Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Immunobiology ; 226(1): 152028, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33242664

RESUMO

BACKGROUND AND AIMS: Pglyrp3 is a bactericidal innate immunity protein known to sustain the habitual gut microbiome and protect against experimental colitis. Intestinal inflammation and metaflammation are commonly associated with a marked reduction of commensal bifidobacteria. Whether Pglyrp3 and bifidobacteria interact synergistically or additively to alleviate metaflammation is unknown. We investigated the extent to which Pglyrp3 and bifidobacteria regulate metaflammation and gut bacterial dysbiosis in DSS-induced mouse models of intestinal inflammation. MATERIAL & METHODS: 8-10 weeks old male mice were used. In both WT and Pglyrp3 -/- experiments, the mice were randomly divided into three groups of 16 mice per group: (1) a control group receiving sterile tap water, (2) an experimental group receiving sterile tap water supplemented with only 5% DSS, and (3) an experimental group receiving sterile tap water supplemented with 5% DSS and 1 × 109 CFU/ml of Bifidobacterium adolescentis (B.a.) for 7 days. Wild-type (WT) littermates of the respective gene (i.e. Pglyrp3) were used as controls throughout the study. Clinical signs of general health and inflammation were monitored daily. Faecal pellet samples were analysed by qRT-PCR for microbial composition. Histology of relevant organs was carried out on day 8. Metabolic parameters and liver inflammation were determined in serum samples. RESULTS: Intestinal inflammation in mice of group 2 were significantly increased compared to those of control group 1. There was a significant difference in mean scores for inflammation severity between DSS-treated WT and DSS-treated Pglyrp3 -/- mice. Buildup of key serum metabolic markers (cholesterol, triglyceride and glucose) was set off by colonic inflammation. qRT-PCR quantification showed that DSS significantly decreased the Clostridium coccoides and Bifidobacterium cell counts while increasing those of Bacteroides group in both WT and Pglyrp3 -/- mice. These manifestations of DSS-induced dysbiosis were significantly attenuated by feeding B.a. Both the local and systemic ill-being of the mice alleviated when they received B.a. DISCUSSION: This study shows that Pglyrp3 facilitates recognition of bifidobacterial cell wall-derived peptidoglycan, thus leading additively to a reduction of metaflammation through an increase in the number of bifidobacteria, which were able to mitigate intestinal immunopathology in the context of Pglyrp3 blockade.


Assuntos
Bifidobacterium adolescentis/fisiologia , Proteínas de Transporte/metabolismo , Colite/metabolismo , Disbiose/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Animais , Terapia Biológica , Proteínas de Transporte/genética , Células Cultivadas , Colite/terapia , Sulfato de Dextrana , Modelos Animais de Doenças , Disbiose/terapia , Microbioma Gastrointestinal , Humanos , Doenças Inflamatórias Intestinais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
2.
Artigo em Inglês | MEDLINE | ID: mdl-33135616

RESUMO

BACKGROUND AND AIMS: Following a fat-rich diet, alterations in gut microbiota contribute to enhanced gut permeability, metabolic endotoxemia, and low grade inflammation-associated metabolic disorders. To better understand whether commensal bifidobacteria influence the expression of key metaflammation-related biomarkers (chemerin, MCP-1, PEDF) and modulate the pro-inflammatory bacteria- and lipid-coupled intracellular signaling pathways, we aimed at i) investigating the influence of the establishment of microbial signaling molecules-based cell-cell contacts on the involved intercellular communication between enterocytes, immune cells, and adipocytes, and ii) assessing their inflammatory mediators' expression profiles within an inflamed adipose tissue model. MATERIAL AND METHODS: Bifidobacterium animalis R101-8 and Escherichia coli TG1, respectively, were added to the apical side of a triple co-culture model consisting of intestinal epithelial HT-29/B6 cell line, human monocyte-derived macrophage cells, and adipose-derived stem cell line in the absence or presence of LPS or palmitic acid. mRNA expression levels of key lipid metabolism genes HILPDA, MCP-1/CCL2, RARRES2, SCD, SFRP2 and TLR4 were determined using TaqMan qRT-PCR. Protein expression levels of cytokines (IL-1ß, IL-6, and TNF-α), key metaflammation-related biomarkers including adipokines (chemerin and PEDF), chemokine (MCP- 1) as well as cellular triglycerides were assessed by cell-based ELISA, while those of p-ERK, p-JNK, p-p38, NF-κB, p-IκBα, pc-Fos, pc-Jun, and TLR4 were assessed by Western blotting. RESULTS: B. animalis R101-8 inhibited LPS- and palmitic acid-induced protein expression of inflammatory cytokines IL-1ß, IL-6, TNF-α concomitant with decreases in chemerin, MCP-1, PEDF, and cellular triglycerides, and blocked NF-kB and AP-1 activation pathway through inhibition of p- IκBα, pc-Jun, and pc-Fos phosphorylation. B. animalis R101-8 downregulated mRNA and protein levels of HILPDA, MCP-1/CCL2, RARRES2, SCD and SFRP2 and TLR4 following exposure to LPS and palmitic acid. CONCLUSION: B. animalis R101-8 improves biomarkers of metaflammation through at least two molecular/signaling mechanisms triggered by pro-inflammatory bacteria/lipids. First, B. animalis R101-8 modulates the coupled intracellular signaling pathways via metabolizing saturated fatty acids and reducing available bioactive palmitic acid. Second, it inhibits NF-kB's and AP-1's transcriptional activities, resulting in the reduction of pro-inflammatory markers. Thus, the molecular basis may be formed by which commensal bifidobacteria improve intrinsic cellular tolerance against excess pro-inflammatory lipids and participate in homeostatic regulation of metabolic processes in vivo.


Assuntos
Bifidobacterium animalis/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/fisiopatologia , Metabolismo dos Lipídeos/fisiologia , Transdução de Sinais/fisiologia , Biomarcadores , Linhagem Celular , Técnicas de Cocultura , Citocinas/biossíntese , Humanos , Metabolismo dos Lipídeos/genética , Lipopolissacarídeos/farmacologia
3.
Hepat Med ; 12: 93-106, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32617026

RESUMO

BACKGROUND AND PURPOSE: In previous investigations, Weissella confusa was shown to lack the metabolic pathway from fructose to mannitol and to produce ethanol when cultivated in the presence of fructose. Hence, we assessed the effect of oral administration of W. confusa (strain NRRL-B-14171) on blood and fecal ethanol concentrations, glucose and lipid metabolism and traits of the metabolic syndrome in Wistar rats (n=27) fed diets with two different fat and fructose levels and with or without the addition of W. confusa during a total intervention time of 15 weeks (105 days). MATERIALS AND METHODS: From week 1 to 6, rats were given a medium fructose and fat (MFru-MF) diet containing 28% fructose and 10% fat without the addition of W. confusa (control group, n=13) or mixed with 30 g per kg diet of lyophilized W. confusa (10.56 ± 0.20 log CFU/g; W. confusa group, n=14). From week 7 to 15, the percentage of dietary fructose and fat in the control and W. confusa group was increased to 56% and 16%, respectively (high fructose-high fat (HFru-HF) diet). RESULTS: In HFru-HF-fed rats, W. confusa was detected in feces, regardless of whether W. confusa was added to the diet or not, but not in rats receiving the MFru-MF diet without added W. confusa or in an additional control group (n=10) fed standard rat food without fructose, increased fat content and W. confusa. This indicates that fecal W. confusa may be derived from orally administered W. confusa as well as - in the case of high fructose and fat intake and obesity of rats - from the intestinal microbiota. As shown by multifactorial ANOVA, blood ethanol, the relative liver weight, serum triglycerides, and serum cholesterol as well as fecal ethanol, ADH, acetate, propionate and butyrate, but not lactate, were significantly higher in the W. confusa - compared to the control group. DISCUSSION: This is the first in vivo trial demonstrating that heterofermentative lactic acid bacteria lacking the mannitol pathway (like W. confusa) can increase fecal and blood ethanol concentrations in mammals on a high fructose-high fat diet. This may explain why W. confusa resulted in hyperlipidemia and may promote development of NAFLD in the host.

4.
Immunobiology ; 225(1): 151874, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31810825

RESUMO

Increased concentration of ferrous iron in the gastrointestinal tract increases the number of various pathogens and induces inflammation. LPS and/or high-fat diet-associated metaflammation is mediated through a quaternary receptor signaling complex containing iron-regulated pathway, IL-6/STAT inflammatory signaling pathway, hepcidin regulatory pathway, and common TLR4/NF-κB signaling pathway. We, therefore, investigated whether bifidobacteria directly or indirectly ameliorate LPS- and/or high-fat diet-associated metaflammation by reduction of intestinal iron concentration and/or the above-mentioned pathways. MATERIAL & METHODS: We used a triple co-culture model of HT-29/B6, HMDM and HepG2 cells with apically added Bifidobacterium pseudolongum (DSMZ 20099), in the absence or presence of iron, LPS or oleate. Expressions of the biomarkers of interest were determined after 24 h incubation by TaqMan qRT-PCR, cell-based ELISA or Western blot. RESULTS: Bifidobacteria inhibited LPS- and oleate-induced protein expression of inflammatory cytokines (IL-6, TNF-α) concomitantly with decreases in cellular TG and iron concentration. Exposure of co-cultured cells to bifidobacteria blocked NF-kB activity through inhibition of IκBα, p38 MAPK, and phosphorylation of NF-kB 65 subunit. TaqMan qRT-PCR and Western blot analysis revealed that bifidobacteria downregulated mRNA and protein expression of BMP6, DMT1, hepcidin, l-ferritin, ferroportin, IL-6, TfR1, Stat3, and TLR4 following exposure to excessive extracellular LPS, oleate and iron. However, the patterns of TLR2 mRNA and protein expression were quite the opposite of those of TLR4. CONCLUSION: Commensal bifidobacteria ameliorate metaflammation/inflammatory responses to excessive extracellular LPS, oleate and iron through at least two molecular/signaling mechanisms: i. modulation of interactions of the hepcidin- and iron-signaling pathways via reduction of excess iron; ii. reduction of pro-inflammatory cytokines and hepcidin production through inhibition of the TLR4/NF-kB pathway. This may be a molecular basis by which commensal bifidobacteria enhance intrinsic cellular tolerance against excess consumption of energy-yielding substrates and/or free iron.


Assuntos
Bifidobacterium/fisiologia , Hepcidinas/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/imunologia , Ferro/metabolismo , Obesidade/imunologia , Biomarcadores/metabolismo , Técnicas de Cocultura , Dieta Hiperlipídica , Células HT29 , Células Hep G2 , Humanos , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/metabolismo , NF-kappa B/metabolismo , Ácido Oleico/metabolismo , Transdução de Sinais , Simbiose , Receptor 4 Toll-Like/metabolismo
5.
Front Microbiol ; 7: 47, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26858714

RESUMO

To gain some specific insight into the roles microorganisms might play in non-alcoholic fatty liver disease (NAFLD), some intestinal and lactic acid bacteria and one yeast (Anaerostipes caccae, Bacteroides thetaiotaomicron, Bifidobacterium longum, Enterococcus fecalis, Escherichia coli, Lactobacillus acidophilus, Lactobacillus fermentum, Lactobacillus plantarum, Weissella confusa, Saccharomyces cerevisiae) were characterized by high performance liquid chromatography for production of ethanol when grown on different carbohydrates: hexoses (glucose and fructose), pentoses (arabinose and ribose), disaccharides (lactose and lactulose), and inulin. Highest amounts of ethanol were produced by S. cerevisiae, L. fermentum, and W. confusa on glucose and by S. cerevisiae and W. confusa on fructose. Due to mannitol-dehydrogenase expressed in L. fermentum, ethanol production on fructose was significantly (P < 0.05) reduced. Pyruvate and citrate, two potential electron acceptors for regeneration of NAD(+)/NADP(+), drastically reduced ethanol production with acetate produced instead in L. fermentum grown on glucose and W. confusa grown on glucose and fructose, respectively. In fecal slurries prepared from feces of four overweight volunteers, ethanol was found to be produced upon addition of fructose. Addition of A. caccae, L. acidophilus, L. fermentum, as well as citrate and pyruvate, respectively, abolished ethanol production. However, addition of W. confusa resulted in significantly (P < 0.05) increased production of ethanol. These results indicate that microorganisms like W. confusa, a hetero-fermentative, mannitol-dehydrogenase negative lactic acid bacterium, may promote NAFLD through ethanol produced from sugar fermentation, while other intestinal bacteria and homo- and hetero-fermentative but mannitol-dehydrogenase positive lactic acid bacteria may not promote NAFLD. Also, our studies indicate that dietary factors interfering with gastrointestinal microbiota and microbial metabolism may be important in preventing or promoting NAFLD.

6.
Clin Nutr ; 34(3): 394-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25042846

RESUMO

BACKGROUND & AIMS: Lactose digestion can be improved in subjects with impaired or completely absent intestinal lactase activity by administration of lactase preparations and particularly of acid lactase, which is active in the stomach, or by yogurt containing live lactic acid bacteria. It is the question, if lactose digestion can be further enhanced by combining these two approaches. METHODS: We investigated in a randomised, placebo-controlled, double-blind, 5-arm crossover study on 24 lactose malabsorbers with variable degrees of lactase deficiency if different lactase preparations and freeze-dried yogurt culture affect gastrointestinal lactose digestion after consuming moderate amounts of lactose (12.5 g) by assessing hydrogen exhalation over 6 h. Furthermore, symptoms of lactose intolerance (excess gas production, abdominal pain, diarrhoea or nausea) were assessed using validated questionnaires. RESULTS: All preparations increased lactose digestion and reduced peak hydrogen exhalation by -27% (yogurt), -29/-33% (3300/9000 FCC(1) ((1) One FCC hydrolyses about 5 or 1.7-2.5 mg lactose in aquous solution or in (artificial) chyme, respectively, according to the FCC-III method of the Committee on Codex Specifications, Food and Nutrition Board, National Research Council. Food Chemicals Codex, 3rd edition. Washington, DC, National Academy Press, 1981 It cannot precisely be defined how much lactose can be hydrolysed in vivo by the consumption of a certain number of FCC units.) units acid lactase from Aspergillus oryzae) or -46%, respectively (3300 FCC units lactase plus yogurt culture combined), as compared with placebo (p < 0.001, Friedman test). The combination preparation had not only the strongest effect, but also showed the lowest variance in H(2)-exhalation values (less malabsorbers with no reduction of H(2)-exhalation) Apart from this, both the higher dose lactase and the combination preparation significantly reduced the symptoms most closely associated with H(2)-exhalation, namely flatulences and abdominal pain, respectively. CONCLUSIONS: The combined administration of freeze-dried yogurt cultures and acid lactase increases lactose digestion more than either freeze-dried yogurt cultures or acid lactase alone, and more lactose malabsorbers benefited from this effect.


Assuntos
Aspergillus oryzae/enzimologia , Bactérias/enzimologia , Lactase/metabolismo , Intolerância à Lactose/terapia , Lactose/metabolismo , Iogurte/microbiologia , Adulto , Estudos Cross-Over , Digestão , Método Duplo-Cego , Feminino , Humanos , Hidrogênio/metabolismo , Hidrólise , Masculino , Adulto Jovem
7.
Immunobiology ; 219(3): 208-17, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24192538

RESUMO

BACKGROUND AND OBJECTIVES: We characterized the immunomodulating potential of a number of lactobacilli isolated from an African fermented food by co-incubation with peripheral blood mononuclear cells (PBMCs). Two strains with different immune modulating properties were genetically compared by suppression subtractive hybridization (SSH). METHODS: From 48 Lactobacillus strains isolated from Kimere, African fermented pearl millet dough, 10 were selected based on their bile salt tolerance. Their effects on secretion by PBMCs of the T-helper cells Th1- and Th2-cytokines IFN-γ and IL-4, respectively, in the presence or absence of staphylococcal enterotoxin A (SEA) were assessed. To study the genetic basis of different immune-modulating properties, a subtracted cDNA library for L. fermentum strains K1-Lb1 (Th1 inducer) and K8-Lb1 (Th1 and Th2 suppressor) was constructed using SSH. Finally, adhesion of these strains to hydrocarbons (relative hydrophobicity) and to human HT-29 colonic epithelial cell line was assessed. RESULTS: Two strains, K1-Lb1 and K4-Lb6, induced basal IFN-γ secretion. Four strains, K1-Lb6, K6-Lb2, K7-Lb1, and K8-Lb1 diminished INF-γ secretion by SEA-stimulated PBMCs. All strains, except K1-Lb1, K2-Lb4, and K9-Lb3, inhibited SEA-stimulated IL-4 secretion. Comparing the genomes of K1-Lb1 and K8-Lb1 by SSH indicated that K1-Lb1 is able to synthetize polysaccharides, for the synthesis of which K1-Lb8 appears to lack enzymes. A difference in the hydrophobicity properties of the surfaces of both strains indicated that this has impact on their surface. CONCLUSION: The K1-Lb1-specific sequences encoding putative glycosyltransferases and enzymes for polysaccharides synthesis may account for the observed differences in immunomodulation and surface properties between the two strains and for mediating potential probiotic effects.


Assuntos
Ductos Biliares/microbiologia , Lactobacillaceae/imunologia , Leucócitos Mononucleares/imunologia , Células Th1/imunologia , Células Th2/imunologia , Células Cultivadas , Sondas de DNA , Enterotoxinas/imunologia , Biblioteca Gênica , Genes Bacterianos/genética , Glicosiltransferases/genética , Humanos , Imunomodulação , Interferon gama/metabolismo , Interleucina-4/metabolismo , Lactobacillaceae/genética , Lactobacillaceae/isolamento & purificação , Leucócitos Mononucleares/microbiologia , Polissacarídeos Bacterianos/biossíntese , Probióticos , Equilíbrio Th1-Th2
8.
Nutr Res ; 33(1): 34-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23351408

RESUMO

Non-fasting triglyceridemia is much closer associated to cardiovascular risk compared to fasting triglyceridemia. We hypothesized that there would be acute differential effects of four common dietary proteins (cod protein, whey isolate, gluten, and casein) on postprandial lipemia in obese non-diabetic subjects. To test the hypothesis we conducted a randomized, acute clinical intervention study with crossover design. We supplemented a fat rich mixed meal with one of four dietary proteins i.e. cod protein, whey protein, gluten or casein. Eleven obese non-diabetic subjects (age: 40-68, body mass index: 30.3-42.0 kg/m(2)) participated and blood samples were drawn in the 8-h postprandial period. Supplementation of a fat rich mixed meal with whey protein caused lower postprandial lipemia (P = .048) compared to supplementation with cod protein and gluten. This was primarily due to lower triglyceride concentration in the chylomicron rich fraction (P = .0293). Thus, we have demonstrated acute differential effects on postprandial metabolism of four dietary proteins supplemented to a fat rich mixed meal in obese non-diabetic subjects. Supplementation with whey protein caused lower postprandial lipemia compared to supplementation with cod and gluten. As postprandial lipemia is closely correlated to cardiovascular disease, long-term dietary supplementation with whey protein may prove beneficial in preventing cardiovascular disease in obese non-diabetic subjects.


Assuntos
Proteínas Alimentares/administração & dosagem , Hiperlipidemias/sangue , Obesidade/sangue , Período Pós-Prandial/efeitos dos fármacos , Adulto , Idoso , Glicemia/análise , Índice de Massa Corporal , Doenças Cardiovasculares/prevenção & controle , Caseínas/administração & dosagem , Estudos Cross-Over , Diabetes Mellitus , Ácidos Graxos não Esterificados/sangue , Feminino , Glutens/administração & dosagem , Humanos , Hiperlipidemias/etiologia , Estilo de Vida , Masculino , Refeições , Pessoa de Meia-Idade , Proteínas do Leite/administração & dosagem , Obesidade/complicações , Triglicerídeos/sangue , Proteínas do Soro do Leite
9.
J Leukoc Biol ; 92(4): 895-911, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22730546

RESUMO

The pathophysiology of IBD is characterized by a complex interaction between genes and the environment. Genetic and environmental differences are attributed to the heterogeneity of the disease pathway and to the epigenetic modifications that lead to altered gene expression in the diseased tissues. The epigenetic machinery consists of short interfering RNA, histone modifications, and DNA methylation. We evaluated the effects of Bifidobacterium breve (DSMZ 20213) and LGG (ATCC 53103), as representatives of commensal probiotics on the expression of IL-17 and IL-23, which play an important role in IBD, and on the epigenetic machinery in a 3D coculture model composed of human intestinal HT-29/B6 or T84 cells and PBMCs. The cells were treated with LPS in the presence or absence of bacteria for 48 h, and the expression of IL-17, IL-23, and CD40 at the mRNA and protein levels was assessed using TaqMan qRT-PCR and ELISA, respectively. Western blotting was used to assess the expression of the MyD88, the degradation of IRAK-1 and IκBα, the expression of the NF-κB p50/p65 subunits, the p-p38 MAPK and p-MEK1, as well as histone modifications. NF-κB activity was assessed by NF-κB-dependent luciferase reporter gene assays. The accumulation of Ac-H4 and DNA methylation was quantitatively assessed using colorimetric assays. B. breve and LGG diminished the LPS-induced expression of IL-17, IL-23, CD40, and histone acetylation, while slightly enhancing DNA methylation. These effects were paralleled by a decrease in the nuclear translocation of NF-κB, as demonstrated by a decrease in the expression of MyD88, degradation of IRAK-1 and IκBα expression of the nuclear NF-κB p50/p65 subunits, p-p38 MAPK and p-MEK1, and NF-κB-dependent luciferase reporter gene activity in LPS-stimulated cells. B. breve and LGG may exert their anti-inflammatory effects in the gut by down-regulating the expression of the IBD-causing factors (IL-23/IL-17/CD40) associated with epigenetic processes involving the inhibition of histone acetylation and the optimal enhancement of DNA methylation, reflected in the limited access of NF-κB to gene promoters and reduced NF-κB-mediated transcriptional activation. We describe a new regulatory mechanism in which commensal probiotics inhibit the NF-κB-mediated transcriptional activation of IBD-causing factors (IL-23/IL-17/CD40), thereby simultaneously reducing histone acetylation and enhancing DNA methylation.


Assuntos
Epigênese Genética , Interleucina-17/antagonistas & inibidores , Interleucina-23/antagonistas & inibidores , Mucosa Intestinal/imunologia , Probióticos/farmacologia , Bifidobacterium , Proliferação de Células , Técnicas de Cocultura , Metilação de DNA , Células HT29 , Histonas/metabolismo , Humanos , Imunidade nas Mucosas , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/fisiologia
10.
Immunobiology ; 217(4): 412-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22099350

RESUMO

Intestinal epithelial cells produce cytokines in response to bacterial peptidoglycan (PGN), which is detected by several classes of pattern-recognition receptors (PRRs) as peptidoglycan recognition proteins (PGlyRPs), Toll-like receptor 2 (TLR2) and NOD receptors. All types of PGlyRPs recognize bacterial peptidoglycan and function in antibacterial innate immunity. In this study, we investigated the role of PGlyRP3 in the response of intestinal epithelial cells (Caco-2) to PGN from pathogenic (Staphylococcus aureus), opportunistic pathogenic (Micrococcus luteus) and non-pathogenic (Bacillus subtilis and Lactobacillus rhamnosus GG) bacteria found in the gut as commensals or in gastroenteritis. All PGNs induced the proinflammatory cytokines IL-12p35, IL-8 and TNF-α and, time-dependently, PGlyRP3, at both the transcription and protein levels. In this context, no differences were observed among the distinct PGN obtained from different bacterial sources. The inflammatory response to PGN is mediated via the TLR2 pathway, since blocking this pathway by inhibiting MyD88 reduced the expression of proinflammatory cytokines. In addition, PGlyRP3 overexpression suppressed, while PGlyRP3 knocking down enhanced the expression of PGN-induced inflammatory cytokines. It is concluded that PGN stimulates inflammatory responses in the intestinal epithelia through activation of the TLR pathway. PGlyRP3 is also stimulated by PGN and has, in contrast to activation of the TLR pathway, an anti-inflammatory effect.


Assuntos
Anti-Inflamatórios/metabolismo , Bacillus subtilis/metabolismo , Proteínas de Transporte/metabolismo , Gastroenterite/imunologia , Mucosa Intestinal/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Micrococcus luteus/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/metabolismo , Anti-Inflamatórios/imunologia , Bacillus subtilis/imunologia , Células CACO-2 , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Gastroenterite/microbiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Lacticaseibacillus rhamnosus/imunologia , Micrococcus luteus/imunologia , Peptidoglicano/imunologia , RNA Interferente Pequeno/genética , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/metabolismo , Transgenes/genética
11.
J Dairy Res ; 78(4): 396-403, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21871144

RESUMO

To investigate matrix-specifity of probiotic effects and particularly of the reduction of antibiotics-associated diarrhea, a controlled, randomized, double-blind study was performed, in which 88 Helicobacter pylori-infected but otherwise healthy subjects were given for eight weeks either a) a probiotic fruit yoghurt "mild" containing Lactobacillus acidophilus LA-5 plus Bifidobacterium lactis BB-12, n = 30), b) the same product but pasteurized after fermentation (n = 29) or c) milk acidified with lactic acid (control, n = 29). During week five, a Helicobacter eradication therapy was performed. Helicobacter activity was measured via 13C-2-urea breath tests and antibiotic-associated diarrhoea and other gastrointestinal complaints were recorded by validated questionnaires. In intervention group a, b and c the mean number of days with diarrhoea was 4, 10 and 10 (P<0·05), the frequency of episodes 17%, 7% and 27% (n.s.), and the change in total symptoms score before antibiotics treatment was -1·4 ± 1·1, -1·2 ± 1·1, 2·6 ± 1·1 points/four weeks (P<0·05). All milk products decreased Helicobacter activity by 18 to 45% without significant differences between groups. The observed decrease in Hel. pylori activity seems to be not or not only due to probiotic bacteria but (rather) to components of acidified milk (most probably lactic acid). Fruit-yogurt-like fermented milk products with living probiotic bacteria significantly shorten the duration of antibiotics-associated diarrhoea and improve gastrointestinal complaints. Fruit yogurt-like fermented milk is a matrix suitable for probiotic bacteria.


Assuntos
Bifidobacterium , Produtos Fermentados do Leite/microbiologia , Diarreia/prevenção & controle , Infecções por Helicobacter/tratamento farmacológico , Lactobacillus acidophilus , Probióticos/uso terapêutico , Adulto , Amoxicilina/administração & dosagem , Amoxicilina/efeitos adversos , Testes Respiratórios , Claritromicina/administração & dosagem , Claritromicina/efeitos adversos , Diarreia/induzido quimicamente , Método Duplo-Cego , Feminino , Frutas , Gastroenteropatias/microbiologia , Gastroenteropatias/terapia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Ureia/análise
12.
Mol Nutr Food Res ; 55(10): 1533-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21710560

RESUMO

SCOPE: Human ß-defensin 2 (hBD-2) is an inducible antimicrobial peptide synthesized by the epithelium to counteract bacterial adherence and invasion. It has been suggested that probiotic bacteria sustain gut barrier function via induction of defensins. The goals of this study were (i) to evaluate the potential immunomodulatory effects of 11 different Lactobacillus fermentum strains isolated from Kimere, an African fermented pearl millet (Pennisetum glaucum) dough, on the hBD-2 secretion by human intestinal CaCo-2 cell line and (ii) to examine genetic differences between two strains of L. fermentum (K2-Lb4 and K11-Lb3) which differed in their effect on the production of hBD-2 in this study. METHODS AND RESULTS: Totally, 46 strains of L. fermentum from Kimere were isolated and characterized using molecular biology methods including pulsed-field gel electrophoresis patterns. After performing time- and dose-experiments, CaCo-2 cells were incubated with or without bacteria for 12 h. L. fermentum PZ1162 was included as the positive control. Cell-free supernatants were analyzed for hBD-2 protein by enzyme-linked immunosorbent assay (ELISA). To identify potential bacterial genes associated with hBD-2 regulation, suppression subtractive hybridization (SSH) was used. Among the 11 strains tested, only two strains of bacteria, K11-Lb3 and K2-Lb6, significantly induced the production of hBD-2 by CaCo-2 cells. This effect was strain-specific, dose-dependent and particularly seems to be bacterial genomic-dependent as manifested by SSH. L. fermentum strains with and without hBD-2 inducing effect differed in genes encoding proteins involved in glycosylation of cell-wall proteins e.g. glycosyltransferase, UDP-N-acetylglucosamine 2-epimerase, rod shape-determining protein MreC, lipoprotein precursors, sugar ABC transporters, and glutamine ABC transporter ATP-binding protein. CONCLUSION: This study implies that certain strains of L. fermentum isolated from Kimere may stimulate the intestinal innate defense through the induction of hBD-2. The molecular basis of hBD-2 induction by L. fermentum strain K11-Lb3 may be based on glycosylated cell-surface structures synthesized with the aid of glycosyltransferase, UDP-N-acetylglucosamine 2-epimerase, and rod shape-determining protein MreC.


Assuntos
Enterócitos/metabolismo , Enterócitos/microbiologia , Limosilactobacillus fermentum/genética , beta-Defensinas/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Células CACO-2 , Carboidratos Epimerases/genética , Carboidratos Epimerases/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Genoma Bacteriano , Glicosilação , Humanos , Hibridização In Situ/métodos , Dados de Sequência Molecular , Pennisetum/microbiologia , Especificidade da Espécie , beta-Defensinas/genética
13.
J Nutr ; 141(5): 971-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21451128

RESUMO

Prebiotic oligosaccharides modulate the intestinal microbiota and beneficially affect the human body by reducing intestinal inflammation. This immunomodulatory effect was assumed to be bacterial in origin. However, some observations suggest that oligosaccharides may exert an antiinflammatory effect per se. We hypothesized that oligosaccharides affect the intestinal immunity via activation of peptidoglycan recognition protein 3 (PGlyRP3), which reduces the expression of proinflammatory cytokines. Caco-2 cells were treated with the oligosaccharides, α3-sialyllactose, or fructooligosaccharides (Raftilose p95), and the effects of these treatments on PGlyRP3 and PPARγ expression, the release and expression of some proinflammatory cytokines, and NF-κB translocation were tested. Both oligosaccharides had antiinflammatory activity; they significantly reduced IL-12 secretion in Caco-2 cells and gene expression of IL-12p35, IL-8, and TNFα. They also reduced the gene expression and nuclear translocation of NF-κB. Both oligosaccharides dose and time dependently induced the production of PGlyRP3, the silencing of which by transfection of Caco-2 cells with specific small interfering RNA targeting PGlyRP3 abolished the antiinflammatory role of both oligosaccharides. Incubation of Caco-2 cells with both oligosaccharides induced PPARγ. Antagonizing PPARγ by culturing the cells with GW9662 for 24 h inhibited the oligosaccharide-induced PGlyRP3 production and the antiinflammatory effect of the oligosaccharides. We conclude that oligosaccharides may exert an antiinflammatory effect by inducing the nuclear receptor PPARγ, which regulates the antiinflammatory PGlyRP3.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Mucosa Intestinal/metabolismo , Oligossacarídeos/metabolismo , PPAR gama/metabolismo , Prebióticos , Células CACO-2 , Proteínas de Transporte/genética , Núcleo Celular/metabolismo , Citocinas/genética , Regulação para Baixo/efeitos dos fármacos , Inativação Gênica , Humanos , Imunomodulação , Mucosa Intestinal/imunologia , Inulina/administração & dosagem , Inulina/análogos & derivados , Oligossacarídeos/administração & dosagem , PPAR gama/antagonistas & inibidores , Transporte Proteico , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição RelA/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
J Nutr ; 140(8): 1411-7, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20534879

RESUMO

The intestinal fatty acid binding protein (FABP2) is involved in lipid metabolism whereby variations in the promoter (haplotypes A/B) and exon 2 (Ala54Thr) are associated with dyslipidemia and insulin resistance. To elucidate which factors determine FABP2 expression in human mucosa, we investigated the association between fat intake, genotypes, biochemical variables, and FABP2 expression. FABP2 gene expression was assessed in duodenal specimens from 100 participants who answered a FFQ and who were genotyped and characterized for traits of metabolic syndrome and further biochemical data. Homozygotes for haplotype A tended to have lower fat intake than B-allele carriers (P = 0.066). Searching for an explanation, we evaluated the orexigenic glucose-dependent insulinotropic polypeptide (GIP) in a subset from the Metabolic Intervention Cohort Kiel. AA homozygotes had lower postprandial GIP concentrations than BB homozygotes. Duodenal FABP2 expression was correlated with (n-3) fatty acid (FA) intake in AA homozygotes (r = 0.49; P = 0.021). It was higher in AA homozygotes than in B-allele carriers after adjustment for (n-3) FA intake (P = 0.049) and was negatively correlated with serum FFA (r = -0.41; P < 0.01). Our data indicate that FABP2 expression depends on (n-3) FA intake and FABP2 genotypes. FABP2 might be involved in regulating food intake and intestinal FA utilization.


Assuntos
Gorduras na Dieta/administração & dosagem , Proteínas de Ligação a Ácido Graxo/genética , Polimorfismo Genético/genética , Adulto , Idoso , Células CACO-2 , Colo/química , DNA/análise , Duodeno/química , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Genótipo , Humanos , Íleo/química , Mucosa Intestinal/química , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
15.
Int Immunopharmacol ; 10(6): 694-706, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20381647

RESUMO

BACKGROUND AND OBJECTIVES: Control of the intracellular Mycobacterium tuberculosis (Mtb), mainly requires an appropriate ratio of Th1/Th2 cytokines to induce autophagy, a physiologically, and immunologically regulated process that has recently been highlighted as an innate defense mechanism against intracellular pathogens. Current vaccines/adjuvants induce both protective Th1 autophagy-promoting cytokines, such as IFN-gamma, and immunosuppressive Th2 autophagy-restraining cytokines, such as IL-4 and IL-13. TB infection itself is also characterized by relatively high levels of Th2 cytokines, which down-regulate Th1 responses and subsequently subvert adequate protective immunity, and a low ratio of IFN-gamma/IL-4. Therefore, there is a need for a safe and non-toxic vaccine/adjuvant that will induce Th1 autophagy-promoting cytokine (IFN-gamma) secretion and suppress the pre-existing subversive Th2 autophagy-restraining cytokines (IL-4 and IL-13). As lactic acid bacteria (LAB) belonging to the natural intestinal microflora and their components have been shown to shift immune responses against other antigens from Th2-type cytokines toward Th1-type cytokines like IFN-gamma, we investigated whether LAB can improve the polarization of Th1/Th2 cytokines and autophagic ability of mononuclear phagocytes in response to Mtb antigen. METHODS: Peripheral blood mononuclear cells (PBMCs), which are a part of the mononuclear phagocyte system and source of crucial macrophage activators in the in vivo situation, and human monocyte-derived macrophages (HMDMs) were treated with Mtb antigen in the presence or absence of two strains of LAB, L. rhammosus GG (LGG) and Bifidobacterium bifidum MF 20/5 (B.b). PBMCs cell culture supernatants were analyzed for the production of the autophagy-promoting factors IFN-gamma, and nitric oxide (NO) and the autophagy-restraining cytokines IL-4 and IL-13, using ELISA and Griess assays to detect the production of cytokines and NO, respectively. In HMDMs, expression of microtubule-associated protein 1 light chain 3 (LC3-I), membrane-associated (LC3-II) forms of LC3 protein and Beclin-1, as hallmarks of autophagy, were assessed using Western blot to detect the autophagy markers. The secreted interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin (IL)-12 and transformig growth factor-beta (TGF-beta), and chemokine (C-C motif) ligand 18 (CCL18) from HMDMs were determined by ELISA. Also, reverse transcription polymerase chain reaction (RT-PCR) analysis was used to assess the mRNA expressions of CCL18 in HMDMs. RESULTS: Treatment of PBMCs with either Mtb antigen or with LAB significantly increased the IFN-gamma and NO production. Combination of Mtb antigen and LAB led to synergistic increase in IFN-gamma, and an additive increase in NO. Treatment with Mtb antigen alone significantly increased the IL-4 and IL-13 production. LAB significantly decreased IL-4 and IL-13 secretion in both unstimulated and Mtb antigen-stimulated PBMCs. The IFN-gamma/IL-4+IL-13 ratio was enhanced, indicating Th1/Th2 polarization. Treatment of macrophages with combined use of Mtb antigen and LAB led to an additive increase in Beclin-1, LC3-II expression, as well as in synergistic increase in IL-12 production. Treatment of macrophages with combined use of Mtb antigen and LAB led to a decrease in IL-6, IL-10, and CCL18 secretion. LAB inhibited the secretion of TGF-beta by Mtb-stimulated macrophages, however not significantly. Treatment of macrophages with combined use of Mtb antigen and LAB led to a decrease in CCL18 mRNA expression. CONCLUSION: Our study implies that LAB may reinforce the response of the mononuclear phagocytes to Mtb antigen by inducing production of the autophagy-promoting factors IFN-gamma and NO, while decreasing the Th2 autophagy-restraining cytokines IL-4 and IL-13. Hence, combination of Mtb antigen and LAB may perhaps be safer in more efficacious TB vaccine formulation.


Assuntos
Antígenos de Bactérias/imunologia , Autofagia/imunologia , Bifidobacterium/imunologia , Interferon gama/imunologia , Lacticaseibacillus rhamnosus/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Adulto , Bifidobacterium/metabolismo , Feminino , Humanos , Interleucina-13/análise , Interleucina-13/metabolismo , Interleucina-4/análise , Interleucina-4/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Macrófagos/microbiologia , Masculino , Proteínas Associadas aos Microtúbulos/imunologia , Monócitos/imunologia , Óxido Nítrico/análise , Óxido Nítrico/metabolismo , Células Th1/imunologia , Células Th2/imunologia , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
16.
Menopause Int ; 15(1): 35-40, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19237621

RESUMO

Among the numerous positive effects of probiotic microorganisms and prebiotic carbohydrates observed in clinical studies--the majority of which, however, does not fulfil the criteria of pharmaceutical verification--some are of specific relevance to female health. The present review addresses--besides some notes concerning the potential microbiota-hormone interactions--the first line with preventive and/or therapeutic applications of probiotic bacteria in order to maintain a balanced intestinal and urogenital flora, as well as in the case of irritable bowel syndrome, constipation (idiopathic slow-transit) and urogenital tract infections. Further aspects are the promotion of bone health and osteoporosis prevention brought about by inulin, oligofructose and galactooligosaccharides. Some further conditions, namely anorexia nervosa, the premenstrual syndrome as well as prevention or alleviation of climacteric and menopausal disorders, for which the use of probiotics is rather hypothetical or is largely studied by alternative medicine practising physicians, are addressed briefly.


Assuntos
Probióticos/uso terapêutico , Feminino , Gastroenteropatias/tratamento farmacológico , Humanos , Osteoporose Pós-Menopausa/tratamento farmacológico , Perimenopausa , Síndrome Pré-Menstrual/tratamento farmacológico , Ensaios Clínicos Controlados Aleatórios como Assunto , Infecções Urinárias/tratamento farmacológico
17.
J Pediatr Gastroenterol Nutr ; 48(1): 82-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19172129

RESUMO

OBJECTIVES: The primary objective of this study was to determine the bifidogenic effect of galacto-oligosaccharides (GOS) in a follow-on formula and the effects on other intestinal bacteria. Secondary objectives were the effects on stool characteristics, growth, and general well-being. PARTICIPANTS AND METHODS: In a multicenter, double-blind study, 159 healthy infants, formula-fed at enrollment (at 4-6 months), were randomized to an experimental follow-on formula supplemented with 5 g/L (GOS) (77 infants), or to a standard follow-on formula (control, 82 infants). Infants were evaluated at enrollment (study day 1 = sd1), after 6 weeks (study day 2 = sd2), and after an additional 12 weeks (study day 3 = sd3). At each study day, a fresh stool sample for the bacterial counts was collected, and the growth parameters were measured. At sd2, urinary specimens were collected for the evaluation of urinary osmolarity. RESULTS: At sd2 and sd3, the GOS group had a higher median number (colony-forming units per gram of stool) of bifidobacteria than did the control group (sd2 GOS 9.2 x 10(9) vs control 4.4 x 10(9), P = 0.012); (sd3 GOS 7.2 x 10(9) vs control 2.4 x 10(9), P = 0.027). Other bacteria did not show any significant differences between the 2 groups at all study days. The GOS produced softer stools but had no effect on stool frequency. The urinary osmolarity (mOsm/L) at sd2 was comparable in both groups. Supplementation had no influence on the incidence of gastrointestinal side effects or on the growth of the infants. CONCLUSIONS: These data indicate that the addition of GOS (5 g/L) to a follow-on formula positively influences the bifidobacteria flora and the stool consistency in infants during the supplementation period at weaning. No local or systemic side effects were recorded.


Assuntos
Bifidobacterium/crescimento & desenvolvimento , Galactose/administração & dosagem , Fórmulas Infantis/administração & dosagem , Oligossacarídeos/administração & dosagem , Bifidobacterium/efeitos dos fármacos , Contagem de Colônia Microbiana , Método Duplo-Cego , Fezes/microbiologia , Feminino , Galactose/efeitos adversos , Humanos , Lactente , Intestinos/microbiologia , Masculino , Oligossacarídeos/efeitos adversos , Concentração Osmolar , Placebos , Urina , Desmame
18.
Eur J Nutr ; 48(1): 45-53, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19030908

RESUMO

BACKGROUND: Milk products are good sources of calcium and their consumption may reduce bone resorption and thus contribute to prevent bone loss. AIM OF THE STUDY: We tested the hypothesis that bedtime consumption of fermented milk supplemented with calcium inhibits the nocturnally enhanced bone resorption more markedly than fermented milk alone, and postulated that this effect was most pronounced when calcium absorption enhancers were added. METHODS: In a controlled, parallel, double-blind intervention study over 2 weeks we investigated the short-term effects of two fermented milks supplemented with calcium from milk minerals (f-milk + Ca, n = 28) or calcium from milk minerals, inulin-type fructans and caseinphosphopeptides (f-milk + Ca + ITF + CPP; n = 29) on calcium and bone metabolism in healthy, postmenopausal women, and compared them with the effect of a fermented control milk without supplements (f-milk, n = 28). At bedtime 175 ml/d of either test milk was consumed. Fasting blood samples and 48 h-urine were collected at baseline and at the end of the intervention. Urine was divided into a pooled daytime and nighttime fraction. Multifactorial ANOVA was performed. RESULTS: Fermented milk independent of a supplement (n = 85) reduced the nocturnal excretion of deoxypyridinoline, a marker of bone resorption, from 11.73 +/- 0.54 before to 9.57 +/- 0.54 micromol/mol creatinine at the end of the intervention (P = 0.005). No effect was seen in the daytime fraction. Differences between the three milks (n = 28 resp. 29) were not significant. Fermented milk reduced bone alkaline phosphatase, a marker of bone formation, from 25.03 +/- 2.08 to 18.96 +/- 2.08 U/l, with no difference between these groups either. Fermented milk increased the nocturnal but not daytime urinary excretion of calcium and phosphorus. The effects on calcium and phosphorus excretion were mainly due to the group supplemented with Ca + ITF + CPP. CONCLUSION: Bedtime consumption of fermented milk reduced the nocturnal bone resorption by decelerating its turnover. Supplemented calcium from milk mineral had no additional effect unless the absorption enhancers ITF + CPP were added. A stimulated intestinal calcium absorption may be assumed, since urinary calcium excretion increased at a constant bone resorption.


Assuntos
Osso e Ossos/metabolismo , Cálcio da Dieta/administração & dosagem , Caseínas/administração & dosagem , Produtos Fermentados do Leite , Frutanos/administração & dosagem , Fosfopeptídeos/administração & dosagem , Idoso , Cálcio/sangue , Cálcio/urina , Ritmo Circadiano , Creatinina/urina , Registros de Dieta , Método Duplo-Cego , Comportamento Alimentar , Feminino , Alimentos Fortificados , Humanos , Inulina , Pessoa de Meia-Idade , Fósforo/sangue , Fósforo/urina , Pós-Menopausa , Inquéritos e Questionários
19.
Immunobiology ; 213(8): 677-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18950596

RESUMO

Among the factors potentially involved in the increased prevalence of allergic diseases, modification of the intestinal flora or lack of microbial exposure during childhood has been proposed. T(H)2-cytokines increase the production of IgE and stimulate mast cells and eosinophils, whereas T(H)1-cytokines, such as IFN-gamma, may suppress IgE synthesis and stimulate the expression of the secretory piece of IgA. Thus, a dysregulation in the expression of T(H)1- and T(H)2-cytokines may contribute to the initiation and maintenance of allergic diseases. Lactobacilli belonging to the natural intestinal microflora were reported to reduce the incidence of atopic dermatitis and the severity of allergic manifestations and to modulate T(H)1/T(H)2 responses. The mechanisms still remain to be elucidated. We sought to assess the effect of different probiotics, Lactobacillus rhamnosus GG, Lactobacillus gasseri (PA16/8), Bifidobacterium bifidum (MP20/5), and Bifidobacterium longum (SP07/3), on the T(H)1 and T(H)2 responses of peripheral blood mononuclear cells (PBMCs) from healthy subjects and from patients with allergy against house dust mite to Staphylococcus enterotoxin A (SEA) and Dermatophagoides pteronyssinus (Dpt). To elucidate the molecular basis of these effects, the effects of bacterial genomic DNA were compared with the effects of viable bacteria. PBMCs from allergic patients and from healthy donors were incubated for 24 or 48 h, respectively, with or without SEA and Dpt allergens. The effects of preincubation with live probiotic bacteria and the effect of their genomic DNA, added simultaneously to cultures and incubated for 24h, were assessed by measuring T(H)1/T(H)2-cytokine production. The tested live Gram-positive probiotic bacteria and their genomic DNA inhibited SEA- and Dpt-stimulated secretion of T(H)2-cytokines (IL-4 and IL-5) and enhanced the stimulation of IFN-gamma. This effect was dose-dependent with a dosage-optimum, which was identical for all lactic acid producing bacteria (LAB) tested (10 bacteria per PBMC) and their DNA (75 ng/ml). Based on the maximal effects achieved with LAB and their DNA, more than 50% of the effects seem to be contributed by DNA. No significant effect was induced by the control, Gram-negative Escherichia coli TG1. Lactobacilli and bifidobacteria reduced SEA-stimulated IL-4 and IL-5 production more effectively in PBMCs from healthy subjects than from allergic patients. In contrast to this, inhibition of Dpt-stimulated IL-4- and IL-5-secretion was more pronounced in cells from allergic subjects. Compared with living LAB, bacterial DNA inhibited IL-4- and IL-5-secretion in a similar manner. SEA- and even more so Dpt-stimulated IFN-gamma stimulation by living LAB was less pronounced in allergic than in healthy subjects, whereas IFN-gamma stimulation by their DNA was more pronounced in allergic subjects. The tested probiotic bacteria as well as their genomic DNA modulated the T(H)1/T(H)2 response to some allergens dose-dependently. DNA seems to contribute to 50% of the effect exerted by living bacteria in this in vitro model. The magnitude of the probiotic effects differed between healthy and allergic subjects. Whether the modulation found for the tested strains might be useful for the prevention and treatment of allergic diseases has to be assessed in clinical trials.


Assuntos
Bifidobacterium , DNA Bacteriano/imunologia , Hipersensibilidade/imunologia , Lacticaseibacillus rhamnosus , Probióticos , Células Th1/imunologia , Células Th2/imunologia , Adulto , Antígenos de Dermatophagoides/imunologia , Citocinas/metabolismo , Relação Dose-Resposta Imunológica , Enterotoxinas/imunologia , Feminino , Humanos , Hipersensibilidade/sangue , Ativação Linfocitária , Masculino , Células Th1/metabolismo , Células Th1/microbiologia , Células Th2/metabolismo , Células Th2/microbiologia
20.
Adv Biochem Eng Biotechnol ; 111: 1-66, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18461293

RESUMO

According to the German definition, probiotics are defined viable microorganisms, sufficient amounts of which reach the intestine in an active state and thus exert positive health effects. Numerous probiotic microorganisms (e.g. Lactobacillus rhamnosus GG, L. reuteri, bifidobacteria and certain strains of L. casei or the L. acidophilus-group) are used in probiotic food, particularly fermented milk products, or have been investigated--as well as Escherichia coli strain Nissle 1917, certain enterococci (Enterococcus faecium SF68) and the probiotic yeast Saccharomyces boulardii--with regard to their medicinal use. Among the numerous purported health benefits attributed to probiotic bacteria, the (transient) modulation of the intestinal microflora of the host and the capacity to interact with the immune system directly or mediated by the autochthonous microflora, are basic mechanisms. They are supported by an increasing number of in vitro and in vivo experiments using conventional and molecular biologic methods. In addition to these, a limited number of randomized, well-controlled human intervention trials have been reported. Well-established probiotic effects are: 1. Prevention and/or reduction of duration and complaints of rotavirus-induced or antibiotic-associated diarrhea as well as alleviation of complaints due to lactose intolerance. 2. Reduction of the concentration of cancer-promoting enzymes and/or putrefactive (bacterial) metabolites in the gut. 3. Prevention and alleviation of unspecific and irregular complaints of the gastrointestinal tracts in healthy people. 4. Beneficial effects on microbial aberrancies, inflammation and other complaints in connection with: inflammatory diseases of the gastrointestinal tract, Helicobacter pylori infection or bacterial overgrowth. 5. Normalization of passing stool and stool consistency in subjects suffering from obstipation or an irritable colon. 6. Prevention or alleviation of allergies and atopic diseases in infants. 7. Prevention of respiratory tract infections (common cold, influenza) and other infectious diseases as well as treatment of urogenital infections. Insufficient or at most preliminary evidence exists with respect to cancer prevention, a so-called hypocholesterolemic effect, improvement of the mouth flora and caries prevention or prevention or therapy of ischemic heart diseases or amelioration of autoimmune diseases (e.g. arthritis). A prebiotic is "a selectively fermented ingredient that allows specific changes, both in the composition and/or activity in the gastrointestinal microflora that confers benefits upon host well being and health", whereas synergistic combinations of pro- and prebiotics are called synbiotics. Today, only bifidogenic, non-digestible oligosaccharides (particularly inulin, its hydrolysis product oligofructose, and (trans)galactooligosaccharides), fulfill all the criteria for prebiotic classification. They are dietary fibers with a well-established positive impact on the intestinal microflora. Other health effects of prebiotics (prevention of diarrhoea or obstipation, modulation of the metabolism of the intestinal flora, cancer prevention, positive effects on lipid metabolism, stimulation of mineral adsorption and immunomodulatory properties) are indirect, i.e. mediated by the intestinal microflora, and therefore less-well proven. In the last years, successful attempts have been reported to make infant formula more breast milk-like by the addition of fructo- and (primarily) galactooligosaccharides.


Assuntos
Dietética/métodos , Medicina Baseada em Evidências/tendências , Alimentos Orgânicos , Trato Gastrointestinal/microbiologia , Fenômenos Fisiológicos da Nutrição/fisiologia , Valor Nutritivo , Probióticos/uso terapêutico , Humanos , Fenômenos Fisiológicos da Nutrição/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA