Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 6(6): e01453-15, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26578675

RESUMO

UNLABELLED: The aim of this study was to investigate the effect of iron (Fe) availability on butyrate production in the complex bacterial ecosystem of the human gut. Hence, different Fe availabilities were mimicked in an in vitro colonic fermentation model (the polyfermenter intestinal model called PolyFermS) inoculated with immobilized gut microbiota from a child and in batch cultures of the butyrate producer Roseburia intestinalis. Shifts in the microbial community (16S rRNA sequencing and quantitative PCR), metabolic activity (high-performance liquid chromatography), and expression of genes involved in butyrate production were assessed. In the PolyFermS, moderate Fe deficiency resulted in a 1.4-fold increase in butyrate production and a 5-fold increase in butyryl-coenzyme A (CoA):acetate CoA-transferase gene expression, while very strong Fe deficiency significantly decreased butyrate concentrations and butyrate-producing bacteria compared with the results under normal Fe conditions. Batch cultures of R. intestinalis grown in a low-Fe environment preferentially produced lactate and had reduced butyrate and hydrogen production, in parallel with upregulation of the lactate dehydrogenase gene and downregulation of the pyruvate:ferredoxin-oxidoreductase gene. In contrast, under high-Fe conditions, R. intestinalis cultures showed enhanced butyrate and hydrogen production, along with increased expression of the corresponding genes, compared with the results under normal-Fe conditions. Our data reveal the strong regulatory effect of Fe on gut microbiota butyrate producers and on the concentrations of butyrate, which contributes to the maintenance of host gut health. IMPORTANCE: Fe deficiency is one of the most common nutritional deficiencies worldwide and can be corrected by Fe supplementation. In this in vitro study, we show that environmental Fe concentrations in a continuous gut fermentation model closely mimicking a child's gut microbiota strongly affect the composition of the gut microbiome and its metabolic activity, particularly butyrate production. The differential expression of genes involved in the butyrate production pathway under different Fe conditions and the enzyme cofactor role of Fe explain the observed modulation of butyrate production. Our data reveal that the level of dietary Fe reaching the colon affects the microbiome, and its essential function of providing the host with beneficial butyrate.


Assuntos
Butiratos/metabolismo , Microbioma Gastrointestinal/fisiologia , Ferro/metabolismo , Butiratos/química , Criança , Cromatografia Líquida de Alta Pressão , Coenzima A-Transferases/genética , Coenzima A-Transferases/metabolismo , Fermentação , Microbioma Gastrointestinal/genética , Bactérias Gram-Positivas/metabolismo , Humanos , Hidrogênio/metabolismo , Ferro/química , L-Lactato Desidrogenase/genética , Piruvato Sintase/genética , RNA Ribossômico 16S/genética
2.
Gut ; 64(5): 731-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25143342

RESUMO

BACKGROUND: In-home iron fortification for infants in developing countries is recommended for control of anaemia, but low absorption typically results in >80% of the iron passing into the colon. Iron is essential for growth and virulence of many pathogenic enterobacteria. We determined the effect of high and low dose in-home iron fortification on the infant gut microbiome and intestinal inflammation. METHODS: We performed two double-blind randomised controlled trials in 6-month-old Kenyan infants (n=115) consuming home-fortified maize porridge daily for 4 months. In the first, infants received a micronutrient powder (MNP) containing 2.5 mg iron as NaFeEDTA or the MNP without iron. In the second, they received a different MNP containing 12.5 mg iron as ferrous fumarate or the MNP without the iron. The primary outcome was gut microbiome composition analysed by 16S pyrosequencing and targeted real-time PCR (qPCR). Secondary outcomes included faecal calprotectin (marker of intestinal inflammation) and incidence of diarrhoea. We analysed the trials separately and combined. RESULTS: At baseline, 63% of the total microbial 16S rRNA could be assigned to Bifidobacteriaceae but there were high prevalences of pathogens, including Salmonella Clostridium difficile, Clostridium perfringens, and pathogenic Escherichia coli. Using pyrosequencing, +FeMNPs increased enterobacteria, particularly Escherichia/Shigella (p=0.048), the enterobacteria/bifidobacteria ratio (p=0.020), and Clostridium (p=0.030). Most of these effects were confirmed using qPCR; for example, +FeMNPs increased pathogenic E. coli strains (p=0.029). +FeMNPs also increased faecal calprotectin (p=0.002). During the trial, 27.3% of infants in +12.5 mgFeMNP required treatment for diarrhoea versus 8.3% in -12.5 mgFeMNP (p=0.092). There were no study-related serious adverse events in either group. CONCLUSIONS: In this setting, provision of iron-containing MNPs to weaning infants adversely affects the gut microbiome, increasing pathogen abundance and causing intestinal inflammation. TRIAL REGISTRATION NUMBER: NCT01111864.


Assuntos
Enterocolite/induzido quimicamente , Alimentos Fortificados/efeitos adversos , Intestinos/microbiologia , Ferro da Dieta/efeitos adversos , Microbiota/efeitos dos fármacos , Anemia Ferropriva/prevenção & controle , Bactérias/isolamento & purificação , Diarreia Infantil/induzido quimicamente , Diarreia Infantil/microbiologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Enterocolite/microbiologia , Fezes/química , Humanos , Lactente , Ferro da Dieta/administração & dosagem , Ferro da Dieta/farmacologia , Complexo Antígeno L1 Leucocitário/metabolismo , Micronutrientes/administração & dosagem , Micronutrientes/efeitos adversos , Micronutrientes/farmacologia
3.
Br J Nutr ; 112(4): 547-56, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-24916165

RESUMO

Fe supplementation is a common strategy to correct Fe-deficiency anaemia in children; however, it may modify the gut microbiota and increase the risk for enteropathogenic infection. In the present study, we studied the impact of Fe supplementation on the abundance of dominant bacterial groups in the gut, faecal SCFA concentration and gut inflammation in children living in rural South Africa. In a randomised, placebo-controlled intervention trial of 38 weeks, 6- to 11-year-old children with Fe deficiency received orally either tablets containing 50 mg Fe as FeSO4 (n 22) for 4 d/week or identical placebo (n 27). In addition, Fe-sufficient children (n 24) were included as a non-treated reference group. Faecal samples were analysed at baseline and at 2, 12 and 38 weeks to determine the effects of Fe supplementation on ten bacterial groups in the gut (quantitative PCR), faecal SCFA concentration (HPLC) and gut inflammation (faecal calprotectin concentration). At baseline, concentrations of bacterial groups in the gut, faecal SCFA and faecal calprotectin did not differ between Fe-deficient and Fe-sufficient children. Fe supplementation significantly improved Fe status in Fe-deficient children and did not significantly increase faecal calprotectin concentration. Moreover, no significant effect of Fe treatment or time × treatment interaction on the concentrations of bacterial groups in the gut or faecal SCFA was observed compared with the placebo treatment. Also, there were no significant differences observed in the concentrations of any of the bacterial target groups or faecal SCFA at 2, 12 or 38 weeks between the three groups of children when correcting for baseline values. The present study suggests that in African children with a low enteropathogen burden, Fe status and dietary Fe supplementation did not significantly affect the dominant bacterial groups in the gut, faecal SCFA concentration or gut inflammation.


Assuntos
Suplementos Nutricionais/efeitos adversos , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Positivas/crescimento & desenvolvimento , Hematínicos/efeitos adversos , Mucosa Intestinal/microbiologia , Ferro da Dieta/efeitos adversos , Trato Gastrointestinal Inferior/microbiologia , Anemia Ferropriva/dietoterapia , Anemia Ferropriva/epidemiologia , Anemia Ferropriva/imunologia , Anemia Ferropriva/microbiologia , Criança , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Feminino , Compostos Ferrosos/administração & dosagem , Gastroenterite/induzido quimicamente , Gastroenterite/epidemiologia , Gastroenterite/imunologia , Gastroenterite/microbiologia , Fármacos Gastrointestinais/efeitos adversos , Fármacos Gastrointestinais/uso terapêutico , Bactérias Gram-Negativas/isolamento & purificação , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/isolamento & purificação , Bactérias Gram-Positivas/metabolismo , Hematínicos/uso terapêutico , Humanos , Incidência , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Ferro da Dieta/uso terapêutico , Complexo Antígeno L1 Leucocitário/química , Complexo Antígeno L1 Leucocitário/metabolismo , Trato Gastrointestinal Inferior/imunologia , Trato Gastrointestinal Inferior/metabolismo , Masculino , Viabilidade Microbiana , Saúde da População Rural , África do Sul/epidemiologia
4.
PLoS One ; 9(3): e93549, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24676135

RESUMO

In regions with a high infectious disease burden, concerns have been raised about the safety of iron supplementation because higher iron concentrations in the gut lumen may increase risk of enteropathogen infection. The aim of this study was to investigate interactions of the enteropathogen Salmonella enterica ssp. enterica Typhimurium with intestinal cells under different iron concentrations encountered in the gut lumen during iron deficiency and supplementation using an in vitro colonic fermentation system inoculated with immobilized child gut microbiota combined with Caco-2/HT29-MTX co-culture monolayers. Colonic fermentation effluents obtained during normal, low (chelation by 2,2'-dipyridyl) and high iron (26.5 mg iron/L) fermentation conditions containing Salmonella or pure Salmonella cultures with similar iron conditions were applied to cellular monolayers. Salmonella adhesion and invasion capacity, cellular integrity and immune response were assessed. Under high iron conditions in pure culture, Salmonella adhesion was 8-fold increased compared to normal iron conditions while invasion was not affected leading to decreased invasion efficiency (-86%). Moreover, cellular cytokines IL-1ß, IL-6, IL-8 and TNF-α secretion as well as NF-κB activation in THP-1 cells were attenuated under high iron conditions. Low iron conditions in pure culture increased Salmonella invasion correlating with an increase in IL-8 release. In fermentation effluents, Salmonella adhesion was 12-fold and invasion was 428-fold reduced compared to pure culture. Salmonella in high iron fermentation effluents had decreased invasion efficiency (-77.1%) and cellular TNF-α release compared to normal iron effluent. The presence of commensal microbiota and bacterial metabolites in fermentation effluents reduced adhesion and invasion of Salmonella compared to pure culture highlighting the importance of the gut microbiota as a barrier during pathogen invasion. High iron concentrations as encountered in the gut lumen during iron supplementation attenuated Salmonella invasion efficiency and cellular immune response suggesting that high iron concentrations alone may not lead to an increased Salmonella invasion.


Assuntos
Mucosa Intestinal/microbiologia , Ferro/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Salmonella typhimurium/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Reatores Biológicos , Células CACO-2 , Células Imobilizadas , Criança , Técnicas de Cocultura , Cultura em Câmaras de Difusão , Fermentação , Células HT29 , Humanos , Interleucina-1beta/biossíntese , Interleucina-1beta/metabolismo , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Mucosa Intestinal/imunologia , Ferro/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Microbiota/fisiologia , Modelos Biológicos , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
5.
Br J Nutr ; 111(12): 2135-45, 2014 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-24555487

RESUMO

The global prevalence of Fe deficiency is high and a common corrective strategy is oral Fe supplementation, which may affect the commensal gut microbiota and gastrointestinal health. The aim of the present study was to investigate the impact of different dietary Fe concentrations on the gut microbiota and gut health of rats inoculated with human faecal microbiota. Rats (8 weeks old, n 40) were divided into five (n 8 each) groups and fed diets differing only in Fe concentration during an Fe-depletion period (12 weeks) and an Fe-repletion period (4 weeks) as follows: (1) Fe-sufficient diet throughout the study period; (2) Fe-sufficient diet followed by 70 mg Fe/kg diet; (3) Fe-depleted diet throughout the study period; (4) Fe-depleted diet followed by 35 mg Fe/kg diet; (5) Fe-depleted diet followed by 70 mg Fe/kg diet. Faecal and caecal samples were analysed for gut microbiota composition (quantitative PCR and pyrosequencing) and bacterial metabolites (HPLC), and intestinal tissue samples were investigated histologically. Fe depletion did not significantly alter dominant populations of the gut microbiota and did not induce Fe-deficiency anaemia in the studied rats. Provision of the 35 mg Fe/kg diet after feeding an Fe-deficient diet significantly increased the abundance of dominant bacterial groups such as Bacteroides spp. and Clostridium cluster IV members compared with that of an Fe-deficient diet. Fe supplementation increased gut microbial butyrate concentration 6-fold compared with Fe depletion and did not affect histological colitis scores. The present results suggest that Fe supplementation enhances the concentration of beneficial gut microbiota metabolites and thus may contribute to gut health.


Assuntos
Bacteroides/crescimento & desenvolvimento , Clostridium/crescimento & desenvolvimento , Colite/prevenção & controle , Colo/microbiologia , Suplementos Nutricionais , Modelos Animais de Doenças , Ferro da Dieta/uso terapêutico , Animais , Bacteroides/imunologia , Bacteroides/isolamento & purificação , Bacteroides/metabolismo , Biomarcadores/metabolismo , Ácido Butírico/metabolismo , Ceco/crescimento & desenvolvimento , Ceco/imunologia , Ceco/metabolismo , Ceco/microbiologia , Criança , Clostridium/imunologia , Clostridium/isolamento & purificação , Clostridium/metabolismo , Colite/imunologia , Colite/metabolismo , Colite/microbiologia , Colo/crescimento & desenvolvimento , Colo/imunologia , Colo/metabolismo , Suplementos Nutricionais/efeitos adversos , Fezes/química , Fezes/microbiologia , Feminino , Vida Livre de Germes , Humanos , Íleo/crescimento & desenvolvimento , Íleo/imunologia , Íleo/metabolismo , Íleo/microbiologia , Imunidade nas Mucosas , Ferro/administração & dosagem , Ferro/análise , Deficiências de Ferro , Ferro da Dieta/efeitos adversos , Ferro da Dieta/análise , Ferro da Dieta/metabolismo , Ratos , Ratos Endogâmicos F344 , Aumento de Peso
6.
PLoS One ; 8(10): e77772, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204958

RESUMO

In vitro gut fermentation modeling offers a useful platform for ecological studies of the intestinal microbiota. In this study we describe a novel Polyfermentor Intestinal Model (PolyFermS) designed to compare the effects of different treatments on the same complex gut microbiota. The model operated in conditions of the proximal colon is composed of a first reactor containing fecal microbiota immobilized in gel beads, and used to continuously inoculate a set of parallel second-stage reactors. The PolyFermS model was validated with three independent intestinal fermentations conducted for 38 days with immobilized human fecal microbiota obtained from three child donors. The microbial diversity of reactor effluents was compared to donor feces using the HITChip, a high-density phylogenetic microarray targeting small subunit rRNA sequences of over 1100 phylotypes of the human gastrointestinal tract. Furthermore, the metabolic response to a decrease of pH from 5.7 to 5.5, applied to balance the high fermentative activity in inoculum reactors, was studied. We observed a reproducible development of stable intestinal communities representing major taxonomic bacterial groups at ratios similar to these in feces of healthy donors, a high similarity of microbiota composition produced in second-stage reactors within a model, and a high time stability of microbiota composition and metabolic activity over 38 day culture. For all tested models, the pH-drop of 0.2 units in inoculum reactors enhanced butyrate production at the expense of acetate, but was accompanied by a donor-specific reorganization of the reactor community, suggesting a concerted metabolic adaptation and trigger of community-specific lactate or acetate cross-feeding pathways in response to varying pH. Our data showed that the PolyFermS model allows the stable cultivation of complex intestinal microbiota akin to the fecal donor and can be developed for the direct comparison of different experimental conditions in parallel reactors continuously inoculated with the exact same microbiota.


Assuntos
Fezes/microbiologia , Trato Gastrointestinal/microbiologia , Metagenoma , Modelos Biológicos , Modelos Teóricos , Bactérias/genética , Bactérias/isolamento & purificação , Butiratos/metabolismo , Criança , DNA Bacteriano/genética , Feminino , Fermentação , Trato Gastrointestinal/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , RNA Ribossômico 16S/genética
7.
FEMS Microbiol Ecol ; 83(1): 161-75, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22845175

RESUMO

Iron (Fe) deficiency affects an estimated 2 billion people worldwide, and Fe supplements are a common corrective strategy. The impact of Fe deficiency and Fe supplementation on the complex microbial community of the child gut was studied using in vitro colonic fermentation models inoculated with immobilized fecal microbiota. Chyme media (all Fe chelated by 2,2'-dipyridyl to 26.5 mg Fe L(-1) ) mimicking Fe deficiency and supplementation were continuously fermented. Fermentation effluent samples were analyzed daily on the microbial composition and metabolites by quantitative PCR, 16S rRNA gene 454-pyrosequencing, and HPLC. Low Fe conditions (1.56 mg Fe L(-1) ) significantly decreased acetate concentrations, and subsequent Fe supplementation (26.5 mg Fe L(-1) ) restored acetate production. High Fe following normal Fe conditions had no impact on the gut microbiota composition and metabolic activity. During very low Fe conditions (0.9 mg Fe L(-1) or Fe chelated by 2,2'-dipyridyl), a decrease in Roseburia spp./Eubacterium rectale, Clostridium Cluster IV members and Bacteroides spp. was observed, while Lactobacillus spp. and Enterobacteriaceae increased consistent with a decrease in butyrate (-84%) and propionate (-55%). The strong dysbiosis of the gut microbiota together with decrease in main gut microbiota metabolites observed with very low iron conditions could weaken the barrier effect of the microbiota and negatively impact gut health.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Fermentação , Ferro/metabolismo , Metagenoma , Consórcios Microbianos , Células Imobilizadas/metabolismo , Criança , Colo/metabolismo , Meios de Cultura/química , DNA Bacteriano/genética , Fezes/microbiologia , Humanos , Deficiências de Ferro , Análise de Sequência de DNA
8.
J Nutr ; 142(2): 271-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22190022

RESUMO

Iron (Fe) deficiency anemia is a global health concern and Fe fortification and supplementation are common corrective strategies. Fe is essential not only for the human host but also for nearly all gut bacteria. We studied the impact of Fe deficiency and Fe repletion on the gut microbiota in rats. Weanling rats were fed an Fe-deficient diet for 24 d and then repleted for 13 d with FeSO4 (n = 15) or electrolytic Fe (n = 14) at 10 and 20 mg Fe · kg diet⁻¹. In addition, one group of rats (n = 8) received the Fe-deficient diet and one group (n = 3) received a Fe-sufficient control diet for all 37 d. Fecal samples were collected at baseline and after the depletion and repletion periods, and colonic tissues were examined histologically. Microbial metabolite composition in cecal water was measured and fecal samples were analyzed for microbial composition with temporal temperature gradient gel electrophoresis and qPCR. Compared to Fe-sufficient rats, Fe-deficient rats had significantly lower concentrations of cecal butyrate (-87%) and propionate (-72%) and the abundance of dominant species was strongly modified, including greater numbers of lactobacilli and Enterobacteriaceae and a large significant decrease of the Roseburia spp./E. rectale group, a major butyrate producer. Repletion with 20 mg FeSO4 · kg diet⁻¹ significantly increased cecal butyrate concentrations and partially restored bacterial populations compared to Fe-deficient rats at endpoint. The effects on the gut microbiota were stronger in rats repleted with FeSO4 than in rats repleted with electrolytic Fe, suggesting ferrous Fe may be more available for utilization by the gut microbiota than elemental Fe. Repletion with FeSO4 significantly increased neutrophilic infiltration of the colonic mucosa compared to Fe-deficient rats. In conclusion, Fe depletion and repletion strongly affect the composition and metabolic activity of rat gut microbiota.


Assuntos
Anemia Ferropriva/tratamento farmacológico , Bactérias/metabolismo , Ceco/microbiologia , Colo/microbiologia , Compostos Ferrosos/uso terapêutico , Ferro/uso terapêutico , Ração Animal , Animais , Peso Corporal , Cromatografia Líquida de Alta Pressão , Dieta , Fezes/microbiologia , Hemoglobinas , Ferro/química , Ferro da Dieta/análise , Ferro da Dieta/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley
9.
Am J Clin Nutr ; 92(6): 1406-15, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20962160

RESUMO

BACKGROUND: Iron is essential for the growth and virulence of many pathogenic enterobacteria, whereas beneficial barrier bacteria, such as lactobacilli, do not require iron. Thus, increasing colonic iron could select gut microbiota for humans that are unfavorable to the host. OBJECTIVE: The objective was to determine the effect of iron fortification on gut microbiota and gut inflammation in African children. DESIGN: In a 6-mo, randomized, double-blind, controlled trial, 6-14-y-old Ivorian children (n = 139) received iron-fortified biscuits, which contained 20 mg Fe/d, 4 times/wk as electrolytic iron or nonfortifoed biscuits. We measured changes in hemoglobin concentrations, inflammation, iron status, helminths, diarrhea, fecal calprotectin concentrations, and microbiota diversity and composition (n = 60) and the prevalence of selected enteropathogens. RESULTS: At baseline, there were greater numbers of fecal enterobacteria than of lactobacilli and bifidobacteria (P < 0.02). Iron fortification was ineffective; there were no differences in iron status, anemia, or hookworm prevalence at 6 mo. The fecal microbiota was modified by iron fortification as shown by a significant increase in profile dissimilarity (P < 0.0001) in the iron group as compared with the control group. There was a significant increase in the number of enterobacteria (P < 0.005) and a decrease in lactobacilli (P < 0.0001) in the iron group after 6 mo. In the iron group, there was an increase in the mean fecal calprotectin concentration (P < 0.01), which is a marker of gut inflammation, that correlated with the increase in fecal enterobacteria (P < 0.05). CONCLUSIONS: Anemic African children carry an unfavorable ratio of fecal enterobacteria to bifidobacteria and lactobacilli, which is increased by iron fortification. Thus, iron fortification in this population produces a potentially more pathogenic gut microbiota profile, and this profile is associated with increased gut inflammation. This trial was registered at controlled-trials.com as ISRCTN21782274.


Assuntos
Bactérias/efeitos dos fármacos , Colo/microbiologia , Alimentos Fortificados , Gastroenterite/epidemiologia , Ferro/farmacologia , Complexo Antígeno L1 Leucocitário/análise , Metagenoma/efeitos dos fármacos , Adolescente , Anemia Ferropriva/tratamento farmacológico , Bactérias/classificação , Pão , Criança , Côte d'Ivoire/epidemiologia , Método Duplo-Cego , Fezes/química , Fezes/microbiologia , Feminino , Gastroenterite/microbiologia , Humanos , Ferro/uso terapêutico , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...