Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nutrients ; 12(7)2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32640639

RESUMO

Commensal gut microbiota and probiotics have numerous effects on the host's metabolic and protective systems, which occur primarily through the intestinal epithelial cell interface. Prebiotics, like galacto-oligosaccharides (GOS) are widely used to modulate their function and abundance. However, important structure-function relations may exist, requiring a detailed structural characterization. Here, we detailed the structural characterization of bovine whey derived oligosaccharide preparations enriched with GOS or not, dubbed GOS-enriched milk oligosaccharides (GMOS) or MOS, respectively. We explore GMOS's and MOS's potential to improve intestinal epithelial barrier function, assessed in a model based on barrier disruptive effects of the Clostridioides difficile toxin A. GMOS and MOS contain mainly GOS species composed of ß1-6- and ß1-3-linked galactoses, and 3'- and 6'-sialyllactose. Both GMOS and MOS, combined with lactobacilli, like Lactobacillus rhamnosus (LPR, NCC4007), gave synergistic epithelial barrier protection, while no such effect was observed with Bifidobacterium longum (BL NCC3001), Escherichia coli (Nissle) or fructo-oligosaccharides. Mechanistically, for barrier protection with MOS, (i) viable LPR was required, (ii) acidification of growth medium was not enough, (iii) LPR did not directly neutralize toxin A, and (iv) physical proximity of LPR with the intestinal epithelial cells was necessary. This is the first study, highlighting the importance of structure-function specificity and the necessity of the simultaneous presence of prebiotic, probiotic and host cell interactions required for a biological effect.


Assuntos
Microbioma Gastrointestinal , Mucosa Intestinal , Oligossacarídeos , Simbióticos , Soro do Leite , Animais , Toxinas Bacterianas/efeitos adversos , Bovinos , Linhagem Celular Tumoral , Enterotoxinas/efeitos adversos , Galactose/química , Galactose/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lactobacillus/metabolismo , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Prebióticos , Probióticos/farmacologia , Substâncias Protetoras/química , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia
2.
Gastroenterology ; 139(6): 2102-2112.e1, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20600016

RESUMO

BACKGROUND & AIMS: Clinical and preclinical studies have associated gastrointestinal inflammation and infection with altered behavior. We investigated whether chronic gut inflammation alters behavior and brain biochemistry and examined underlying mechanisms. METHODS: AKR mice were infected with the noninvasive parasite Trichuris muris and given etanercept, budesonide, or specific probiotics. Subdiaphragmatic vagotomy was performed in a subgroup of mice before infection. Gastrointestinal inflammation was assessed by histology and quantification of myeloperoxidase activity. Serum proteins were measured by proteomic analysis, circulating cytokines were measured by fluorescence activated cell sorting array, and serum tryptophan and kynurenine were measured by liquid chromatography. Behavior was assessed using light/dark preference and step-down tests. In situ hybridization was used to assess brain-derived neurotrophic factor (BDNF) expression in the brain. RESULTS: T muris caused mild to moderate colonic inflammation and anxiety-like behavior that was associated with decreased hippocampal BDNF messenger RNA (mRNA). Circulating tumor necrosis factor-α and interferon-γ, as well as the kynurenine and kynurenine/tryptophan ratio, were increased. Proteomic analysis showed altered levels of several proteins related to inflammation and neural function. Administration of etanercept, and to a lesser degree of budesonide, normalized behavior, reduced cytokine and kynurenine levels, but did not influence BDNF expression. The probiotic Bifidobacterium longum normalized behavior and BDNF mRNA but did not affect cytokine or kynurenine levels. Anxiety-like behavior was present in infected mice after vagotomy. CONCLUSIONS: Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry, which can be normalized by inflammation-dependent and -independent mechanisms, neither of which requires the integrity of the vagus nerve.


Assuntos
Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Colite/fisiopatologia , Hipocampo/fisiologia , Tricuríase/fisiopatologia , Animais , Ansiedade/imunologia , Ansiedade/parasitologia , Fator Neurotrófico Derivado do Encéfalo/genética , Doença Crônica , Colite/imunologia , Colite/parasitologia , Citocinas/sangue , Cinurenina/sangue , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Proteômica , RNA Mensageiro/metabolismo , Tricuríase/imunologia , Trichuris , Triptofano/sangue , Vagotomia , Nervo Vago/imunologia , Nervo Vago/fisiopatologia
3.
PLoS One ; 5(1): e8666, 2010 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20072622

RESUMO

Carrageenan (CGN) is a high molecular weight sulphated polysaccharide derived from red seaweeds. In rodents, its degraded forms (dCGN) can induce intestinal inflammation associated with macrophage recruitment and activation. The aim of this study was: 1) to analyze the size-dependent effects of dCGN on colon inflammation in vivo, and 2) to correlate these effects with monocyte/macrophage proliferation, cytokine production and expression of various cell surface antigens including ICAM-1 adhesion molecule. Peripheral blood monocytes (PBM) and THP-1 monocytic cells were cultured in the presence of either 10 or 40 kDa, dCGN. The 40 kDa, but not the 10 kDa dCGN, induced colitis in in vivo. Degraded CGN inhibited THP-1 cell proliferation in vitro, arresting the cells in G1 phase. In addition, dCGN increased ICAM-1 expression in both PBM and THP-1 cells with a major effect seen after 40 kDa dCGN exposure. Also, dCGN stimulated monocyte aggregation in vitro that was prevented by incubation with anti-ICAM-1 antibody. Finally, dCGN stimulated TNF-alpha expression and secretion by both PBM and THP-1 cells. All these effects were linked to NF-kappaB activation. These data strongly suggest that the degraded forms of CGN have a pronounced effect on monocytes, characteristic of an inflammatory phenotype.


Assuntos
Carragenina/toxicidade , Colite/induzido quimicamente , Molécula 1 de Adesão Intercelular/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Colite/metabolismo , Primers do DNA , Citometria de Fluxo , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Mamm Genome ; 20(8): 516-27, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19711126

RESUMO

Dysregulation of intestinal epithelial cell performance is associated with an array of pathologies whose onset mechanisms are incompletely understood. While whole-genomics approaches have been valuable for studying the molecular basis of several intestinal diseases, a thorough analysis of gene expression along the healthy gastrointestinal tract is still lacking. The aim of this study was to map gene expression in gastrointestinal regions of healthy human adults and to implement a procedure for microarray data analysis that would allow its use as a reference when screening for pathological deviations. We analyzed the gene expression signature of antrum, duodenum, jejunum, ileum, and transverse colon biopsies using a biostatistical method based on a multivariate and univariate approach to identify region-selective genes. One hundred sixty-six genes were found responsible for distinguishing the five regions considered. Nineteen had never been described in the GI tract, including a semaphorin probably implicated in pathogen invasion and six novel genes. Moreover, by crossing these genes with those retrieved from an existing data set of gene expression in the intestine of ulcerative colitis and Crohn's disease patients, we identified genes that might be biomarkers of Crohn's and/or ulcerative colitis in ileum and/or colon. These include CLCA4 and SLC26A2, both implicated in ion transport. This study furnishes the first map of gene expression along the healthy human gastrointestinal tract. Furthermore, the approach implemented here, and validated by retrieving known gene profiles, allowed the identification of promising new leads in both healthy and disease states.


Assuntos
Biomarcadores/metabolismo , Gastroenteropatias/genética , Trato Gastrointestinal/metabolismo , Expressão Gênica , Adulto , Feminino , Gastroenteropatias/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Adulto Jovem
5.
J Proteome Res ; 8(4): 2090-105, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19281268

RESUMO

Coevolution shapes interorganismal crosstalk leading to profound and diverse cellular and metabolic changes as observed in gut dysbiosis in human diseases. Here, we modulated a simplified gut microbiota using pro-, pre-, and synbiotics to assess the depth of systemic metabolic exchanges in mice, using a multicompartmental modeling approach with metabolic signatures from 10 tissue/fluid compartments. The nutritionally induced microbial changes modulated host lipid, carbohydrate, and amino acid metabolism at a panorganismal scale. Galactosyl-oligosaccharides reduced lipogenesis, triacylglycerol incorporation into lipoproteins and triglyceride concentration in the liver and the kidney. Those changes were not correlated with decreased plasma lipoproteins that were specifically induced by L. rhamnosus supplementation. Additional alteration of transmethylation metabolic pathways (homocysteine-betaine) was observed in the liver and the pancreas following pre- and synbiotic microbial modulation, which may be of interest for control of glucose metabolism and insulin sensitivity. Probiotics also reduced hepatic glycogen and glutamine and adrenal ascorbate with inferred effects on energy homeostasis, antioxidation, and steroidogenesis. These studies show the breadth and the depth of gut microbiome modulations of host biochemistry and reveal that major mammalian metabolic processes are under symbiotic homeostatic control.


Assuntos
Trato Gastrointestinal/microbiologia , Metaboloma , Metagenoma , Animais , Feminino , Trato Gastrointestinal/metabolismo , Humanos , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Ressonância Magnética Nuclear Biomolecular , Probióticos/metabolismo
6.
Mol Syst Biol ; 4: 205, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18628745

RESUMO

Gut microbiome-host metabolic interactions affect human health and can be modified by probiotic and prebiotic supplementation. Here, we have assessed the effects of consumption of a combination of probiotics (Lactobacillus paracasei or L. rhamnosus) and two galactosyl-oligosaccharide prebiotics on the symbiotic microbiome-mammalian supersystem using integrative metabolic profiling and modeling of multiple compartments in germ-free mice inoculated with a model of human baby microbiota. We have shown specific impacts of two prebiotics on the microbial populations of HBM mice when co-administered with two probiotics. We observed an increase in the populations of Bifidobacterium longum and B. breve, and a reduction in Clostridium perfringens, which were more marked when combining prebiotics with L. rhamnosus. In turn, these microbial effects were associated with modulation of a range of host metabolic pathways observed via changes in lipid profiles, gluconeogenesis, and amino-acid and methylamine metabolism associated to fermentation of carbohydrates by different bacterial strains. These results provide evidence for the potential use of prebiotics for beneficially modifying the gut microbial balance as well as host energy and lipid homeostasis.


Assuntos
Genoma/genética , Intestinos/microbiologia , Lactobacillus/genética , Lactobacillus/metabolismo , Modelos Animais , Probióticos , Biologia de Sistemas , Animais , Peso Corporal , Ceco/metabolismo , Ácidos Graxos/metabolismo , Fezes/microbiologia , Feminino , Genoma/efeitos dos fármacos , Humanos , Lactente , Intestinos/efeitos dos fármacos , Fígado/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Probióticos/farmacologia
7.
J Nutr ; 138(1): 123-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18156414

RESUMO

Fructooligosaccharides (FOS) are considered prebiotics because of their ability to promote growth of specific beneficial gut bacteria, such as bifidobacteria. Some studies reported potential immune-modulating properties. The aim of this study was to investigate the effect of FOS:inulin mix on murine response to Salmonella vaccine and evaluate the relevance toward protection against Salmonella infection. Balb/c mice were fed a diet containing 5% FOS:inulin mix or a control diet 1 wk before oral immunization with a suboptimal dose of live attenuated Salmonella typhimurium vaccine. Four weeks after vaccination, mice were infected with LD100 of virulent S. typhimurium. Specific blood Salmonella immunoglobulin G and fecal immunoglobulin A significantly increased in mice fed the diet containing prebiotics compared with control mice 4 wk postimmunization. Peritoneal macrophage phagocytic activity also significantly increased in FOS:inulin-fed mice at 1 wk postimmunization compared with control mice. No detectable effects were observed on the percentage of lymphoid cell subsets in the spleen. However, production of cytokines, interferon-gamma, interleukin-12, and tumor necrosis factor alpha, was numerically increased in spleen cell cultures stimulated with mitogens from FOS:inulin-fed mice 1 and 4 wk postimmunization. Salmonella translocation to lymphoid organs was not affected by feeding FOS:inulin. However, the improved response to Salmonella vaccine was concomitant with an increase in the survival rate of FOS:inulin-fed mice upon challenge with virulent Salmonella. No detectable effects were observed on the composition or the metabolic activity of the microbiota. Overall, the data suggest that a diet supplemented with FOS:inulin mix stimulates mucosal immunity and seems to improve efficacy of an oral vaccine.


Assuntos
Dieta , Inulina/administração & dosagem , Oligossacarídeos/administração & dosagem , Vacinas contra Salmonella/imunologia , Administração Oral , Animais , Anticorpos Antibacterianos , Citocinas/metabolismo , Feminino , Imunoglobulina A/metabolismo , Imunoglobulina G/sangue , Inulina/farmacologia , Metaloporfirinas , Camundongos , Camundongos Endogâmicos BALB C , Oligossacarídeos/farmacologia , Fagócitos , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/administração & dosagem , Baço/citologia , Baço/metabolismo
8.
Genomics ; 91(1): 70-7, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18035521

RESUMO

The intestinal mucus layer and endogenous microbiota are strongly intertwined and this contributes to the maintenance of the epithelial barrier and ultimately of gut homeostasis. To understand the molecular foundations of such relationship, we investigated if the nature of the microbiota transcriptionally regulates mucus layer composition in vivo. We found that the expression of mucins 1 to 4 and trefoil factor 3 was down-regulated in the ileum and colon of conventional and reconventionalized mice compared with germ-free animals. Conversely, very limited colon-restricted changes in transmembrane mucins were detected in mice colonized with human adult or baby microbiota. Moreover, by microarray analysis, the murine endogenous microbiota was found to modulate genes putatively involved in mucin secretion. These findings show that a well-established microbial community participates in the regulation of the gut mucus layer and that its composition and adequacy to the host are key factors in this process.


Assuntos
Regulação para Baixo/fisiologia , Vida Livre de Germes/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucinas/biossíntese , Adulto , Animais , Colo/metabolismo , Colo/microbiologia , Perfilação da Expressão Gênica , Humanos , Íleo/metabolismo , Íleo/microbiologia , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos C3H , Mucinas/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fator Trefoil-3
9.
J Pediatr Gastroenterol Nutr ; 43(1): 16-24, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16819372

RESUMO

OBJECTIVES: Psychological stress during the neonatal period results in intestinal barrier dysfunction and growth alterations later in life. We aimed to restore impaired barrier function and growth rate by a nutritional intervention. METHODS: Male rat pups (n = 84) were assigned to 1 of 2 rearing conditions from postnatal day (PND) 2 to PND14: S, separated 3 h/d from their mothers, or H, 15 min/d handled controls. From PND15 to PND35, rats received a control diet or a similar diet adapted to contain arachidonic and docosahexaenoic acids, galacto- and fructo-oligosaccharides and Lactobacillus paracasei NCC2461. RESULTS: Maternal separation had only a minor impact on the measured gut barrier parameters at PND15, whereas it severely affected them at PND35. At this age, intestinal permeability to macromolecules was higher, mucin content in small intestinal tissues was lower and microbiota composition was altered in S compared with H animals. Feeding the adapted diet normalized the intestinal permeability, although it did not restore intestinal mucin content or microbiota. In addition, the adapted diet improved the growth rate recovery of the S animals after weaning and resulted in increased villus length in small intestine. CONCLUSION: Our results suggest that an adapted diet containing specific long-chain polyunsaturated fatty acids, prebiotics and probiotics can revert the negative imprinting of neonatal stress on both intestinal barrier function and growth.


Assuntos
Alimentos Formulados , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/fisiopatologia , Intestino Delgado/crescimento & desenvolvimento , Intestino Delgado/fisiopatologia , Estresse Psicológico/fisiopatologia , Animais , Animais Recém-Nascidos , Ácido Araquidônico/administração & dosagem , Ácido Araquidônico/metabolismo , Peso Corporal , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/metabolismo , Ingestão de Alimentos , Homeostase , Absorção Intestinal , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Lactobacillus/metabolismo , Masculino , Privação Materna , Mucinas/metabolismo , Oligossacarídeos/administração & dosagem , Oligossacarídeos/metabolismo , Permeabilidade , Probióticos/administração & dosagem , Probióticos/metabolismo , Ratos , Ratos Long-Evans , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Fatores de Tempo , Desmame
10.
J Nutr ; 135(12): 2753-61, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16317116

RESUMO

The mucus layer covering the epithelium is one of the main lines of defense of the colonic barrier. Both mucus gel and mucin expressions are altered during colonic inflammation and could be involved in epithelial repair. We postulated that modulating colonic mucus and mucins by probiotic supplementation could contribute to healing inflammatory mucosa. Our aim in this study was to determine whether probiotics could repair dextran-sodium sulfate (DSS)-induced chronic colitis in mice, and whether modifications of the colonic mucins could be involved. For that purpose, the VSL#3 probiotic mixture of 8 lactic acid bacteria probiotic strains was administered daily for 2 wk to mice with a mucosa impaired by a mild DSS treatment, and to mice with a normal mucosa. Probiotic strains survived in the gastrointestinal tract, increased the cecal concentrations of bifidobacteria, and modified cecal microflora metabolic activity in both DSS-treated and healthy mice. However, probiotic supplementation did not reverse the inflammation induced by DSS at either the macroscopic or histological level. Concurrently, probiotics did not modify the colonic mucus barrier, in terms of either mucin gene expression or adherent mucus layer thickness. In conclusion, the modification of microflora by supplementation with the VSL#3 probiotic mixture did not help to repair the colonic barrier breakdown caused by DSS treatment. The potential healing roles of mucins were neither confirmed nor invalidated by this study.


Assuntos
Colite/induzido quimicamente , Colite/terapia , Mucosa Intestinal/fisiologia , Probióticos/uso terapêutico , Animais , Colo/microbiologia , Sulfato de Dextrana , Modelos Animais de Doenças , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/fisiologia , Probióticos/administração & dosagem
11.
Physiol Genomics ; 23(2): 235-45, 2005 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-16131529

RESUMO

The postnatal maturation of the gut, partially modulated by bacterial colonization, ends up in the establishment of an efficient barrier to luminal antigens and bacteria. The use of broad-spectrum antibiotics in pediatric practices alters the gut bacterial colonization and, consequently, may impair the maturation of the gut barrier function. To test this hypothesis, suckling Sprague-Dawley rats received a daily intragastric gavage of antibiotic (Clamoxyl; an amoxicillin-based commercial preparation) or saline solution from postnatal day 7 (d7) until d17 or d21. Luminal microbiota composition and global gene expression profile were analyzed on samples from small intestine and colon of each group. The treatment with Clamoxyl resulted in the almost-complete eradication of Lactobacillus in the whole intestine and in a drastic reduction of colonic total aerobic and anaerobic bacteria, in particular Enterobacteriacae and Enterococcus. The global gene expression analysis revealed that Clamoxyl affects the maturation process of 249 and 149 Affymetrix probe sets in the proximal and distal small intestine, respectively, and 163 probe sets in the colon. The expression of genes coding for Paneth cell products (defensins, matrilysin, and phospholipase A2) was significantly downregulated by the Clamoxyl treatment. A significant downregulation of major histocompatibility complex (MHC) class Ib and II genes, involved in antigen presentation, was also observed. Conversely, mast cell proteases expression was upregulated. These results suggest that early treatment with a large-spectrum antibiotic deeply affects the gut barrier function at the suckling-weaning interface, a period during which the gut is challenged by an array of novel food-borne antigens.


Assuntos
Amoxicilina/farmacologia , Colo/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Controladores do Desenvolvimento/genética , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Envelhecimento , Animais , Animais Recém-Nascidos , Apresentação de Antígeno/genética , Regulação para Baixo/genética , Feminino , Intestinos/crescimento & desenvolvimento , Intestinos/microbiologia , Masculino , Mastócitos/enzimologia , Peptídeo Hidrolases/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima/genética
12.
Curr Opin Clin Nutr Metab Care ; 6(5): 581-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12913677

RESUMO

PURPOSE OF REVIEW: This paper will summarize the most recent clinical and experimental data on the effects of prebiotics in inflammatory and cancerous diseases of the large intestine. RECENT FINDINGS: Animal studies, as well as data obtained in in-vitro cell culture systems, have underlined the potential of certain prebiotics to protect against inflammatory and cancerous processes in the large intestine. Clinical trials are now in progress to assess the relevance of these promising results. The biochemical mechanisms are still incompletely deciphered, but both the promotion of lactic acid-producing bacteria and the production of short-chain fatty acids, particularly butyrate, during the fermentation of prebiotics could be key factors. SUMMARY: Enteric resident bacteria are involved in inflammatory bowel diseases and may contribute to colonic carcinogenesis. Dietary manipulation of the flora may thus represent a useful aid to prevent or to treat these diseases, and this could be a place for prebiotics. Inulin-like prebiotics have shown encouraging results in animal models, but clinical and epidemiological trials are necessary to define their efficacy in humans. In the next few years, important advances are expected in understanding the interactions between prebiotics, intestinal flora and the colonic mucosa in health and diseases, enabling the improvement of therapy as well as better nutritional handling of susceptible individuals.


Assuntos
Neoplasias do Colo/terapia , Trato Gastrointestinal/microbiologia , Doenças Inflamatórias Intestinais/terapia , Probióticos/uso terapêutico , Animais , Neoplasias do Colo/microbiologia , Modelos Animais de Doenças , Ácidos Graxos Voláteis/biossíntese , Fermentação , Humanos , Doenças Inflamatórias Intestinais/microbiologia
13.
J Clin Gastroenterol ; 37(2): 105-18, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12869879

RESUMO

The inaugural meeting of the International Scientific Association for Probiotics and Prebiotics (ISAPP) was held May 3 to May 5 2002 in London, Ontario, Canada. A group of 63 academic and industrial scientists from around the world convened to discuss current issues in the science of probiotics and prebiotics. ISAPP is a non-profit organization comprised of international scientists whose intent is to strongly support and improve the levels of scientific integrity and due diligence associated with the study, use, and application of probiotics and prebiotics. In addition, ISAPP values its role in facilitating communication with the public and healthcare providers and among scientists in related fields on all topics pertinent to probiotics and prebiotics. It is anticipated that such efforts will lead to development of approaches and products that are optimally designed for the improvement of human and animal health and well being. This article is a summary of the discussions, conclusions, and recommendations made by 8 working groups convened during the first ISAPP workshop focusing on the topics of: definitions, intestinal flora, extra-intestinal sites, immune function, intestinal disease, cancer, genetics and genomics, and second generation prebiotics.


Assuntos
Probióticos , Animais , Bifidobacterium/genética , Engenharia Genética , Genoma Bacteriano , Humanos , Intestinos/microbiologia , Lactobacillus/genética , Probióticos/uso terapêutico
14.
Proc Nutr Soc ; 62(1): 95-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12740048

RESUMO

Short-chain fatty acids (SCFA) affect local and remote motility of the gastrointestinal tract by mechanisms that are not completely understood. In the large intestine where they are produced, they inhibit peristaltic activity and may stimulate tonic activity. When present in the terminal ileum as a result of reflux of colon contents, they elicit propulsive contractions. These local motor effects could involve a neuro-hormonal sensory mechanism located in the mucosa of the terminal ileum and proximal colon. Finally, through a humoral pathway probably involving polypeptide YY release, ileal and colonic SCFA modify upper motility by inducing relaxation of the proximal stomach and lower oesophageal sphincter and reducing gastric emptying. One characteristic feature of the SCFA effects is the dose-dependency of the gastrointestinal motor responses. Indeed, the effects occur only below or above a threshold of SCFA concentration in lumen contents. One putative physiological role of the motor effects of SCFA might be to maintain the physico-chemical balance of the lumen environment in the terminal ileum and proximal colon. Another role might be to co-regulate motility of the upper intestine. The clinical relevance of these effects is unclear. However, some recent findings suggest that excessive SCFA concentrations might induce adverse effects on gastrointestinal and colonic motility and sensitivity in certain diseases such as inflammatory bowel disease and gastro-oesophageal reflux disease.


Assuntos
Bactérias/metabolismo , Ácidos Graxos Voláteis/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Ácido Láctico/farmacologia , Animais , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/fisiologia , Relação Dose-Resposta a Droga , Fermentação , Humanos , Íleo/efeitos dos fármacos , Íleo/microbiologia , Íleo/fisiologia , Peptídeo YY/metabolismo
15.
Proc Nutr Soc ; 62(1): 101-6, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12740064

RESUMO

Short-chain fatty acids (SCFA), particularly butyrate, were shown to regulate cell proliferation in vitro and in vivo. Indeed, butyrate is the major fuel for colonic epithelial cells, and it can influence cell proliferation through the release of growth factors or gastrointestinal peptides such as gastrin, or through modulation of mucosal blood flow. Lastly, SCFA can act directly on genes regulating cell proliferation, and butyrate is the main SCFA to display such an effect. Butyrate inhibits histone deacetylase, which will allow histone hyperacetylation. Such hyperacetylation leads to transcription of several genes, including p21/Cip1. Moreover, it will allow cyclin D3 hyper-expression by inhibiting its degradation. The induction of the cyclin-dependent kinase inhibitory protein p21/Cip1 accounts for cell arrest in the G1 phase of the cell cycle. However, in the absence of p21 other mechanisms are initiated, leading to inhibition of cell proliferation.


Assuntos
Ciclo Celular/efeitos dos fármacos , Colo/citologia , Ácidos Graxos Voláteis/farmacologia , Acetilação/efeitos dos fármacos , Butiratos/farmacologia , Divisão Celular/efeitos dos fármacos , Colo/efeitos dos fármacos , Ciclina D3 , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Expressão Gênica , Inibidores de Histona Desacetilases , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos
17.
J Nutr ; 133(1): 21-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12514261

RESUMO

Fructooligosaccharides (FOS) increase the growth of lactic acid bacteria (LAB) and promote butyrate and lactate production. Because of these properties, FOS may benefit intestinal inflammation. The purpose of this study was to investigate the effect of FOS on colitis in rats and determine which factors are involved. Groups of rats with intracolonic trinitrobenzene sulfonic acid (TNBS)-induced colitis received intragastric infusions of 9 g/L NaCl, 1 g/d FOS or 10(11) colony-forming units (cfu)/d LAB (Experiment 1), or intracolonic infusions of 9 g/L NaCl, butyrate, lactate or butyrate + lactate with or without 10(9.5) cfu/d LAB (Experiment 2). Each infusion was administered twice daily for 14 d. Intragastric FOS reduced the gross score for inflammation (P < 0.001), myeloperoxidase (MPO) activity (P < 0.001) and pH (P < 0.001), and increased lactate (P = 0.02) and butyrate concentrations (P < 0.001) as well as LAB counts in the cecum (P < 0.01). Intragastric LAB (10(11) cfu/d) had the same beneficial effects as FOS and modified the cecal composition similarly. High doses of intracolonic butyrate and lactate reduced the indices of inflammation (P < 0.001), whereas administration of the lower concentrations found in the colon tended to decrease (P < 0.1) the gross score for inflammation and MPO activity. Addition of LAB (10(9.5) cfu/d) to the organic acids was necessary to reproduce the significant FOS-induced effects on these variables. Thus, under the experimental conditions used, FOS reduced intestinal inflammatory activity mainly by increasing LAB counts in the intestine.


Assuntos
Butiratos/farmacologia , Colite/tratamento farmacológico , Lactatos/farmacologia , Oligossacarídeos/uso terapêutico , Análise de Variância , Animais , Butiratos/administração & dosagem , Colite/induzido quimicamente , Lactatos/administração & dosagem , Masculino , Oligossacarídeos/administração & dosagem , Peroxidase/metabolismo , Ratos , Ratos Wistar , Ácido Trinitrobenzenossulfônico/toxicidade
18.
Peptides ; 24(12): 1929-34, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15127944

RESUMO

Chronic nutritional disorders such as protein malnutrition are associated with delayed gastric emptying and increased postprandial cholecystokinin (CCK) levels. This study investigated the mechanisms involved in gastric emptying adaptation to low-protein diet. Two groups of 12 rats were adapted to a low-protein (LPD) or standard diet (SD) for 3 weeks. As compared to rats fed a SD, in rats adapted to a LPD gastric emptying was delayed, whereas postprandial CCK levels were increased. LPD enhanced antral muscle contractile response to CCK and cerulein without altering response to acetylcholine. This increased contractility was associated with up-regulation of CCK-A receptor mRNA levels in antral muscle. Our data suggest that modulation of gastric emptying after adaptation to a low-protein diet involves up-regulation of both CCK-A receptors and CCK-induced contraction of antral smooth muscle.


Assuntos
Adaptação Fisiológica , Colecistocinina/antagonistas & inibidores , Dieta com Restrição de Proteínas , Proteínas Alimentares/administração & dosagem , Esvaziamento Gástrico/efeitos dos fármacos , Animais , Peso Corporal , Colecistocinina/metabolismo , Colecistocinina/farmacologia , Proteínas Alimentares/metabolismo , Masculino , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , RNA Mensageiro/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores da Colecistocinina/efeitos dos fármacos , Receptores da Colecistocinina/metabolismo , Fatores de Tempo
19.
Gastroenterol Clin Biol ; 27(11): 987-91, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14732844

RESUMO

BACKGROUND: Leptin, a protein with a cytokine-like structure, is produced predominantly by adipocytes. It appears to play a key role in immune responses by increasing the secretion of Th1 and pro-inflammatory cytokines. As fat-wrapping is a characteristic feature of Crohn's disease (CD), and as increased leptin levels have been reported in animal models of intestinal inflammation, this study investigated whether mesenteric adipose tissue could be a source of leptin in human inflammatory bowel disease (IBD). AIM: To quantify the expression of leptin mRNA in mesenteric adipose tissue of patients with CD or ulcerative colitis (UC). METHODS: Specimens were obtained from mesenteric white adipose tissue close to healthy and inflammatory small intestine and/or colon in patients with CD or UC and, for controls, from apparently healthy mesentery of patients operated for carcinoma of the right colon. The expression of leptin mRNA was assessed by reverse transcription-competitive polymerase chain reaction. RESULTS: Leptin mRNA levels were significantly higher in mesenteric adipose tissue of CD and UC patients than in controls (P<0.05). In CD and UC, concentrations were not significantly different in mesenteric fat specimens, whether contiguous to macroscopically normal or grossly abnormal intestine. CONCLUSIONS: This study provides the first evidence of a novel abnormality of the mesentery of patients with IBD. Overexpression of leptin mRNA in mesenteric adipose tissue may contribute to (a) the inflammatory process, (b) enhancement of mesenteric TNF alpha expression in CD (as recently reported), and/or (c) the anorexia frequently reported during flares of IBD.


Assuntos
Tecido Adiposo , Regulação da Expressão Gênica , Doenças Inflamatórias Intestinais/genética , Leptina/genética , Mesentério , RNA Mensageiro/genética , Adulto , Feminino , Humanos , Masculino , RNA Mensageiro/análise
20.
Reprod Nutr Dev ; 43(4): 347-56, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14971826

RESUMO

According to Burkitt's hypothesis, dietary fibres may protect against the development of colorectal cancer. In rats, studies have shown that only butyrate-producing fibres are protective. In parallel, in humans, non-steroidal anti-inflammatory drugs, which target cyclooxygenases, have been shown to display a protective effect against colorectal cancer. Among them, COX-2-selective inhibitors which present less side effects than non-selective agents, are promising as chemopreventive agents. Our aim was to analyse the effect of an association between butyrate-producing fibres and the COX-2 inhibitor on the development of aberrant crypt foci (ACF) in rats. Fisher F344 rats were fed with (1) a standard low fibre control diet; (2) the standard diet supplemented with 1500 ppm celecoxib; (3) a diet supplemented with 6% fructo-oligosaccharide (FOS); and (4) a diet with both celecoxib and FOS. Three weeks later, the rats were injected twice with azoxymethane and the number of ACF was determined 15 weeks later. In the control group, 43.8 +/- 6.4 ACF were found. This number was not significantly modified by the addition of FOS or celecoxib alone to the diet. However, the association of FOS and celecoxib resulted in a 61% reduction in the number of ACF (P < 0.01). The number of aberrant crypt per foci was also reduced. Thus, although no significant effect of celecoxib or FOS alone was identified, the association of butyrate-producing fibre and celecoxib was effective in preventing the development of ACF. This preliminary study argues for a strong protective effect of such an association which deserves further studies.


Assuntos
Anticarcinógenos/administração & dosagem , Colo/patologia , Fibras na Dieta/administração & dosagem , Oligossacarídeos/administração & dosagem , Sulfonamidas/administração & dosagem , Animais , Azoximetano/antagonistas & inibidores , Butiratos/metabolismo , Celecoxib , Colo/efeitos dos fármacos , Colo/metabolismo , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/prevenção & controle , Inibidores de Ciclo-Oxigenase/farmacologia , Masculino , Neoplasias Experimentais , Oligossacarídeos/metabolismo , Pirazóis , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA