Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutrients ; 12(3)2020 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-32110880

RESUMO

The importance of gut microbiota in health and disease is being highlighted by numerous research groups worldwide. Atherosclerosis, the leading cause of heart disease and stroke, is responsible for about 50% of all cardiovascular deaths. Recently, gut dysbiosis has been identified as a remarkable factor to be considered in the pathogenesis of cardiovascular diseases (CVDs). In this review, we briefly discuss how external factors such as dietary and physical activity habits influence host-microbiota and atherogenesis, the potential mechanisms of the influence of gut microbiota in host blood pressure and the alterations in the prevalence of those bacterial genera affecting vascular tone and the development of hypertension. We will also be examining the microbiota as a therapeutic target in the prevention of CVDs and the beneficial mechanisms of probiotic administration related to cardiovascular risks. All these new insights might lead to novel analysis and CVD therapeutics based on the microbiota.


Assuntos
Aterosclerose/microbiologia , Aterosclerose/patologia , Microbioma Gastrointestinal , Animais , Aterosclerose/prevenção & controle , Aterosclerose/terapia , Transplante de Microbiota Fecal , Humanos , Terapia de Alvo Molecular , Medicina de Precisão , Probióticos/uso terapêutico
2.
Microorganisms ; 7(3)2019 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-30832423

RESUMO

Specific microbial profiles and changes in intestinal microbiota have been widely demonstrated to be associated with the pathogenesis of a number of extra-intestinal (obesity and metabolic syndrome) and intestinal (inflammatory bowel disease) diseases as well as other metabolic disorders, such as non-alcoholic fatty liver disease and type 2 diabetes. Thus, maintaining a healthy gut ecosystem could aid in avoiding the early onset and development of these diseases. Furthermore, it is mandatory to evaluate the alterations in the microbiota associated with pathophysiological conditions and how to counteract them to restore intestinal homeostasis. This review highlights and critically discusses recent literature focused on identifying changes in and developing gut microbiota-targeted interventions (probiotics, prebiotics, diet, and fecal microbiota transplantation, among others) for the above-mentioned pathologies. We also discuss future directions and promising approaches to counteract unhealthy alterations in the gut microbiota. Altogether, we conclude that research in this field is currently in its infancy, which may be due to the large number of factors that can elicit such alterations, the variety of related pathologies, and the heterogeneity of the population involved. Further research on the effects of probiotics, prebiotics, or fecal transplantations on the composition of the human gut microbiome is necessary.

3.
BMC Microbiol ; 15(1): 79, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25887178

RESUMO

BACKGROUND: The action of probiotics has been studied in vitro in cells isolated from both mice and humans, particularly enterocytes (IECs), dendritic cells (DCs) and co-cultures of peripheral DCs and IECs. Peripheral DCs and murine DCs differ from human gut DCs, and to date there are no data on the action of any probiotic on co-cultured human IECs and human intestinal DCs. To address this issue, a novel transwell model was used. Human IECs (Caco-2 cells) grown in the upper chamber of transwell filters were co-cultured with intestinal-like human DCs grown in the basolateral compartment of the transwells. The system was apically exposed for 4 h to live probiotic L. paracasei CNCM I-4034 obtained from the faeces of breastfed infants or to its cell-free culture supernatant (CFS) and challenged with Salmonella typhi. The secretion of pro- and anti-inflammatory cytokines in the basolateral compartment was determined by immunoassay, and the DC expression pattern of 20 TLR signaling pathway genes was analysed by PCR array. RESULTS: The presence of the live probiotic alone significantly increased IL-1ß, IL-6, IL-8, TGF-ß2, RANTES and IP-10 levels and decreased IL-12p40, IL-10, TGF- ß1 and MIP-1α levels. This release was correlated with a significant increase in the expression of almost all TLR signaling genes. By contrast, incubation of the co-culture with CFS increased IL-1ß, IL-6, TGF-ß2 and IP-10 production only when Salmonella was present. This induction was correlated with an overall decrease in the expression of all TLR genes except TLR9, which was strongly up-regulated. CONCLUSIONS: The data presented here clearly indicate that L. paracasei CNCM I-4034 significantly increases the release of pro-inflammatory cytokines, enhances TLR signaling pathway activation and stimulates rather than suppresses the innate immune system. Furthermore, our findings provide evidence that the effects of probiotics in the presence of IECs and DCs differ from the effects of probiotics on cultures of each cell type alone, as reported by us earlier. Thus, co-culture systems such as the one described here are needed to characterise the effects of probiotics in vitro, highlighting the potential utility of such co-cultures as a model system.

4.
PLoS One ; 9(5): e98401, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24852284

RESUMO

We have previously described the safety and immunomodulatory effects of Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036 in healthy volunteers. The scope of this work was to evaluate the effects of these probiotic strains on the hepatic steatosis of obese rats. We used the Zucker rat as a genetic model of obesity. Zucker-Lepr(fa/fa) rats received one of three probiotic strains, a mixture of L. paracasei CNCM I-4034 and B. breve CNCM I-4035, or a placebo for 30 days. An additional group of Zucker-lean+/fa rats received a placebo for 30 days. No alterations in intestinal histology, in the epithelial, lamina propria, muscular layers of the ileal or colonic mucosa, or the submucosae, were observed in any of the experimental groups. Triacylglycerol content decreased in the liver of Zucker-Lepr(fa/fa) rats that were fed L. rhamnosus, B. breve, or the mixture of B. breve and L. paracasei. Likewise, the area corresponding to neutral lipids was significantly smaller in the liver of all four groups of Zucker-Lepr(fa/fa) rats that received probiotics than in rats fed the placebo. Zucker-Lepr(fa/fa) rats exhibited significantly greater serum LPS levels than Zucker-lean+/fa rats upon administration of placebo for 30 days. In contrast, all four groups of obese Zucker-Lepr(fa/fa) rats that received LAB strains exhibited serum LPS concentrations similar to those of Zucker-lean+/fa rats. Serum TNF-α levels decreased in the Zucker-Lepr(fa/fa) rats that received B. breve, L. rhamnosus, or the mixture, whereas L. paracasei feeding decreased IL-6 levels in the serum of Zucker-Lepr(fa/fa) rats. In conclusion, the probiotic strains reduced hepatic steatosis in part by lowering serum LPS, and had an anti-inflammatory effect in obese Zucker rats.


Assuntos
Bifidobacterium , Fígado Gorduroso/terapia , Lactobacillus , Probióticos , Adipocinas/sangue , Animais , Citocinas/sangue , Lipopolissacarídeos/sangue , Masculino , Obesidade/sangue , Ratos , Ratos Zucker
5.
Br J Nutr ; 111(10): 1727-37, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24480321

RESUMO

The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance to the commensal microbiota and food antigens. Different strains of probiotics possess the ability to finely regulate the activation of dendritic cells (DC), polarising the subsequent activity of T-cells. Nevertheless, information about their underlying mechanisms of action is scarce. In the present study, we investigated the immunomodulatory effects of a potentially probiotic strain, Lactobacillus rhamnosus CNCM I-4036, and its cell-free culture supernatant (CFS) on human DC challenged with Escherichia coli. The results showed that the levels of pro-inflammatory cytokines such as IL-1ß, IL-6, IL-8 and IL-12p70 were higher in the cells treated with live L. rhamnosus than in the cells treated with the CFS. In the presence of E. coli, the supernatant was more effective than the probiotic bacteria in reducing the secretion of pro-inflammatory cytokines. In addition, live L. rhamnosus potently induced the production of transforming growth factor (TGF)-ß1 and TGF-ß2, whereas the CFS increased the secretion of TGF-ß1. However, in the presence of E. coli, both treatments restored the levels of TGF-ß. The probiotic strain L. rhamnosus CNCM I-4036 and its CFS were able to activate the Toll-like receptor signalling pathway, enhancing innate immunity. The two treatments induced gene transcription of TLR-9. Live L. rhamnosus activated the expression of TLR-2 and TLR-4 genes, whereas the CFS increased the expression of TLR-1 and TLR-5 genes. In response to the stimulation with probiotic/CFS and E. coli, the expression of each gene tested was notably increased, with the exception of TNF-α and NFKBIA. In conclusion, the CFS exhibited an extraordinary ability to suppress the production of pro-inflammatory cytokines by DC, and may be used as an effective and safer alternative to live bacteria.


Assuntos
Citocinas/metabolismo , Células Dendríticas/citologia , Escherichia coli/isolamento & purificação , Intestinos/microbiologia , Lacticaseibacillus rhamnosus/metabolismo , Probióticos/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Sistema Livre de Células/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Expressão Gênica , Humanos , Imunoensaio , Intestinos/imunologia , Lacticaseibacillus rhamnosus/imunologia , Reação em Cadeia da Polimerase , Transdução de Sinais , Receptores Toll-Like/genética
6.
J Pediatr Gastroenterol Nutr ; 57(4): 461-6, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23752082

RESUMO

OBJECTIVES: There are many differences in the fecal infant microbiota associated with various feeding methods. The aim of this study was to examine the major differences in the fecal microbiota of breast-fed (BF) and formula-fed (FF) infants and to describe the principal bacterial components that would explain the variability in the predominant bacterial families and genus clusters. METHODS: Fecal samples from 58 infants, 31 of whom were exclusively BF and 27 of whom were exclusively FF with a standard formula in agreement with the European Society for Pediatric Gastroenterology, Hepatology, and Nutrition recommendations, were analyzed by fluorescent in situ hybridization combined with flow cytometry. Principal component analysis was used to maximize the information gained for the predominant bacterial families and genus clusters using a minimal number of bacterial groups. RESULTS: The predominant detected group was Bifidobacterium, followed by Enterobacteriaceae and Bacteroides in both BF and FF infants. The Lactobacillus group was the only independent variable associated with FF infants. We also found that 3 principal components were sufficient to describe the association between the bacterial group, genus, and species studied in BF and FF infants; however, these components differed between BF and FF infants. For the former, the 3 factors found were Bifidobacterium/Enterobacteriaceae, Lactobacillus/Bacteroides, and Clostridium coccoides/Atopobium; for the latter, Bifidobacterium/Enterobacteriaceae, Bacteroides and C coccoides were observed. CONCLUSIONS: There is a clear clustering of components of infant microbiota based on the feeding method.


Assuntos
Bactérias , Alimentação com Mamadeira , Aleitamento Materno , Colo/microbiologia , Fezes/microbiologia , Microbiota , Humanos , Lactente , Fórmulas Infantis , Análise de Componente Principal , Especificidade da Espécie
7.
PLoS One ; 8(3): e59370, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555025

RESUMO

Dendritic cells (DCs) constitute the first point of contact between gut commensals and our immune system. Despite growing evidence of the immunomodulatory effects of probiotics, the interactions between the cells of the intestinal immune system and bacteria remain largely unknown. Indeed,, the aim of this work was to determine whether the probiotic Bifidobacterium breve CNCM I-4035 and its cell-free culture supernatant (CFS) have immunomodulatory effects in human intestinal-like dendritic cells (DCs) and how they respond to the pathogenic bacterium Salmonella enterica serovar Typhi, and also to elucidate the molecular mechanisms involved in these interactions. Human DCs were directly challenged with B. breve/CFS, S. typhi or a combination of these stimuli for 4 h. The expression pattern of genes involved in Toll-like receptor (TLR) signaling pathway and cytokine secretion was analyzed. CFS decreased pro-inflammatory cytokines and chemokines in human intestinal DCs challenged with S. typhi. In contrast, the B. breve CNCM I-4035 probiotic strain was a potent inducer of the pro-inflammatory cytokines and chemokines tested, i.e., TNF-α, IL-8 and RANTES, as well as anti-inflammatory cytokines including IL-10. CFS restored TGF-ß levels in the presence of Salmonella. Live B.breve and its supernatant enhanced innate immune responses by the activation of TLR signaling pathway. These treatments upregulated TLR9 gene transcription. In addition, CFS was a more potent inducer of TLR9 expression than the probiotic bacteria in the presence of S. typhi. Expression levels of CASP8 and IRAK4 were also increased by CFS, and both treatments induced TOLLIP gene expression. Our results indicate that the probiotic strain B. breve CNCM I-4035 affects the intestinal immune response, whereas its supernatant exerts anti-inflammatory effects mediated by DCs. This supernatant may protect immune system from highly infectious agents such as Salmonella typhi and can down-regulate pro-inflammatory pathways.


Assuntos
Bifidobacterium/fisiologia , Meios de Cultivo Condicionados/farmacologia , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Probióticos/farmacologia , Salmonella typhi/imunologia , Caspase 8/genética , Caspase 8/imunologia , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Células Dendríticas/citologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Salmonella typhi/crescimento & desenvolvimento , Transdução de Sinais , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
8.
Br J Nutr ; 109 Suppl 2: S27-34, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23360878

RESUMO

Ideally, cell models should resemble the in vivo conditions; however, in most in vitro experimental models, epithelial cells are cultivated as monolayers, in which the establishment of functional epithelial features is not achieved. To overcome this problem, co-culture experiments with probiotics, dendritic cells and intestinal epithelial cells and three-dimensional models attempt to reconcile the complex and dynamic interactions that exist in vivo between the intestinal epithelium and bacteria on the luminal side and between the epithelium and the underlying immune system on the basolateral side. Additional models include tissue explants, bioreactors and organoids. The present review details the in vitro models used to study host-microbe interactions and explores the new tools that may help in understanding the molecular mechanisms of these interactions.


Assuntos
Células Dendríticas/imunologia , Interações Hospedeiro-Parasita , Mucosa Intestinal/imunologia , Modelos Biológicos , Probióticos , Animais , Reatores Biológicos/microbiologia , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/microbiologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Macrófagos/citologia , Macrófagos/imunologia , Organoides/imunologia , Organoides/microbiologia , Técnicas de Cultura de Tecidos
9.
Br J Nutr ; 109 Suppl 2: S35-50, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23360880

RESUMO

According to the FAO and the WHO, probiotics are 'live microorganisms which, when administered in adequate amounts, confer a health benefit on the host'. The strains most frequently used as probiotics include lactic acid bacteria and bifidobacteria, which are isolated from traditional fermented products and the gut, faeces and breast milk of human subjects. The identification of microorganisms is the first step in the selection of potential probiotics. The present techniques, including genetic fingerprinting, gene sequencing, oligonucleotide probes and specific primer selection, discriminate closely related bacteria with varying degrees of success. Additional molecular methods, such as denaturing gradient gel electrophoresis/temperature gradient gel electrophoresis and fluorescence in situ hybridisation, are employed to identify and characterise probiotics. The ability to examine fully sequenced genomes has accelerated the application of genetic approaches to the elucidation of the functional roles of probiotics. One of the best-demonstrated clinical benefits of probiotics is the prevention and treatment of acute and antibiotic-associated diarrhoea;however, there is mounting evidence for a potential role for probiotics in the treatment of allergies and intestinal, liver and metabolic diseases. There are various mechanisms by which probiotics exert their beneficial effects: regulation of intestinal permeability, normalisation of host intestinal microbiota, improvement of gut immune barrier function, and adjustment between pro- and anti-inflammatory cytokines. The number of studies carried out to test the effects of probiotics in vitro and in animals is enormous. However, the most reliable method of assessing the therapeutic benefits of any probiotic strain is the use of randomised, placebo-controlled trials, which are reviewed in this article [corrected].


Assuntos
Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/isolamento & purificação , Trato Gastrointestinal/microbiologia , Lactobacillales/crescimento & desenvolvimento , Lactobacillales/isolamento & purificação , Probióticos/isolamento & purificação , Animais , Bifidobacterium/classificação , Bifidobacterium/imunologia , Produtos Fermentados do Leite/microbiologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/fisiologia , Humanos , Absorção Intestinal , Mucosa Intestinal/microbiologia , Lactobacillales/classificação , Lactobacillales/imunologia , Viabilidade Microbiana , Leite Humano/microbiologia , Permeabilidade , Probióticos/efeitos adversos , Probióticos/uso terapêutico
10.
Br J Nutr ; 109 Suppl 2: S51-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23360881

RESUMO

The aim of the present study was to isolate, identify and characterise novel strains of lactic acid bacteria and bifidobacteria with probiotic properties from the faeces of exclusively breast-fed infants. Of the 4680 isolated colonies, 758 exhibited resistance to low pH and tolerance to high concentrations of bile salts; of these, only forty-two exhibited a strong ability to adhere to enterocytes in vitro. The identities of the isolates were confirmed by 16S ribosomal RNA (rRNA) sequencing, which permitted the grouping of the forty-two bacteria into three different strains that showed more than 99 % sequence identity with Lactobacillus paracasei, Lactobacillus rhamnosus and Bifidobacterium breve, respectively. The strain identification was confirmed by sequencing the 16S-23S rRNA intergenic spacer regions. Strains were assayed for enzymatic activity and carbohydrate utilisation, and they were deposited in the Collection Nationale de Cultures de Microorganismes (CNCM) of the Institute Pasteur and named L. paracasei CNCM I-4034, B. breve CNCM I-4035 and L. rhamnosus CNCM I-4036. The strains were susceptible to antibiotics and did not produce undesirable metabolites, and their safety was assessed by acute ingestion in immunocompetent and immunosuppressed BALB/c mouse models. The three novel strains inhibited in vitro the meningitis aetiological agent Listeria monocytogenes and human rotavirus infections. B. breve CNCM I-4035 led to a higher IgA concentration in faeces and plasma of mice. Overall, these results suggest that L. paracasei CNCM I-4034, B. breve CNCM I-4035 and L. rhamnosus CNCM I-4036 should be considered as probiotic strains, and their human health benefits should be further evaluated.


Assuntos
Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/isolamento & purificação , Aleitamento Materno , Fezes/microbiologia , Lactobacillus/crescimento & desenvolvimento , Lactobacillus/isolamento & purificação , Probióticos/isolamento & purificação , Animais , Antibiose , Aderência Bacteriana , Bifidobacterium/classificação , Bifidobacterium/imunologia , Enterócitos/microbiologia , Feminino , Humanos , Imunidade nas Mucosas , Hospedeiro Imunocomprometido , Recém-Nascido , Lactobacillus/classificação , Lactobacillus/imunologia , Lacticaseibacillus rhamnosus/classificação , Lacticaseibacillus rhamnosus/crescimento & desenvolvimento , Lacticaseibacillus rhamnosus/imunologia , Lacticaseibacillus rhamnosus/isolamento & purificação , Listeria monocytogenes/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Probióticos/efeitos adversos , Probióticos/uso terapêutico , Rotavirus/crescimento & desenvolvimento , Espanha , Organismos Livres de Patógenos Específicos
11.
Br J Nutr ; 109 Suppl 2: S63-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23360882

RESUMO

Numerous in vitro and in vivo studies conducted using different probiotic micro-organisms have demonstrated their ability to interfere with the growth and virulence of a variety of enteropathogens. The reported beneficial effects of the use of probiotics to complement antibiotic therapy or prevent diarrhoea or gastrointestinal infection in infants have increased in recent years. In the present study, we demonstrated the capacity of supernatants obtained from three novel probiotics (Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036) isolated from the faeces of breastfed infants to inhibit the growth of enterotoxigenic and enteropathogenic (EPEC) bacteria, such as Escherichia coli, Salmonella and Shigella. To assess their potential antimicrobial activity, the 17 and 24 h cell-free supernatants broth concentrates (10×) having 1, 2 or 4 % of the three probiotics were incubated with EPEC bacteria strains. After 17 h of co-culture, the supernatants were able to inhibit the growth of E. coli, Salmonella and Shigella up to 40, 55 and 81 %, respectively. However, the inhibitory capacity of some supernatants was maintained or completely lost when the supernatants (pH 3·0) were neutralised (pH 6·5). Overall, these results demonstrated that L. paracasei CNCM I-4034, B. breve CNCM I-4035 and L. rhamnosus CNCM I-4036 produce compounds that exhibited strain-specific inhibition of enterobacteria and have the potential to be used as probiotics in functional foods.


Assuntos
Antibiose , Bifidobacterium/isolamento & purificação , Aleitamento Materno , Fezes/microbiologia , Gastroenterite/prevenção & controle , Lactobacillus/isolamento & purificação , Probióticos/isolamento & purificação , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Meios de Cultivo Condicionados/metabolismo , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/patogenicidade , Escherichia coli Enterotoxigênica/crescimento & desenvolvimento , Escherichia coli Enterotoxigênica/patogenicidade , Gastroenterite/microbiologia , Humanos , Concentração de Íons de Hidrogênio , Recém-Nascido , Lactobacillus/crescimento & desenvolvimento , Lactobacillus/metabolismo , Lacticaseibacillus rhamnosus/crescimento & desenvolvimento , Lacticaseibacillus rhamnosus/isolamento & purificação , Lacticaseibacillus rhamnosus/metabolismo , Viabilidade Microbiana , Probióticos/metabolismo , Probióticos/uso terapêutico , Salmonella typhi/crescimento & desenvolvimento , Salmonella typhi/patogenicidade , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade , Shigella sonnei/crescimento & desenvolvimento , Shigella sonnei/patogenicidade , Espanha , Fatores de Tempo
12.
Ann Nutr Metab ; 61(2): 160-74, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23037511

RESUMO

Probiotics are live microorganisms that provide health benefits to the host when ingested in adequate amounts. The strains most frequently used as probiotics include lactic acid bacteria and bifidobacteria. Probiotics have demonstrated significant potential as therapeutic options for a variety of diseases, but the mechanisms responsible for these effects have not been fully elucidated yet. Several important mechanisms underlying the antagonistic effects of probiotics on various microorganisms include the following: modification of the gut microbiota, competitive adherence to the mucosa and epithelium, strengthening of the gut epithelial barrier and modulation of the immune system to convey an advantage to the host. Accumulating evidence demonstrates that probiotics communicate with the host by pattern recognition receptors, such as toll-like receptors and nucleotide-binding oligomerization domain-containing protein-like receptors, which modulate key signaling pathways, such as nuclear factor-ĸB and mitogen-activated protein kinase, to enhance or suppress activation and influence downstream pathways. This recognition is crucial for eliciting measured antimicrobial responses with minimal inflammatory tissue damage. A clear understanding of these mechanisms will allow for appropriate probiotic strain selection for specific applications and may uncover novel probiotic functions. The goal of this systematic review was to explore probiotic modes of action focusing on how gut microbes influence the host.


Assuntos
Probióticos/metabolismo , Animais , Anti-Infecciosos/metabolismo , Aderência Bacteriana , Bifidobacterium/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/microbiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lactobacillaceae/metabolismo , Metagenoma , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Animais , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais
13.
PLoS One ; 7(8): e43197, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905233

RESUMO

Probiotic bacteria have been shown to modulate immune responses and could have therapeutic effects in allergic and inflammatory disorders. However, little is known about the signalling pathways that are engaged by probiotics. Dendritic cells (DCs) are antigen-presenting cells that are involved in immunity and tolerance. Monocyte-derived dendritic cells (MoDCs) and murine DCs are different from human gut DCs; therefore, in this study, we used human DCs generated from CD34+ progenitor cells (hematopoietic stem cells) harvested from umbilical cord blood; those DCs exhibited surface antigens of dendritic Langerhans cells, similar to the lamina propria DCs in the gut. We report that both a novel probiotic strain isolated from faeces of exclusively breast-fed newborn infants, Lactobacillus paracasei CNCM I-4034, and its cell-free culture supernatant (CFS) decreased pro-inflammatory cytokines and chemokines in human intestinal DCs challenged with Salmonella. Interestingly, the supernatant was as effective as the bacteria in reducing pro-inflammatory cytokine expression. In contrast, the bacterium was a potent inducer of TGF-ß2 secretion, whereas the supernatant increased the secretion of TGF-ß1 in response to Salmonella. We also showed that both the bacteria and its supernatant enhanced innate immunity through the activation of Toll-like receptor (TLR) signalling. These treatments strongly induced the transcription of the TLR9 gene. In addition, upregulation of the CASP8 and TOLLIP genes was observed. This work demonstrates that L. paracasei CNCM I-4034 enhanced innate immune responses, as evidenced by the activation of TLR signalling and the downregulation of a broad array of pro-inflammatory cytokines. The use of supernatants like the one described in this paper could be an effective and safe alternative to using live bacteria in functional foods.


Assuntos
Citocinas/metabolismo , Células Dendríticas/citologia , Intestinos/citologia , Lactobacillus/metabolismo , Infecções por Salmonella/metabolismo , Salmonella/metabolismo , Receptores Toll-Like/metabolismo , Antígenos CD34/biossíntese , Caspase 8/metabolismo , Quimiocinas/metabolismo , Técnicas de Cocultura , Células Dendríticas/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Imunidade Inata/fisiologia , Inflamação , Intestinos/microbiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Probióticos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...