only L. plantarum CNCM I-4547 treatment decreased the colonic expression of TLR-4. After B. subtilis CU1, colonic expression of cystic fibrosis transmembrane conductance regulator (CFTR) was reduced and that of Na+/H+ exchanger 3 (NHE3) increased. B. subtilis CU1 may increase the capacity of the colon to absorb excess of water in diarrheic conditions by acting on CFTR and NHE3 expression. The two probiotics strains showed an impact on diarrhea through limitation of water excretion that may involve paracellular permeability or electrolyte transport for L. plantarum CNCM I-4547 and B. subtilis CU1 respectively.

4.
Cell Microbiol ; 20(11): e12871, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29920917

RESUMO

Prostaglandin E2 (PGE2 ) plays a critical role in intestinal mucosal tolerance and barrier integrity. Cyclooxygenase-2 (COX-2)-dependent PGE2 production involves mobilisation of arachidonic acid. Lactobacillus rhamnosus GG (LbGG) is one of the most widely used probiotics reported to colonise the colonic mucosa. LbGG contributes to the protection of the small intestine against radiation injury through the repositioning of mucosal COX-2 expressing cells. However, it is unknown if LbGG modulates PGE2 production in the colonic mucosa under homeostasis and the major cellular elements involved in these processes. Colonic epithelial and CD90+ mesenchymal stromal cells, also known as (myo) fibroblasts (CMFs), are abundant innate immune cells in normal colonic mucosa able to produce PGE2 . Herein, we tested the hypothesis that under colonic mucosal homeostasis, LbGG modulates the eicosanoid pathway resulting in increased PGE2 production in both epithelial and stromal cells. Among the five tested human colonic epithelial cell lines, only exposure of Caco-2 to LbGG for 24 hr led to the mobilisation of arachidonic acid with concomitant increase in the components within the leukotriene and COX-2-dependent PGE2 pathways. By contrast, CMFs isolated from the normal human colonic mucosa responded to LbGG with increased expression of COX-2 and PGE2 in the prostaglandin pathway, but not 5-LO in the leukotriene pathway. Oral gavage of C57BL/6 mice for 5 days with LbGG (5 × 108 Colony-Forming Unit (CFU)/dose) increased COX-2 expression in the colonic mucosa. The majority of cells upregulating COX-2 protein expression were located in the colonic lamina propria and colocalised with α-SMA+ cells corresponding to the CMF phenotype. This process was myeloid differentiation factor-88-dependent, because silencing of myeloid differentiation factor-88 expression in CMFs abrogated LbGG-induced upregulation of COX-2 in culture and in vivo. Taken together, our data suggest that LbGG increases release of COX-2-mediated PGE2 , contributing to the maintenance of mucosal homeostasis in the colon and CMFs are among the major contributors to this process.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Lacticaseibacillus rhamnosus , Fator 88 de Diferenciação Mieloide/metabolismo , Probióticos/farmacologia , Administração Oral , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Ácido Araquidônico/metabolismo , Células CACO-2 , Colo/citologia , Colo/microbiologia , Homeostase , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/genética , Miofibroblastos/metabolismo , Miofibroblastos/microbiologia , Probióticos/administração & dosagem
5.
Front Immunol ; 8: 88, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28239378

RESUMO

Enterococcus species, principally Enterococcus faecium are used as probiotics since a long time with preference in animal applications but safety considerations were updated and also new uses as probiotics can be envisaged. Fifteen Enterococcus strains isolated from different foods were identified and analyzed for virulence factors and antibiotic resistance. Three Enterococcus durans strains were selected to study their immunomodulatory properties on PBMC and Caco2 cells. Two strains presented a profile toward a mild inflammatory Th1 response considering TNF-α/IL-10 and IL-1ß/IL-10 cytokines ratios. The third strain EP1, presented an anti-inflammatory potential and was selected for in vivo studies. In mice, the strain was well tolerated and did not cause any adverse effects. EP1 administration increased the amount of IgA+ cells in mesenteric lymph node (MLN) after 7 days of administration. In fecal samples, the IgA content increased gradually and significantly from day 7 to day 21 in treated group. Additionally, IL-17, IL-6, IL-1ß, IFN-γ, and CXCL1 gene expression significantly decreased on day 21 in Peyer's patches and IL-17 decreased in MLN. Mice treated with the probiotic showed significant lower mRNA levels of pro-inflammatory cytokines and mucins in the ileum at day 7 while their expression was normalized at day 21. Colonic expression of il-1ß, il6, and mucins remain diminished at day 21. Ileum and colon explants from treated mice stimulated in vitro with LPS showed a significant reduction in IL-6 and an increase in IL-10 secretion suggesting an in vivo protective effect of the probiotic treatment against a proinflammatory stimulus. Interestingly, analysis of feces microbiota demonstrated that EP1 administration increase the amount of Faecalibacterium prausnitzii, a butyrate-producing bacteria, which is known for its anti-inflammatory effects. In conclusion, we demonstrated that EP1 strain is a strong sIgA inducer and possess mucosal anti-inflammatory properties. This strain also modulates gut microbiota increasing Faecalibacterium prausnitzii, a functionally important bacterium. Thus, E. durans EP1 is not only a good candidate to increases F. prausnitzii in some cases of dysbiosis but can also be interesting in gut inflammatory disorders therapy.

6.
Regul Toxicol Pharmacol ; 83: 54-65, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27825987

RESUMO

Bacillus subtilis CU1 is a recently described probiotic strain with beneficial effects on immune health in elderly subjects. The following work describes a series of studies supporting the safety of the strain for use as an ingredient in food and supplement preparations. Using a combination of 16S rDNA and gyrB nucleotide analyses, the species was identified as a member of the Bacillus subtilis complex (B. subtilis subsp. spizizenii). Further characterization of the organism at the strain level was achieved using random amplified polymorphic DNA polymerase chain reaction (RAPD PCR) and pulsed field gel electrophoresis (PFGE) analyses. B. subtilis CU1 did not demonstrate antibiotic resistance greater than existing regulatory cutoffs against clinically important antibiotics, did not induce hemolysis or produce surfactant factors, and was absent of toxigenic activity in vitro. Use of B. subtilis CU1 as a probiotic has recently been evaluated in a 16-week randomized, double-blind, placebo-controlled, parallel-arm study, in which 2 × 109 spores per day of B. subtilis CU1 were administered for a total 40 days to healthy elderly subjects (4 consumption periods of 10 days separated by 18-day washouts). This work describes safety related endpoints not previously reported. B. subtilis CU1 was safe and well-tolerated in the clinical subjects without undesirable physiological effects on markers of liver and kidney function, complete blood counts, hemodynamic parameters, and vital signs.


Assuntos
Bacillus subtilis/fisiologia , Inocuidade dos Alimentos , Probióticos/toxicidade , Idoso , Antibacterianos/farmacologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/genética , Bacillus subtilis/patogenicidade , Qualidade de Produtos para o Consumidor , Impressões Digitais de DNA , DNA Girase/genética , DNA Bacteriano/genética , Método Duplo-Cego , Farmacorresistência Bacteriana , Eletroforese em Gel de Campo Pulsado , Feminino , Hemólise , Humanos , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/genética , Ribotipagem , Medição de Risco , Fatores de Tempo
7.
Antimicrob Agents Chemother ; 60(6): 3445-54, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27001810

RESUMO

Although the use of probiotics based on Bacillus strains to fight off intestinal pathogens and antibiotic-associated diarrhea is widespread, the mechanisms involved in producing their beneficial effects remain unclear. Here, we studied the ability of compounds secreted by the probiotic Bacillus clausii strain O/C to counteract the cytotoxic effects induced by toxins of two pathogens, Clostridium difficile and Bacillus cereus, by evaluating eukaryotic cell viability and expression of selected genes. Coincubation of C. difficile and B. cereus toxic culture supernatants with the B. clausii supernatant completely prevented the damage induced by toxins in Vero and Caco-2 cells. The hemolytic effect of B. cereus was also avoided by the probiotic supernatant. Moreover, in these cells, the expression of rhoB, encoding a Rho GTPase target for C. difficile toxins, was normalized when C. difficile supernatant was pretreated using the B. clausii supernatant. All of the beneficial effects observed with the probiotic were abolished by the serine protease inhibitor phenylmethylsulfonyl fluoride (PMSF). Suspecting the involvement of a secreted protease in this protective effect, a protease was purified from the B. clausii supernatant and identified as a serine protease (M-protease; GenBank accession number Q99405). Experiments on Vero cells demonstrated the antitoxic activity of the purified protease against pathogen supernatants. This is the first report showing the capacity of a protease secreted by probiotic bacteria to inhibit the cytotoxic effects of toxinogenic C. difficile and B. cereus strains. This extracellular compound could be responsible, at least in part, for the protective effects observed for this human probiotic in antibiotic-associated diarrhea.


Assuntos
Bacillus cereus/patogenicidade , Bacillus clausii/metabolismo , Toxinas Bacterianas/toxicidade , Clostridioides difficile/patogenicidade , Probióticos/farmacologia , Subtilisinas/metabolismo , Animais , Células CACO-2 , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Meios de Cultivo Condicionados/farmacologia , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Fluoreto de Fenilmetilsulfonil/farmacologia , Inibidores de Proteases/farmacologia , Subtilisinas/antagonistas & inibidores , Células Vero , Proteína rhoB de Ligação ao GTP/metabolismo
8.
Immun Ageing ; 12: 24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26640504

RESUMO

BACKGROUND: Bacillus probiotics health benefits have been until now quite poorly studied in the elderly population. This study aimed to assess the effects of Bacillus subtilis CU1 consumption on immune stimulation and resistance to common infectious disease (CID) episodes in healthy free-living seniors. RESULTS: One hundred subjects aged 60-74 were included in this randomized, double-blind, placebo-controlled, parallel-arms study. Subjects consumed either the placebo or the probiotic (2.10(9) B. subtilis CU1 spores daily) by short periodical courses of 10 days intermittently, alternating 18-day course of break. This scheme was repeated 4 times during the study. Symptoms of gastrointestinal and upper/lower respiratory tract infections were recorded daily by the subjects throughout the study (4 months). Blood, saliva and stool samples were collected in a predefined subset of the first forty-four subjects enrolled in the study. B. subtilis CU1 supplementation did not statistically significantly decrease the mean number of days of reported CID symptoms over the 4-month of study (probiotic group: 5.1 (7.0) d, placebo group: 6.6 (7.3) d, P = 0.2015). However, in the subset of forty-four randomized subjects providing biological samples, we showed that consumption of B. subtilis CU1 significantly increased fecal and salivary secretory IgA concentrations compared to the placebo. A post-hoc analysis on this subset showed a decreased frequency of respiratory infections in the probiotc group compared to the placebo group. CONCLUSION: Taken together, our study provides evidence that B. subtilis CU1 supplementation during the winter period may be a safe effective way to stimulate immune responses in elderly subjects.

9.
Diabetes Metab J ; 39(5): 448-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26566505

RESUMO

[This corrects the article on p. 291 in vol. 39, PMID: 26301190.].

10.
Diabetes Metab J ; 39(4): 291-303, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26301190

RESUMO

Over the past decade, growing evidence has established the gut microbiota as one of the most important determinants of metabolic disorders such as obesity and type 2 diabetes. Indeed, obesogenic diet can drastically alter bacterial populations (i.e., dysbiosis) leading to activation of pro-inflammatory mechanisms and metabolic endotoxemia, therefore promoting insulin resistance and cardiometabolic disorders. To counteract these deleterious effects, probiotic strains have been developed with the aim of reshaping the microbiome to improve gut health. In this review, we focus on benefits of widely used probiotics describing their potential mechanisms of action, especially their ability to decrease metabolic endotoxemia by restoring the disrupted intestinal mucosal barrier. We also discuss the perspective of using new bacterial strains such as butyrate-producing bacteria and the mucolytic Akkermansia muciniphila, as well as the use of prebiotics to enhance the functionality of probiotics. Finally, this review introduces the notion of genetically engineered bacterial strains specifically developed to deliver anti-inflammatory molecules to the gut.

11.
Microbiology (Reading) ; 161(Pt 4): 708-18, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25635270

RESUMO

The vaginal microbiota of healthy, fertile women is dominated by lactobacilli. As a defence mechanism, these bacteria produce H2O2 to discourage colonization of the vagina by undesirable micro-organisms. In particular, Lactobacillus jensenii CECT 4306 is a strong producer of H2O2 and has been found to protect itself from the bactericidal effects of this compound through the activity of extracellular peroxidases. However, this peroxidase activity is dependent on the presence of Fe(3+), which is found in elevated concentrations in the vaginal mucosa as a consequence of the menstrual discharge. The aim of the present work was to evaluate whether Fe(3+) is able to modulate other potential probiotic properties of strain 4306. We found that Fe(3+) enhances the adhesion of L. jensenii CECT 4306 to mucin and to HT-29 and HT-29 MTX cells, and, in addition, improves the anti-inflammatory profile, as judged by an increase in the ratio of IL-10/IL-12p70 that were secreted by macrophages. A comparison of total, secreted and surface proteins produced in the presence and absence of Fe(3+) revealed significant differences in the concentration of the moonlighting protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In conclusion, Fe(3+) seems to improve the probiotic characteristics of L. jensenii CECT 4306, and future research of the interactions of this strain with its vaginal environment may reveal further information about different aspects of its probiotic potential.


Assuntos
Ferro/metabolismo , Lactobacillus/metabolismo , Probióticos , Vagina/microbiologia , Aderência Bacteriana , Proteínas de Bactérias , Linhagem Celular , Feminino , Compostos Férricos/metabolismo , Humanos , Imunomodulação , Microbiota , Mucosa/imunologia , Mucosa/microbiologia , Proteoma , Proteômica , Vagina/imunologia
12.
Antonie Van Leeuwenhoek ; 106(4): 693-706, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25090957

RESUMO

Probiotics represent a potential strategy to influence the host's immune system thereby modulating immune response. Lipoteichoic Acid (LTA) is a major immune-stimulating component of Gram-positive cell envelopes. This amphiphilic polymer, anchored in the cytoplasmic membrane by means of its glycolipid component, typically consists of a poly (glycerol-phosphate) chain with D-alanine and/or glycosyl substitutions. LTA is known to stimulate macrophages in vitro, leading to secretion of inflammatory mediators such as Nitric Oxide (NO). This study investigates the structure-activity relationship of purified LTA from three probiotic Bacillus strains (Bacillus cereus CH, Bacillus subtilis CU1 and Bacillus clausii O/C). LTAs were extracted from bacterial cultures and purified. Chemical modification by means of hydrolysis at pH 8.5 was performed to remove D-alanine. The molecular structure of native and modified LTAs was determined by (1)H NMR and GC-MS, and their inflammatory potential investigated by measuring NO production by RAW 264.7 macrophages. Structural analysis revealed several differences between the newly characterized LTAs, mainly relating to their D-alanylation rates and poly (glycerol-phosphate) chain length. We observed induction of NO production by LTAs from B. subtilis and B. clausii, whereas weaker NO production was observed with B. cereus. LTA dealanylation abrogated NO production independently of the glycolipid component, suggesting that immunomodulatory potential depends on D-alanine substitutions. D-alanine may control the spatial configuration of LTAs and their recognition by cell receptors. Knowledge of molecular mechanisms behind the immunomodulatory abilities of probiotics is essential to optimize their use.


Assuntos
Alanina/análise , Alanina/imunologia , Bacillus/química , Lipopolissacarídeos/análise , Lipopolissacarídeos/imunologia , Probióticos/química , Ácidos Teicoicos/análise , Ácidos Teicoicos/imunologia , Animais , Bacillus/imunologia , Linhagem Celular , Cromatografia Gasosa-Espectrometria de Massas , Hidrólise , Fatores Imunológicos/análise , Fatores Imunológicos/química , Fatores Imunológicos/imunologia , Lipopolissacarídeos/química , Macrófagos/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Camundongos , Estrutura Molecular , Óxido Nítrico/metabolismo , Relação Estrutura-Atividade , Ácidos Teicoicos/química
13.
Biomed Res Int ; 2014: 351204, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24991549

RESUMO

Inflammatory bowel disease (IBD) is an autoimmune disease characterized by a chronic inflammation of the gastrointestinal tract mucosa and is related to an abnormal immune response to commensal bacteria. Our aim of the present work has been to explore the levels of antibodies (IgG and IgA) raised against extracellular proteins produced by LAB and its association with IBD. We analyzed, by Western-blot and ELISA, the presence of serum antibodies (IgA and IgG) developed against extracellular protein fractions produced by different food bacteria from the genera Bifidobacterium and Lactobacillus. We used a sera collection consisting of healthy individuals (HC, n = 50), Crohn's disease patients (CD, n = 37), and ulcerative colitis patients (UC, n = 15). Levels of IgA antibodies developed against a cell-wall hydrolase from Lactobacillus casei subsp. rhamnosus GG (CWH) were significantly higher in the IBD group (P < 0.002; n = 52). The specificity of our measurements was confirmed by measuring IgA antibodies developed against the CWH peptide 365-VNTSNQTAAVSAS-377. IBD patients appeared to have different immune response to food bacteria. This paper sets the basis for developing systems for early detection of IBD, based on the association of high levels of antibodies developed against extracellular proteins from food and probiotic bacteria.


Assuntos
Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Doenças Inflamatórias Intestinais/imunologia , Lacticaseibacillus casei/imunologia , Probióticos , Adulto , Anticorpos/sangue , Anticorpos/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/isolamento & purificação , Bifidobacterium/imunologia , Bifidobacterium/patogenicidade , Colite Ulcerativa/sangue , Colite Ulcerativa/microbiologia , Doença de Crohn/sangue , Doença de Crohn/microbiologia , Feminino , Microbiologia de Alimentos , Humanos , Imunidade Celular , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Doenças Inflamatórias Intestinais/sangue , Doenças Inflamatórias Intestinais/patologia , Lactobacillus/imunologia , Lactobacillus/patogenicidade , Lacticaseibacillus casei/patogenicidade , Masculino , Pessoa de Meia-Idade
14.
Biomed Res Int ; 2014: 208974, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24955346

RESUMO

Lactobacilli are generally regarded as safe; however, certain strains have been associated with cases of infection. Our workgroup has already assessed many functional properties of Lactobacillus kefiri, but parameters regarding safety must be studied before calling them probiotics. In this work, safety aspects and antimicrobial activity of L. kefiri strains were studied. None of the L. kefiri strains tested caused α- or ß-hemolysis. All the strains were susceptible to tetracycline, clindamycin, streptomycin, ampicillin, erythromycin, kanamycin, and gentamicin; meanwhile, two strains were resistant to chloramphenicol. On the other hand, all L. kefiri strains were able to inhibit both Gram(+) and Gram(-) pathogens. Regarding the in vitro results, L. kefiri CIDCA 8348 was selected to perform in vivo studies. Mice treated daily with an oral dose of 10(8) CFU during 21 days showed no signs of pain, lethargy, dehydration, or diarrhea, and the histological studies were consistent with those findings. Moreover, no differences in proinflammatory cytokines secretion were observed between treated and control mice. No translocation of microorganisms to blood, spleen, or liver was observed. Regarding these findings, L. kefiri CIDCA 8348 is a microorganism isolated from a dairy product with a great potential as probiotic for human or animal use.


Assuntos
Produtos Fermentados do Leite/efeitos adversos , Lactobacillus/metabolismo , Probióticos/administração & dosagem , Animais , Produtos Fermentados do Leite/microbiologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Humanos , Lactobacillus/patogenicidade , Camundongos , Testes de Sensibilidade Microbiana , Probióticos/efeitos adversos
15.
J Dairy Res ; 81(1): 16-23, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24168928

RESUMO

We investigated the mucus-binding properties of aggregating and non-aggregating potentially probiotic strains of kefir-isolated Lactobacillus kefiri, using different substrates. All the strains were able to adhere to commercial gastric mucin (MUCIN) and extracted mucus from small intestine (SIM) and colon (CM). The extraction of surface proteins from bacteria using LiCl or NaOH significantly reduced the adhesion of three selected strains (CIDCA 8348, CIDCA 83115 and JCM 5818); although a significant proportion (up to 50%) of S-layer proteins were not completely eliminated after treatments. The surface (S-layer) protein extracts from all the strains of Lb. kefiri were capable of binding to MUCIN, SIM or CM, and no differences were observed among them. The addition of their own surface protein extract increased adhesion of CIDCA 8348 and 83115 to MUCIN and SIM, meanwhile no changes in adhesion were observed for JCM 5818. None of the seven sugars tested had the ability to inhibit the adhesion of whole bacteria to the three mucus extracts. Noteworthy, the degree of bacterial adhesion reached in the presence of their own surface protein (S-layer) extract decreased to basal levels in the presence of some sugars, suggesting an interaction between the added sugar and the surface proteins. In conclusion, the ability of these food-isolated bacteria to adhere to gastrointestinal mucus becomes an essential issue regarding the biotechnological potentiality of Lb. kefiri for the food industry.


Assuntos
Aderência Bacteriana , Mucosa Gástrica/microbiologia , Mucosa Intestinal/microbiologia , Lactobacillus/fisiologia , Muco/microbiologia , Probióticos , Animais , Aderência Bacteriana/efeitos dos fármacos , Colo , Produtos Fermentados do Leite/microbiologia , Hexoses/farmacologia , Intestino Delgado , Proteínas de Membrana/farmacologia , Suínos
16.
PLoS One ; 7(5): e36262, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606249

RESUMO

The human gastrointestinal tract is exposed to a huge variety of microorganisms, either commensal or pathogenic; at this site, a balance between immunity and immune tolerance is required. Intestinal dendritic cells (DCs) control the mechanisms of immune response/tolerance in the gut. In this paper we have identified a peptide (STp) secreted by Lactobacillus plantarum, characterized by the abundance of serine and threonine residues within its sequence. STp is encoded in one of the main extracellular proteins produced by such species, which includes some probiotic strains, and lacks cleavage sites for the major intestinal proteases. When studied in vitro, STp expanded the ongoing production of regulatory IL-10 in human intestinal DCs from healthy controls. STp-primed DC induced an immunoregulatory cytokine profile and skin-homing profile on stimulated T-cells. Our data suggest that some of the molecular dialogue between intestinal bacteria and DCs may be mediated by immunomodulatory peptides, encoded in larger extracellular proteins, secreted by commensal bacteria. These peptides may be used for the development of nutraceutical products for patients with IBD. In addition, this kind of peptides seem to be absent in the gut of inflammatory bowel disease patients, suggesting a potential role as biomarker of gut homeostasis.


Assuntos
Proteínas de Bactérias/imunologia , Células Dendríticas/imunologia , Intestinos/imunologia , Lactobacillus plantarum/imunologia , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sequência de Bases , Biomarcadores , DNA Bacteriano/genética , Humanos , Tolerância Imunológica , Técnicas In Vitro , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Intestinos/citologia , Lactobacillus plantarum/genética , Metagenoma/imunologia , Dados de Sequência Molecular , Probióticos
17.
Curr Microbiol ; 64(6): 592-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22461079

RESUMO

The aim of this study was to study the interference of the extracellular proteins produced by Lactobacillus plantarum BMCM12 with the adhesion of some well-known gut pathogens. The extracellular proteins secreted by L. plantarum BMCM12 in MRS broth were precipitated, resolved by SDS-PAGE, and identified by tandem mass spectrometry. Discordances between the observed and the theoretical molecular masses of several proteins suggested the presence of protein glycosylation, corroborated with specific glycoprotein staining after protein de-glycosylation using trifluoromethanesulfonic acid. Experiments of exclusion, competition, or prevention of the pathogen adhesion to mucin were performed using BMCM12 extracellular proteins, using Escherichia coli LMG2092 and Salmonella enterica subsp. enterica LMG15860. Extracellular proteins from BMCM12 reduced significantly the adhesion of the pathogens when they were added prior to adhesion assays. These proteins play thus important roles in preventing pathogen adhesion to the mucin layer.


Assuntos
Antibiose , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Escherichia coli/fisiologia , Lactobacillus plantarum/fisiologia , Mucinas/metabolismo , Salmonella enterica/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Lactobacillus plantarum/metabolismo , Peso Molecular , Espectrometria de Massas em Tandem
18.
FEMS Microbiol Lett ; 328(2): 166-73, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22224921

RESUMO

In the present work, the adhesion of 43 human lactobacilli isolates to mucin has been studied. The most adherent strains were selected, and their capacities to adhere to three epithelial cell lines were studied. All intestinal strains and one vaginal isolate adhered to HT-29 cells. The latter was the most adherent to Caco-2 cells, although two of the intestinal isolates were also highly adherent. Moreover, five of the eight strains strongly adhered to HeLa cells. The binding of an Actinomyces neuii clinical isolate to HeLa cells was enhanced by two of the lactobacilli and by their secreted proteins, while those of another two strains almost abolished it. None of the strains were able to interfere with the adhesion of Candida albicans to HeLa cells. The components of the extracellular proteome of all strains were identified by MALDI-TOF/MS. Among them, a collagen-binding A precursor and aggregation-promoting factor-like proteins are suggested to participate on adhesion to Caco-2 and HeLa cells, respectively. In this way, several proteins with LysM domains might explain the ability of some bacterial supernatants to block A. neuii adhesion to HeLa cell cultures. Finally, glyceraldehyde 3-phosphate dehydrogenase (GAPDH) could explain the good adhesion of some strains to mucin.


Assuntos
Aderência Bacteriana , Lactobacillus/fisiologia , Probióticos/administração & dosagem , Vagina/microbiologia , Actinomyces/fisiologia , Antibacterianos/química , Proteínas de Bactérias/química , Células CACO-2 , Candida albicans/fisiologia , Meios de Cultura/química , Feminino , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/química , Células HT29 , Células HeLa , Humanos , Mucinas/química , Ligação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
19.
FEMS Microbiol Lett ; 318(2): 101-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21323981

RESUMO

Bacillus cereus CH is a probiotic strain used in human nutrition whose adhesion to mucin is dependent on its surface-associated flagellin. Flagellins from the surface of several probiotic Bacillus strains were efficiently extracted with 5 M LiCl and identified by peptide fingerprinting. Based on the proteomic analysis, cloning of the gene coding for the flagellin of B. cereus CH was performed in the lactococcal vector pNZ8110 under the control of a nisin-inducible promoter. The resulting strain, Lactococcus lactis CH, produced a surface-associated flagellin after 6 h of induction with nisin. The recombinant Lactococcus strain adhered strongly to mucin-coated polystyrene plates, whilst inhibiting competitively the adhesion of the pathogens Escherichia coli LMG2092 and Salmonella enterica ssp. enterica LMG15860 to the same molecule. Strain CH could be used in further experimentation for the characterization of the molecular mechanism of action of this probiotic B. cereus CH flagellin.


Assuntos
Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Enterobacteriaceae/fisiologia , Flagelina/metabolismo , Lactococcus/fisiologia , Mucinas/metabolismo , Aderência Bacteriana , Escherichia coli/fisiologia , Flagelina/genética , Regulação Bacteriana da Expressão Gênica , Engenharia Genética , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Lactococcus/genética , Modelos Biológicos , Ligação Proteica , Salmonella enterica/fisiologia
20.
Appl Environ Microbiol ; 77(3): 1123-6, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21131525

RESUMO

In the present work, we describe the adhesion capabilities of a recombinant Lactococcus lactis strain producing an extracellular protein from Lactobacillus plantarum. Our results show that this protein may offer the bacterium a mechanism to bind to N-acetylglucosamine-containing polymers, such as human mucins, present in different environments.


Assuntos
Adesinas Bacterianas/metabolismo , Células CACO-2/metabolismo , Quitina/metabolismo , Lactobacillus plantarum/metabolismo , Mucinas/metabolismo , Ligação Proteica , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Lactobacillus plantarum/genética , Lactobacillus plantarum/fisiologia , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA