Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Clin Nutr ; 115(1): 142-153, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34617558

RESUMO

BACKGROUND: Bovine milk-derived oligosaccharides (MOS) containing primarily galacto-oligosaccharides with inherent concentrations of sialylated oligosaccharides can be added to infant formula to enhance the oligosaccharide profile. OBJECTIVE: To investigate the effects of an MOS-supplemented infant formula on gut microbiota and intestinal immunity. METHODS: In a double-blind, randomized, controlled trial, healthy term formula-fed infants aged 21-26 d either received an intact protein cow milk-based formula (control group, CG, n = 112) or the same formula containing 7.2 g MOS/L (experimental group, EG, n = 114) until the age of 6 mo. Exclusively human milk-fed infants (HFI, n = 70) from an observational study served as the reference. Fecal samples collected at baseline, and the ages of 2.5 and 4 mo were assessed for microbiota (16S ribosomal RNA-based approaches), metabolites, and biomarkers of gut health and immune response. RESULTS: Aged 2.5 and 4 mo, redundancy analysis (P = 0.002) and average phylogenetic distance (P < 0.05) showed that the overall microbiota composition in EG was different from CG and closer to that of HFI. Similarly, EG caesarean-born infants were different from CG caesarean- or vaginally born infants and approaching HFI vaginally born infants. Relative bifidobacteria abundance was higher in EG compared with CG (P < 0.05) approaching HFI. At the age of 4 mo, counts of Clostridioides difficile and Clostridium perfringens were ∼90% (P < 0.001) and ∼65% (P < 0.01) lower in EG compared with CG, respectively. Geometric LS mean (95% CI) fecal secretory IgA in EG was twice that of CG [70 (57, 85) compared with 34 (28, 42) mg/g, P < 0.001] and closer to HFI. Fecal oral polio vaccine-specific IgA was ∼50% higher in EG compared with CG (P = 0.065). Compared with CG, EG and HFI had lower fecal calcium excretion (by ∼30%, P < 0.005) and fecal pH (P < 0.001), and higher lactate concentration (P < 0.001). CONCLUSIONS: Infant formula with MOS shifts the gut microbiota and metabolic signature closer to that of HFI, has a strong bifidogenic effect, reduces fecal pathogens, and improves the intestinal immune response.


Assuntos
Suplementos Nutricionais , Microbioma Gastrointestinal , Fórmulas Infantis/química , Fenômenos Fisiológicos da Nutrição do Lactente , Oligossacarídeos/administração & dosagem , Animais , Método Duplo-Cego , Fezes/microbiologia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Leite/química , Leite Humano/química , Estudos Observacionais como Assunto , Filogenia , RNA Ribossômico 16S/análise
2.
Nutrients ; 13(5)2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-34066577

RESUMO

Prevalence of anaemia among Nigerian toddlers is reported to be high, and may cause significant morbidity, affects brain development and function, and results in weakness and fatigue. Although, iron fortification can reduce anaemia, yet the effect on gut microbiota is unclear. This open-label randomised study in anaemic malnourished Nigerian toddlers aimed to decrease anaemia without affecting pathogenic gut bacteria using a multi-nutrient fortified dairy-based drink. The test product was provided daily in different amounts (200, 400 or 600 mL, supplying 2.24, 4.48 and 6.72 mg of elemental iron, respectively) for 6 months. Haemoglobin, ferritin, and C-reactive protein concentrations were measured to determine anaemia, iron deficiency (ID) and iron deficiency anaemia (IDA) prevalence. Faecal samples were collected to analyse gut microbiota composition. All three dosages reduced anaemia prevalence, to 47%, 27% and 18%, respectively. ID and IDA prevalence was low and did not significantly decrease over time. Regarding gut microbiota, Enterobacteriaceae decreased over time without differences between groups, whereas Bifidobacteriaceae and pathogenic E. coli were not affected. In conclusion, the multi-nutrient fortified dairy-based drink reduced anaemia in a dose-dependent way, without stimulating intestinal potential pathogenic bacteria, and thus appears to be safe and effective in treating anaemia in Nigerian toddlers.


Assuntos
Anemia Ferropriva/prevenção & controle , Bebidas , Transtornos da Nutrição Infantil/prevenção & controle , Compostos Ferrosos/administração & dosagem , Alimentos Fortificados , Micronutrientes/administração & dosagem , Anemia Ferropriva/epidemiologia , Anemia Ferropriva/microbiologia , Transtornos da Nutrição Infantil/epidemiologia , Transtornos da Nutrição Infantil/microbiologia , Pré-Escolar , Laticínios , Relação Dose-Resposta a Droga , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Lactente , Masculino , Nigéria/epidemiologia , Prevalência
3.
Eur J Nutr ; 60(6): 3423-3436, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33634319

RESUMO

PURPOSE: Adverse effects of iron fortification/supplements such as Micronutrient Powder (MNP) on gut microbiota have previously been found in infection-prone African settings. This study examined the adversaries of a low-iron MNP compared with the standard MNP on the composition of gut microbiota in Bangladeshi children exposed to a high concentration of iron from potable groundwater. METHODS: A randomized controlled trial was conducted in 2- to 5-year-old children, drinking groundwater with a high concentration of iron (≥ 2 mg/L). Children were randomized to receive one sachet per day of either standard MNP (12.5 mg iron) or low-iron MNP (5 mg iron), for 2 months. A sub-sample of 53 children was considered for paired assessment of the gut microbiome by 16S rRNA amplicon sequencing. RESULTS: At baseline, the gut microbiota consisted of Bifidobacteriaceae (15.6%), Prevotellaceae (12.2%), Lactobacillaceae (3.6%), Clostridiaceae (4.1%) and Enterobacteriaceae (2.8%). Overall, there was no significant treatment effect of the low-iron MNP compared to the standard MNP. However, an apparent treatment effect was observed in children with a relative adult-like microbiota, with a higher relative abundance of potentially pathogenic Enterobacteriaceae after receiving the standard MNP compared to the low-iron MNP. This effect, however, was statistically non-significant (p = 0.07). CONCLUSION: In Bangladeshi children drinking iron-rich groundwater, a low-iron MNP supplementation did not have a significant impact on their gut microbiota profile/composition compared to the standard MNP. The trial registration number is ISRCTN60058115; Date of registration 03/07/2019; retrospectively registered.


Assuntos
Anemia Ferropriva , Microbioma Gastrointestinal , Água Subterrânea , Adulto , Pré-Escolar , Suplementos Nutricionais , Humanos , Lactente , Ferro , Micronutrientes , Pós , RNA Ribossômico 16S/genética
4.
Pediatr Allergy Immunol ; 31(7): 745-754, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32426882

RESUMO

BACKGROUND: Bioactive proteins and human milk oligosaccharides (HMOs), important ingredients in breast milk, that protect against infections are lacking in young child formula (YCF). This study investigated the effects of new YCFs on respiratory and gastrointestinal infections in toddlers. METHODS: Four hundred and sixty one healthy Chinese children aged 1-2.5 years were recruited in this randomized, controlled, double-blind, parallel-group clinical trial of different YCFs. They were randomly assigned to either standard milk formula (YCF-Ref) or one of three new YCFs containing bioactive proteins and/or the HMO 2'-fucosyllactose (2'-FL) and/or milk fat for six months. Primary outcomes were incidence of upper respiratory tract infection (URTI) and duration of gastrointestinal tract infections (GITI). RESULTS: There were no significant between-group differences in primary outcomes. For secondary outcomes, subjects receiving 2'-FL-supplemented YCF had longer URTI. Subjects receiving YCF supplemented with milk fat and intact bioactive proteins, and 2'-FL at levels found in breast milk, had more GITI episodes and shorter time to first GITI but similar effects on URTI duration than YCF-Ref recipients. No effects on URTI and GITI were observed in toddlers receiving YCF with bioactive proteins at lower levels than breast milk. Occurrence of adverse events and anthropometry were similar in all groups. CONCLUSIONS: All three YCFs supplemented with different combinations of intact bioactive proteins, 2'-FL, and milk fat are safe in toddlers. No difference is detected among YCFs on URTI incidence and GITI duration. Further studies are needed to verify these findings especially in infants who may benefit most from the immune-boosting effects of bioactive proteins and HMOs.


Assuntos
Gastroenteropatias/epidemiologia , Fórmulas Infantis/química , Infecções Respiratórias/epidemiologia , Povo Asiático , Pré-Escolar , Suplementos Nutricionais , Método Duplo-Cego , Feminino , Gastroenteropatias/prevenção & controle , Humanos , Incidência , Lactente , Masculino , Leite Humano/química , Oligossacarídeos/administração & dosagem , Oligossacarídeos/química , Infecções Respiratórias/prevenção & controle , Resultado do Tratamento , Trissacarídeos/administração & dosagem , Trissacarídeos/química
5.
Nutrients ; 11(11)2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31671757

RESUMO

There is little data on human milk oligosaccharide (HMO) composition in Sub-Saharan Africa. Iron fortificants adversely affect the infant gut microbiota, while co-provision of prebiotic galacto-oligosaccharides (GOS) mitigates most of the adverse effects. Whether variations in maternal HMO profile can influence the infant response to iron and/or GOS fortificants is unknown. The aim of this study was to determine HMO profiles and the secretor/non-secretor phenotype of lactating Kenyan mothers and investigate their effects on the maternal and infant gut microbiota, and on the infant response to a fortification intervention with 5 mg iron (2.5 mg as sodium iron ethylenediaminetetraacetate and 2.5 mg as ferrous fumarate) and 7.5 g GOS. We studied mother-infant pairs (n = 80) participating in a 4-month intervention trial in which the infants (aged 6.5-9.5 months) received daily a micronutrient powder without iron, with iron or with iron and GOS. We assessed: (1) maternal secretor status and HMO composition; (2) effects of secretor status on the maternal and infant gut microbiota in a cross-sectional analysis at baseline of the intervention trial; and (3) interactions between secretor status and intervention groups during the intervention trial on the infant gut microbiota, gut inflammation, iron status, growth and infectious morbidity. Secretor prevalence was 72% and HMOs differed between secretors and non-secretors and over time of lactation. Secretor status did not predict the baseline composition of the maternal and infant gut microbiota. There was a secretor-status-by-intervention-group interaction on Bifidobacterium (p = 0.021), Z-scores for length-for-age (p = 0.022) and weight-for-age (p = 0.018), and soluble transferrin receptor (p = 0.041). In the no iron group, longitudinal prevalence of diarrhea was higher among infants of non-secretors (23.8%) than of secretors (10.4%) (p = 0.001). In conclusion, HMO profile may modulate the infant gut microbiota response to fortificant iron; compared to infants of secretor mothers, infants of non-secretor mothers may be more vulnerable to the adverse effect of iron but also benefit more from the co-provision of GOS.


Assuntos
Suplementos Nutricionais , Ferro/administração & dosagem , Micronutrientes/administração & dosagem , Leite Humano/química , Oligossacarídeos/administração & dosagem , Oligossacarídeos/química , Adulto , Bactérias/classificação , Bactérias/genética , Método Duplo-Cego , Feminino , Microbioma Gastrointestinal , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Ferro/metabolismo , Quênia , Masculino , Micronutrientes/química , Mães , Oligossacarídeos/classificação , Oligossacarídeos/metabolismo , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Adulto Jovem
6.
Hemodial Int ; 21 Suppl 1: S28-S36, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28328083

RESUMO

Patients with chronic kidney disease (CKD) and loss of kidney function are at increased risk for morbidity and mortality. The risks of CKD are attributed to "uremia," an increased concentration of uremic retention solutes (toxins) in the plasma. Recently, a colo-renal axis became clearly apparent and uremia has been associated with an altered gut microbiome composition and metabolism. There is a high prevalence of anemia in patients with CKD, for which patients are often treated with oral or intravenous iron. Recent in vivo and in vitro studies have reported adverse effects of oral iron supplementation on the gut microbiota composition, gut metabolome, and intestinal health, which in turn may result in an increased production of uremic toxins. It may also affect circulating levels of other microbe-derived molecules, that can act as mediators of immune regulation. Changes in body iron levels have also been reported to exert subtle effects on host immune function by modulating immune cell proliferation and differentiation, and by directly regulating cytokine formation and antimicrobial immune effector mechanisms. Based on the foregoing it is conceivable that oral iron supplementation in iron deficient predialysis CKD patients adversely changes gut microbiota composition, the gut and systemic metabolome, and host immunity and infection. Future studies are needed to confirm these hypotheses and to assess whether, compared to IV iron supplementation, oral iron supplementation negatively impacts on morbidity of CKD, and whether these adverse effects depend on the iron bioavailability of the iron formulation to the microbiota.


Assuntos
Suplementos Nutricionais , Microbioma Gastrointestinal , Ferro/administração & dosagem , Metaboloma , Insuficiência Renal Crônica/metabolismo , Administração Oral , Humanos , Probióticos/farmacologia , Insuficiência Renal Crônica/microbiologia
7.
Eur J Immunol ; 45(9): 2553-67, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26046550

RESUMO

Orally administrated iron is suspected to increase susceptibility to enteric infections among children in infection endemic regions. Here we investigated the effect of dietary iron on the pathology and local immune responses in intestinal infection models. Mice were held on iron-deficient, normal iron, or high iron diets and after 2 weeks they were orally challenged with the pathogen Citrobacter rodentium. Microbiome analysis by pyrosequencing revealed profound iron- and infection-induced shifts in microbiota composition. Fecal levels of the innate defensive molecules and markers of inflammation lipocalin-2 and calprotectin were not influenced by dietary iron intervention alone, but were markedly lower in mice on the iron-deficient diet after infection. Next, mice on the iron-deficient diet tended to gain more weight and to have a lower grade of colon pathology. Furthermore, survival of the nematode Caenorhabditis elegans infected with Salmonella enterica serovar Typhimurium was prolonged after iron deprivation. Together, these data show that iron limitation restricts disease pathology upon bacterial infection. However, our data also showed decreased intestinal inflammatory responses of mice fed on high iron diets. Thus additionally, our study indicates that the effects of iron on processes at the intestinal host-pathogen interface may highly depend on host iron status, immune status, and gut microbiota composition.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Infecções por Enterobacteriaceae/patologia , Mucosa Intestinal/patologia , Intestinos/patologia , Ferro da Dieta/administração & dosagem , Salmonelose Animal/metabolismo , Proteínas de Fase Aguda/biossíntese , Proteínas de Fase Aguda/imunologia , Animais , Peso Corporal/imunologia , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Citrobacter rodentium/imunologia , Dieta/métodos , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Fezes/microbiologia , Feminino , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Ferro da Dieta/efeitos adversos , Complexo Antígeno L1 Leucocitário/biossíntese , Complexo Antígeno L1 Leucocitário/imunologia , Lipocalina-2 , Lipocalinas/biossíntese , Lipocalinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/imunologia , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Salmonelose Animal/mortalidade , Salmonella typhimurium/imunologia , Análise de Sobrevida
8.
Front Microbiol ; 6: 1481, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26779139

RESUMO

Oral iron administration in African children can increase the risk for infections. However, it remains unclear to what extent supplementary iron affects the intestinal microbiome. We here explored the impact of iron preparations on microbial growth and metabolism in the well-controlled TNO's in vitro model of the large intestine (TIM-2). The model was inoculated with a human microbiota, without supplementary iron, or with 50 or 250 µmol/L ferrous sulfate, 50 or 250 µmol/L ferric citrate, or 50 µmol/L hemin. High resolution responses of the microbiota were examined by 16S rDNA pyrosequencing, microarray analysis, and metagenomic sequencing. The metabolome was assessed by fatty acid quantification, gas chromatography-mass spectrometry (GC-MS), and (1)H-NMR spectroscopy. Cultured intestinal epithelial Caco-2 cells were used to assess fecal water toxicity. Microbiome analysis showed, among others, that supplementary iron induced decreased levels of Bifidobacteriaceae and Lactobacillaceae, while it caused higher levels of Roseburia and Prevotella. Metagenomic analyses showed an enrichment of microbial motility-chemotaxis systems, while the metabolome markedly changed from a saccharolytic to a proteolytic profile in response to iron. Branched chain fatty acids and ammonia levels increased significantly, in particular with ferrous sulfate. Importantly, the metabolite-containing effluent from iron-rich conditions showed increased cytotoxicity to Caco-2 cells. Our explorations indicate that in the absence of host influences, iron induces a more hostile environment characterized by a reduction of microbes that are generally beneficial, and increased levels of bacterial metabolites that can impair the barrier function of a cultured intestinal epithelial monolayer.

9.
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
10.
FEMS Microbiol Rev ; 38(6): 1202-34, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25205464

RESUMO

Iron is abundantly present on earth, essential for most microorganisms and crucial for human health. Human iron deficiency that is nevertheless highly prevalent in developing regions of the world can be effectively treated by oral iron administration. Accumulating evidence indicates that excess of unabsorbed iron that enters the colonic lumen causes unwanted side effects at the intestinal host-microbiota interface. The chemical properties of iron, the luminal environment and host iron withdrawal mechanisms, especially during inflammation, can turn the intestine in a rather stressful milieu. Certain pathogenic enteric bacteria can, however, deal with this stress at the expense of other members of the gut microbiota, while their virulence also seems to be stimulated in an iron-rich intestinal environment. This review covers the multifaceted aspects of nutritional iron stress with respect to growth, composition, metabolism and pathogenicity of the gut microbiota in relation to human health. We aim to present an unpreceded view on the dynamic effects and impact of oral iron administration on intestinal host-microbiota interactions to provide leads for future research and other applications.


Assuntos
Intestinos/microbiologia , Ferro da Dieta/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/imunologia , Biodiversidade , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Intestinos/imunologia , Ferro/química , Ferro/metabolismo , Ferro da Dieta/metabolismo , Oxirredução , Estresse Fisiológico/efeitos dos fármacos
11.
Antimicrob Agents Chemother ; 58(3): 1664-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24379194

RESUMO

Oral iron therapy can increase the abundance of bacterial pathogens, e.g., Salmonella spp., in the large intestine of African children. Carvacrol is a natural compound with antimicrobial activity against various intestinal bacterial pathogens, among which is the highly prevalent Salmonella enterica serovar Typhimurium. This study aimed to explore a presumed interaction between carvacrol and bacterial iron handling and to assess the potential of carvacrol in preventing the increase of bacterial pathogenicity during high iron availability. S. Typhimurium was cultured with increasing concentrations of iron and carvacrol to study the effects of these combined interventions on growth, adhesion to intestinal epithelial cells, and iron uptake/influx in both bacterial and epithelial cells. In addition, the ability of carvacrol to remove iron from the high-affinity ligand transferrin and an Fe-dye complex was examined. Carvacrol retarded growth of S. Typhimurium at all iron conditions. Furthermore, iron-induced epithelial adhesion was effectively reduced by carvacrol at high iron concentrations. The reduction of growth and virulence by carvacrol was not paralleled by a change in iron uptake or influx into S. Typhimurium. In contrast, bioavailability of iron for epithelial cells was moderately decreased under these conditions. Further, carvacrol was shown to lack the properties of an iron binding molecule; however, it was able to weaken iron-ligand interactions by which it may possibly interfere with bacterial virulence. In conclusion, our in vitro data suggest that carvacrol has the potential to serve as a novel dietary supplement to prevent pathogenic overgrowth and colonization in the large intestine during oral iron therapy.


Assuntos
Antibacterianos/farmacologia , Mucosa Intestinal/microbiologia , Ferro/farmacologia , Monoterpenos/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Células CACO-2/microbiologia , Cimenos , Relação Dose-Resposta a Droga , Compostos Férricos/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Salmonella typhimurium/patogenicidade , Virulência/efeitos dos fármacos
12.
Clin Chem ; 59(3): 527-35, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23232066

RESUMO

BACKGROUND: The iron-regulating hormone hepcidin is a promising biomarker in the diagnosis of iron disorders. Concentrations of hepcidin have been shown to increase during the day in individuals who are following a regular diet. It is currently unknown whether these increases are determined by an innate rhythm or by other factors. We aimed to assess the effect of dietary iron on hepcidin concentrations during the day. METHODS: Within a 7-day interval, 32 volunteers received an iron-deficient diet on 1 day and the same diet supplemented with 65 mg ferrous fumarate at 0815 and 1145 on another day. Blood was drawn to assess ferritin, hepcidin-25, and transferrin saturation (TS) throughout both days at 4 time points between 0800 (fasted) and 1600. A linear mixed model for repeated data was used to analyze the effect of iron intake on TS and hepcidin concentrations. RESULTS: Baseline values of hepcidin at 0800 correlated significantly with ferritin (r = 0.61). During the day of an iron-deficient diet the mean TS was similar both in men and in women, whereas hepcidin increased. During the day with iron supplementation the mean TS was significantly higher both in men and in women, and the mean hepcidin was moderately but significantly higher in women (1.0 nmol/L, 95% CI, 0.2-1.8) but not in men (0.0 nmol/L, 95% CI, -0.8 to 0.8). CONCLUSIONS: Our data demonstrate that ferritin sets the basal hepcidin concentrations and suggest that innate diurnal rhythm rather than dietary iron mediates the daily hepcidin variations. These findings will be useful for optimizing sampling protocols and will facilitate the interpretation of hepcidin as an iron biomarker.


Assuntos
Peptídeos Catiônicos Antimicrobianos/sangue , Ritmo Circadiano , Ferro da Dieta/administração & dosagem , Adolescente , Adulto , Suplementos Nutricionais , Feminino , Ferritinas/sangue , Hepcidinas , Humanos , Masculino , Pessoa de Meia-Idade , Transferrina/análise
13.
PLoS One ; 7(1): e29968, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22272265

RESUMO

Recent trials have questioned the safety of untargeted oral iron supplementation in developing regions. Excess of luminal iron could select for enteric pathogens at the expense of beneficial commensals in the human gut microflora, thereby increasing the incidence of infectious diseases. The objective of the current study was to determine the effect of high iron availability on virulence traits of prevalent enteric pathogens at the host-microbe interface. A panel of enteric bacteria was cultured under iron-limiting conditions and in the presence of increasing concentrations of ferric citrate to assess the effect on bacterial growth, epithelial adhesion, invasion, translocation and epithelial damage in vitro. Translocation and epithelial integrity experiments were performed using a transwell system in which Caco-2 cells were allowed to differentiate to a tight epithelial monolayer mimicking the intestinal epithelial barrier. Growth of Salmonella typhimurium and other enteric pathogens was increased in response to iron. Adhesion of S. typhimurium to epithelial cells markedly increased when these bacteria were pre-incubated with increasing iron concentration (P = 0.0001), whereas this was not the case for the non-pathogenic Lactobacillus plantarum (P = 0.42). Cellular invasion and epithelial translocation of S. typhimurium followed the trend of increased adhesion. Epithelial damage was increased upon incubation with S. typhimurium or Citrobacter freundii that were pre-incubated under iron-rich conditions. In conclusion, our data fit with the consensus that oral iron supplementation is not without risk as iron could, in addition to inducing pathogenic overgrowth, also increase the virulence of prevalent enteric pathogens.


Assuntos
Mucosa Intestinal/metabolismo , Ferro/metabolismo , Salmonella typhimurium/crescimento & desenvolvimento , Aderência Bacteriana/efeitos dos fármacos , Translocação Bacteriana/efeitos dos fármacos , Disponibilidade Biológica , Células CACO-2 , Citrobacter freundii/crescimento & desenvolvimento , Citrobacter freundii/fisiologia , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/fisiologia , Epitélio/metabolismo , Epitélio/microbiologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/fisiologia , Compostos Férricos/farmacologia , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Ferro da Dieta/administração & dosagem , Ferro da Dieta/metabolismo , Ferro da Dieta/farmacocinética , Lactobacillus plantarum/crescimento & desenvolvimento , Lactobacillus plantarum/fisiologia , Salmonella typhimurium/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA