Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Microbiol ; 18(1): 110, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30189859

ABSTRACT

BACKGROUND: The effects of gut microbiota on human traits are expected to be small to moderate and adding the complexity of the human diseases, microbiome research demands big sample sizes. Fecal samples for such studies are mostly self-collected by participants at home. This imposes an extra level of complexity as sample collection and storage can be challenging. Effective, low-burden collection and storage methods allowing fecal samples to be transported properly and ensuring optimal quality and quantity of bacterial DNA for upstream analyses are necessary. Moreover, accurate assessment of the microbiome composition also depends on bacterial DNA extraction method. The aim of this study was to evaluate the reliability and efficiency of the OMNIgene•GUT kit as a participant-fecal friendly collection method (storage at room temperature for 24 h (O24h) or 7 days (O7d)) in comparison to the standard collection method (Fresh, storage at 4 °C for less than 24 h) in terms of amount of variability and information content accounting for two common DNA extraction methods. RESULTS: Fourteen fecal samples were collected from healthy individuals (7 males, 7 females). Collection and storage methods did not differ significantly in terms of DNA concentration and Shannon diversity index. Phylum relative abundance showed significant differences for Bacteroidetes, Actinobacteria and Cyanobacteria. The differences were observed between control (Fresh) and O24h methods, but not between Fresh and O7d. These differences were not seen when performing bacterial DNA quantification based on three bacterial groups: Bacteroides spp., Bifidobacterium spp. and Clostridium cluster IV, which represent three major phyla: Bacteroidetes, Actinobacteria and Firmicutes respectively. The two DNA extraction methods differ in terms of DNA quantity, quality, bacterial diversity and bacterial relative abundance. Furthermore, principal component analysis revealed differences in microbial structure, which are driven by the DNA extraction methods more than the collection/storage methods. CONCLUSION: Our results have highlighted the potential of using the OMNIgene•GUT kit for collection and storage at ambient temperature, which is convenient for studies aiming to collect large samples by giving participants the possibility to send samples by post. Importantly, we revealed that the choice of DNA extraction method have an impact on the microbiome profiling.


Subject(s)
Bacteria/isolation & purification , Feces/microbiology , Gastrointestinal Microbiome , Specimen Handling/methods , Bacteria/classification , Bacteria/genetics , DNA, Bacterial/genetics , Female , Humans , Male , RNA, Ribosomal, 16S/genetics , Sample Size
2.
Gut ; 66(11): 1956-1967, 2017 11.
Article in English | MEDLINE | ID: mdl-28774885

ABSTRACT

OBJECTIVE: Iron-containing micronutrient powders (MNPs) reduce anaemia in African infants, but the current high iron dose (12.5 mg/day) may decrease gut Bifidobacteriaceae and Lactobacillaceae, and increase enteropathogens, diarrhoea and respiratory tract infections (RTIs). We evaluated the efficacy and safety of a new MNP formula with prebiotic galacto-oligosaccharides (GOS) combined with a low dose (5 mg/day) of highly bioavailable iron. DESIGN: In a 4-month, controlled, double-blind trial, we randomised Kenyan infants aged 6.5-9.5 months (n=155) to receive daily (1) a MNP without iron (control); (2) the identical MNP but with 5 mg iron (2.5 mg as sodium iron ethylenediaminetetraacetate and 2.5 mg as ferrous fumarate) (Fe group); or (3) the identical MNP as the Fe group but with 7.5 g GOS (FeGOS group). RESULTS: Anaemia decreased by ≈50% in the Fe and FeGOS groups (p<0.001). Compared with the control or FeGOS group, in the Fe group there were (1) lower abundances of Bifidobacterium and Lactobacillus and higher abundances of Clostridiales (p<0.01); (2) higher abundances of virulence and toxin genes (VTGs) of pathogens (p<0.01); (3) higher plasma intestinal fatty acid-binding protein (a biomarker of enterocyte damage) (p<0.05); and (4) a higher incidence of treated RTIs (p<0.05). In contrast, there were no significant differences in these variables comparing the control and FeGOS groups, with the exception that the abundance of VTGs of all pathogens was significantly lower in the FeGOS group compared with the control and Fe groups (p<0.01). CONCLUSION: A MNP containing a low dose of highly bioavailable iron reduces anaemia, and the addition of GOS mitigates most of the adverse effects of iron on the gut microbiome and morbidity in African infants. TRIAL REGISTRATION NUMBER: NCT02118402.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Ferric Compounds/adverse effects , Ferrous Compounds/adverse effects , Gastrointestinal Microbiome/drug effects , Micronutrients/adverse effects , Oligosaccharides , Prebiotics , Double-Blind Method , Edetic Acid/adverse effects , Edetic Acid/therapeutic use , Female , Ferric Compounds/therapeutic use , Ferrous Compounds/therapeutic use , Humans , Infant , Kenya , Male , Micronutrients/therapeutic use , Oligosaccharides/administration & dosage , Prebiotics/administration & dosage , Prebiotics/microbiology
3.
Eur J Immunol ; 45(9): 2553-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26046550

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans/drug effects , Enterobacteriaceae Infections/pathology , Intestinal Mucosa/pathology , Intestines/pathology , Iron, Dietary/administration & dosage , Salmonella Infections, Animal/metabolism , Acute-Phase Proteins/biosynthesis , Acute-Phase Proteins/immunology , Animals , Body Weight/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Citrobacter rodentium/immunology , Diet/methods , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Feces/microbiology , Female , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Iron, Dietary/adverse effects , Leukocyte L1 Antigen Complex/biosynthesis , Leukocyte L1 Antigen Complex/immunology , Lipocalin-2 , Lipocalins/biosynthesis , Lipocalins/immunology , Mice , Mice, Inbred C57BL , Oncogene Proteins/biosynthesis , Oncogene Proteins/immunology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella typhimurium/immunology , Survival Analysis
4.
Gut ; 64(5): 731-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25143342

ABSTRACT

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.


Subject(s)
Enterocolitis/chemically induced , Food, Fortified/adverse effects , Intestines/microbiology , Iron, Dietary/adverse effects , Microbiota/drug effects , Anemia, Iron-Deficiency/prevention & control , Bacteria/isolation & purification , Diarrhea, Infantile/chemically induced , Diarrhea, Infantile/microbiology , Dose-Response Relationship, Drug , Double-Blind Method , Enterocolitis/microbiology , Feces/chemistry , Humans , Infant , Iron, Dietary/administration & dosage , Iron, Dietary/pharmacology , Leukocyte L1 Antigen Complex/metabolism , Micronutrients/administration & dosage , Micronutrients/adverse effects , Micronutrients/pharmacology
5.
Int J Syst Evol Microbiol ; 64(Pt 5): 1600-1616, 2014 May.
Article in English | MEDLINE | ID: mdl-24480908

ABSTRACT

A Gram-positive staining, rod-shaped, non-motile, spore-forming obligately anaerobic bacterium, designated CRIBT, was isolated from the gastro-intestinal tract of a rat and characterized. The major cellular fatty acids of strain CRIBT were saturated and unsaturated straight-chain C12-C19 fatty acids, with C16:0 being the predominant fatty acid. The polar lipid profile comprised six glycolipids, four phospholipids and one lipid that did not stain with any of the specific spray reagents used. The only quinone was MK-6. The predominating cell-wall sugars were glucose and galactose. The peptidoglycan type of strain CRIBT was A1σ lanthionine-direct. The genomic DNA G+C content of strain CRIBT was 28.1 mol%. On the basis of 16S rRNA gene sequence similarity, strain CRIBT was most closely related to a number of species of the genus Clostridium, including Clostridium lituseburense (97.2%), Clostridium glycolicum (96.2%), Clostridium mayombei (96.2%), Clostridium bartlettii (96.0%) and Clostridium irregulare (95.5%). All these species show very low 16S rRNA gene sequence similarity (<85%) to the type strain of Clostridium butyricum, the type species of the genus Clostridium. DNA-DNA hybridization with closely related reference strains indicated reassociation values below 32%. On the basis of phenotypic and genetic studies, a novel genus, Romboutsia gen. nov., is proposed. The novel isolate CRIBT (=DSM 25109T=NIZO 4048T) is proposed as the type strain of the type species, Romboutsia ilealis gen. nov., sp. nov., of the proposed novel genus. It is proposed that C. lituseburense is transferred to this genus as Romboutsia lituseburensis comb. nov. Furthermore, the reclassification into novel genera is proposed for C. bartlettii, as Intestinibacter bartlettii gen. nov., comb. nov. (type species of the genus), C. glycolicum, as Terrisporobacter glycolicus gen. nov., comb. nov. (type species of the genus), C. mayombei, as Terrisporobacter mayombei gen. nov., comb. nov., and C. irregulare, as Asaccharospora irregularis gen. nov., comb. nov. (type species of the genus), on the basis of additional data collected in this study. In addition, an emendation of the species Peptostreptococcus anaerobius and the order Eubacteriales is provided.


Subject(s)
Gram-Positive Endospore-Forming Rods/classification , Ileum/microbiology , Phylogeny , Rats, Sprague-Dawley/microbiology , Animals , Bacterial Typing Techniques , Base Composition , DNA, Bacterial/genetics , Fatty Acids/chemistry , Glycolipids/chemistry , Gram-Positive Endospore-Forming Rods/genetics , Gram-Positive Endospore-Forming Rods/isolation & purification , Molecular Sequence Data , Nucleic Acid Hybridization , Peptidoglycan/chemistry , Phospholipids/chemistry , RNA, Ribosomal, 16S/genetics , Rats , Sequence Analysis, DNA , Vitamin K 2/analogs & derivatives , Vitamin K 2/chemistry
7.
Nutrients ; 15(22)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38004196

ABSTRACT

Microbiota colonization and development in early life is impacted by various host intrinsic (genetic) factors, but also diet, lifestyle, as well as environmental and residential factors upon and after birth. To characterize the impact of maternal nutrition and environmental factors on vaginally born infant gut microbiota composition, we performed an observational study in five distinct geographical areas in Vietnam. Fecal samples of infants (around 39 days old) and fecal and breast milk samples of their mothers (around 28 years) were collected. The microbiota composition of all samples was analyzed by 16S rRNA gene Illumina sequencing and a bioinformatics workflow based on QIIME. In addition, various breast milk components were determined. Strong associations between the geographically determined maternal diet and breast milk composition as well as infant fecal microbiota were revealed. Most notable was the association of urban Ha Noi with relatively high abundances of taxa considered pathobionts, such as Klebsiella and Citrobacter, at the expense of Bifidobacterium. Breast milk composition was most distinct in rural Ha Long Bay, characterized by higher concentrations of, e.g., docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), selenium, and vitamin B12, while it was characterized by, e.g., iron, zinc, and α-linolenic acid (ALA) in Ha Noi. Breast milk iron levels were positively associated with infant fecal Klebsiella and negatively with Bifidobacterium, while the EPA and DHA levels were positively associated with Bifidobacterium. In conclusion, differences between five regions in Vietnam with respect to both maternal breast milk and infant gut microbiota composition were revealed, most likely in part due to maternal nutrition. Thus, there could be opportunities to beneficially steer infant microbiota development in a more desired (rural instead of urban) direction through the mother's diet.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Female , Humans , Infant , Milk, Human/microbiology , Mothers , Gastrointestinal Microbiome/genetics , RNA, Ribosomal, 16S/genetics , Vietnam , Docosahexaenoic Acids , Iron , Breast Feeding , Feces/microbiology
8.
Microorganisms ; 11(9)2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37764197

ABSTRACT

BACKGROUND: The intestinal microbiota plays an important role in the etiology of obesity. Sleeve gastrectomy (SG) is a frequently performed and effective therapy for morbid obesity. OBJECTIVE: To investigate the effect of sleeve gastrectomy on the fecal microbiota of individuals with morbid obesity and to examine whether shifts in microbiota composition are associated with markers of inflammation and intestinal barrier function. METHODS: Fecal and blood samples of healthy individuals (n = 27) and morbidly obese individuals pre-SG (n = 24), and at 2 months (n = 13) and 6 months post-SG (n = 9) were collected. The 16SrRNA gene was sequenced to assess microbiota composition. Fecal calprotectin, plasma inflammatory markers and intestinal permeability markers (multi-sugar test) were determined. RESULTS: Fecal microbiota composition between morbidly obese and lean individuals was significantly different. The fecal microbiota composition changed significantly 2 and 6 months post-SG (p = 0.008) compared to pre-SG but not towards a more lean profile. The post-SG microbiota profile was characterized by an increase in facultative anaerobic bacteria, characteristic for the upper gastrointestinal tract. No correlations were found between inflammatory markers, intestinal permeability and microbial profile changes. CONCLUSIONS: Fecal microbiota composition in morbidly obese individuals changed significantly following SG. This change might be explained by functional changes induced by the SG procedure.

9.
Appl Environ Microbiol ; 77(21): 7749-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21926217

ABSTRACT

Prophylactic probiotic therapy has shown beneficial effects in an experimental rat model for acute pancreatitis on the health status of the animals. Mechanisms by which probiotic therapy interferes with severity of acute pancreatitis and associated sepsis, however, are poorly understood. The aims of this study were to identify the probiotic-induced changes in the gut microbiota and to correlate these changes to disease outcome. Duodenum and ileum samples were obtained from healthy and diseased rats subjected to pancreatitis for 7 days and prophylactically treated with either a multispecies probiotic mixture or a placebo. Intestinal microbiota was characterized by terminal-restriction fragment length polymorphism (T-RFLP) analyses of PCR-amplified 16S rRNA gene fragments. These analyses showed that during acute pancreatitis the host-specific ileal microbiota was replaced by an "acute pancreatitis-associated microbiota." This replacement was not reversed by administration of the probiotic mixture. An increase, however, was observed in the relative abundance of a novel bacterial phylotype most closely related to Clostridium lituseburense and referred to as commensal rat ileum bacterium (CRIB). Specific primers targeting the CRIB 16S rRNA gene sequence were developed to detect this phylotype by quantitative PCR. An ileal abundance of CRIB 16S rRNA genes of more than 7.5% of the total bacterial 16S rRNA gene pool was correlated with reduced duodenal bacterial overgrowth, reduced bacterial translocation to remote organs, improved pancreas pathology, and reduced proinflammatory cytokine levels in plasma. Our current findings and future studies involving this uncharacterized bacterial phylotype will contribute to unraveling one of the potential mechanisms of probiotic therapy.


Subject(s)
Biodiversity , Biological Therapy/methods , Clostridium/classification , Gastrointestinal Tract/microbiology , Pancreatitis, Acute Necrotizing/complications , Probiotics/administration & dosage , Sepsis/prevention & control , Animals , Clostridium/genetics , Clostridium/isolation & purification , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Duodenum/microbiology , Ileum/microbiology , Molecular Sequence Data , Phylogeny , Polymorphism, Restriction Fragment Length , RNA, Ribosomal, 16S/genetics , Rats , Sequence Analysis, DNA
10.
Ann Surg ; 250(5): 712-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19801929

ABSTRACT

OBJECTIVES: To determine the relation between intestinal barrier dysfunction, bacterial translocation, and clinical outcome in patients with predicted severe acute pancreatitis and the influence of probiotics on these processes. SUMMARY OF BACKGROUND DATA: Randomized, placebo-controlled, multicenter trial on probiotic prophylaxis (Ecologic 641) in patients with predicted severe acute pancreatitis (PROPATRIA). METHODS: Excretion of intestinal fatty acid binding protein (IFABP, a parameter for enterocyte damage), recovery of polyethylene glycols (PEGs, a parameter for intestinal permeability), and excretion of nitric oxide (NOx, a parameter for bacterial translocation) were assessed in urine of 141 patients collected 24 to 48 h after start of probiotic or placebo treatment and 7 days thereafter. RESULTS: IFABP concentrations in the first 72 hours were higher in patients who developed bacteremia (P = 0.03), infected necrosis (P = 0.01), and organ failure (P = 0.008). PEG recovery was higher in patients who developed bacteremia (PEG 4000, P = 0.001), organ failure (PEG 4000, P < 0.0001), or died (PEG 4000, P = 0.009). Probiotic prophylaxis was associated with an increase in IFABP (median 362 vs. 199 pg/mL; P = 0.02), most evidently in patients with organ failure (P = 0.001), and did not influence intestinal permeability. Overall, probiotics decreased NOx (P = 0.05) but, in patients with organ failure, increased NOx (P = 0.001). CONCLUSIONS: Bacteremia, infected necrosis, organ failure, and mortality were all associated with intestinal barrier dysfunction early in the course of acute pancreatitis. Overall, prophylaxis with this specific combination of probiotic strains reduced bacterial translocation, but was associated with increased bacterial translocation and enterocyte damage in patients with organ failure.


Subject(s)
Bacterial Translocation , Intestines/physiopathology , Pancreatitis/microbiology , Pancreatitis/physiopathology , Probiotics/therapeutic use , Acute Disease , Bacteremia/complications , Bacteremia/diagnosis , Bacteremia/prevention & control , Double-Blind Method , Enterocytes/pathology , Fatty Acid-Binding Proteins/urine , Humans , Intestines/pathology , Nitric Oxide/urine , Pancreatitis/pathology , Permeability , Polyethylene Glycols/pharmacokinetics
11.
Lancet ; 371(9613): 651-659, 2008 02 23.
Article in English | MEDLINE | ID: mdl-18279948

ABSTRACT

BACKGROUND: Infectious complications and associated mortality are a major concern in acute pancreatitis. Enteral administration of probiotics could prevent infectious complications, but convincing evidence is scarce. Our aim was to assess the effects of probiotic prophylaxis in patients with predicted severe acute pancreatitis. METHODS: In this multicentre randomised, double-blind, placebo-controlled trial, 298 patients with predicted severe acute pancreatitis (Acute Physiology and Chronic Health Evaluation [APACHE II] score > or =8, Imrie score > or =3, or C-reactive protein >150 mg/L) were randomly assigned within 72 h of onset of symptoms to receive a multispecies probiotic preparation (n=153) or placebo (n=145), administered enterally twice daily for 28 days. The primary endpoint was the composite of infectious complications--ie, infected pancreatic necrosis, bacteraemia, pneumonia, urosepsis, or infected ascites--during admission and 90-day follow-up. Analyses were by intention to treat. This study is registered, number ISRCTN38327949. FINDINGS: One person in each group was excluded from analyses because of incorrect diagnoses of pancreatitis; thus, 152 individuals in the probiotics group and 144 in the placebo group were analysed. Groups were much the same at baseline in terms of patients' characteristics and disease severity. Infectious complications occurred in 46 (30%) patients in the probiotics group and 41 (28%) of those in the placebo group (relative risk 1.06, 95% CI 0.75-1.51). 24 (16%) patients in the probiotics group died, compared with nine (6%) in the placebo group (relative risk 2.53, 95% CI 1.22-5.25). Nine patients in the probiotics group developed bowel ischaemia (eight with fatal outcome), compared with none in the placebo group (p=0.004). INTERPRETATION: In patients with predicted severe acute pancreatitis, probiotic prophylaxis with this combination of probiotic strains did not reduce the risk of infectious complications and was associated with an increased risk of mortality. Probiotic prophylaxis should therefore not be administered in this category of patients.


Subject(s)
Pancreatitis/prevention & control , Probiotics/therapeutic use , Acute Disease , Adult , Aged , Aged, 80 and over , Double-Blind Method , Female , Humans , Logistic Models , Male , Middle Aged , Netherlands , Pancreatitis/complications , Placebos , Statistics, Nonparametric , Treatment Outcome
12.
Microbiome ; 6(1): 10, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29325581

ABSTRACT

BACKGROUND: While almost all infants are infected with respiratory syncytial virus (RSV) before the age of 2 years, only a small percentage develops severe disease. Previous studies suggest that the nasopharyngeal microbiome affects disease development. We therefore studied the effect of the nasopharyngeal microbiome on viral load and mucosal cytokine responses, two important factors influencing the pathophysiology of RSV disease. To determine the relation between (i) the microbiome of the upper respiratory tract, (ii) viral load, and (iii) host mucosal inflammation during an RSV infection, nasopharyngeal microbiota profiles of RSV infected infants (< 6 months) with different levels of disease severity and age-matched healthy controls were determined by 16S rRNA marker gene sequencing. The viral load was measured using qPCR. Nasopharyngeal CCL5, CXCL10, MMP9, IL6, and CXCL8 levels were determined with ELISA. RESULTS: Viral load in nasopharyngeal aspirates of patients associates significantly to total nasopharyngeal microbiota composition. Healthy infants (n = 21) and RSV patients (n = 54) display very distinct microbial patterns, primarily characterized by a loss in commensals like Veillonella and overrepresentation of opportunistic organisms like Haemophilus and Achromobacter in RSV-infected individuals. Furthermore, nasopharyngeal microbiota profiles are significantly different based on CXCL8 levels. CXCL8 is a chemokine that was previously found to be indicative for disease severity and for which we find Haemophilus abundance as the strongest predictor for CXCL8 levels. CONCLUSIONS: The nasopharyngeal microbiota in young infants with RSV infection is marked by an overrepresentation of the genus Haemophilus. We present that this bacterium is associated with viral load and mucosal CXCL8 responses, both which are involved in RSV disease pathogenesis.


Subject(s)
Haemophilus/classification , Interleukin-8/metabolism , Nasopharynx/microbiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Female , Haemophilus/genetics , Haemophilus/isolation & purification , Hospitalization , Humans , Infant , Infant, Newborn , Male , RNA, Ribosomal, 16S/genetics , Respiratory Syncytial Virus Infections/immunology , Sequence Analysis, DNA/methods , Up-Regulation , Viral Load
13.
Nutr Rev ; 65(11): 469-89, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18038940

ABSTRACT

Probiotics and prebiotics have long been appreciated for their positive influences on gut health. Research on the mechanisms and effects of these agents shows that their impact reaches beyond the intestine. Effects on the microecology and pathology of the oral cavity, stomach, and vaginal tract have been observed. Likely mediated through immune influences, systemic effects such as reduced severity of colds or other respiratory conditions, impact on allergy incidence and symptoms, and reduced absences from work or daycare have also been noted. These observations, among others, suggest a broader spectrum of influence than commonly considered for these unique substances.


Subject(s)
Digestive System/microbiology , Immune System/microbiology , Probiotics , Respiratory System/microbiology , Urogenital System/microbiology , Animals , Chickens , Dietary Supplements/microbiology , Digestive System/immunology , Female , Humans , Hypersensitivity , Immune System/immunology , Male , Randomized Controlled Trials as Topic , Rats , Respiratory System/immunology , Urogenital System/immunology
14.
Surgery ; 141(4): 470-80, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17383524

ABSTRACT

BACKGROUND: Infection of pancreatic necrosis by gut bacteria is a major cause of morbidity and mortality in patients with severe acute pancreatitis. Use of prophylactic antibiotics remains controversial. The aim of this experiment was assess if modification of intestinal flora with specifically designed multispecies probiotics reduces bacterial translocation or improves outcome in a rat model of acute pancreatitis. METHODS: Male Sprague-Dawley rats were allocated into 3 groups: (1) controls (sham-operated, no treatment), (2) pancreatitis and placebo, and (3) pancreatitis and probiotics. Acute pancreatitis was induced by intraductal glycodeoxycholate and intravenous cerulein infusion. Daily probiotics or placebo was administered intragastrically from 5 days prior until 7 days after induction of pancreatitis. Tissue and fluid samples were collected for microbiologic and quantitative real-time PCR analysis of bacterial translocation. RESULTS: Probiotics reduced duodenal bacterial overgrowth of potential pathogens (Log(10) colony-forming units [CFU]/g 5.0 +/- 0.7 [placebo] vs 3.5 +/- 0.3 CFU/g [probiotics], P < .05), resulting in reduced bacterial translocation to extraintestinal sites, including the pancreas (5.38 +/- 1.0 CFU/g [placebo] vs 3.1 +/- 0.5 CFU/g [probiotics], P < .05). Accordingly, health scores were better and late phase mortality was reduced: 27% (4/15, placebo) versus 0% (0/13, probiotics), respectively, P < .05. CONCLUSIONS: This experiment supports the hypothesis that modification of intestinal flora with multispecies probiotics results in reduced bacterial translocation, morbidity, and mortality in the course of experimental acute pancreatitis.


Subject(s)
Bacterial Translocation/drug effects , Bifidobacterium , Lactobacillus , Pancreatitis, Acute Necrotizing/therapy , Probiotics/therapeutic use , Animals , Duodenum/microbiology , Male , Pancreatitis, Acute Necrotizing/microbiology , Probiotics/pharmacology , Rats , Rats, Sprague-Dawley
15.
Clin Nutr ; 26(4): 450-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17544549

ABSTRACT

BACKGROUND & AIMS: Although the potential for probiotics is investigated in an increasing variety of diseases, there is little or no consensus regarding the desired probiotic properties for a particular disease in question, nor about the final design of the probiotic. Specific strain selection procedures were undertaken to design a disease-specific multispecies probiotic. METHODS: From a strain collection of 69 different lactic acid bacteria a primary selection was made of 14 strains belonging to different species showing superior survival in a simulated gastrointestinal environment. Functional tests like antimicrobial activity against a range of clinical isolates and cytokine inducing capacity in cultured human peripheral blood mononuclear cells were used to further identify potential strains. RESULTS: Specific strains inhibited growth of clinical isolates whereas others superiorly induced the anti-inflammatory cytokine IL-10. Based on functional tests and general criteria regarding probiotic design and safety, a selection of the following six strains was made (Ecologic 641); Bifidobacterium bifidum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus salivarius and Lactococcus lactis. Combination of these strains resulted in a wider antimicrobial spectrum, superior induction of IL-10 and silencing of pro-inflammatory cytokines as compared to the individual components. CONCLUSIONS: Application of strict criteria during the design of a disease-specific probiotic prior to implementation in clinical trials may provide a rational basis for use of probiotics.


Subject(s)
Antibiosis , Bifidobacterium/physiology , Cytokines/biosynthesis , Inflammation/prevention & control , Lactobacillus/physiology , Probiotics , Bifidobacterium/growth & development , Bifidobacterium/immunology , Coculture Techniques , Critical Illness/therapy , Cytokines/immunology , Humans , Lactobacillus/growth & development , Lactobacillus/immunology , Species Specificity , Time Factors , Tissue Culture Techniques
16.
Sci Rep ; 7(1): 8327, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827640

ABSTRACT

The establishment of the infant gut microbiota is a highly dynamic process dependent on extrinsic and intrinsic factors. We characterized the faecal microbiota of 4 breastfed infants and 4 formula-fed infants at 17 consecutive time points during the first 12 weeks of life. Microbiota composition was analysed by a combination of 16S rRNA gene sequencing and quantitative PCR (qPCR). In this dataset, individuality was a major driver of microbiota composition (P = 0.002) and was more pronounced in breastfed infants. A developmental signature could be distinguished, characterized by sequential colonisation of i) intrauterine/vaginal birth associated taxa, ii) skin derived taxa and other typical early colonisers such as Streptococcus and Enterobacteriaceae, iii) domination of Bifidobacteriaceae, and iv) the appearance of adultlike taxa, particularly species associated with Blautia, Eggerthella, and the potential pathobiont Clostridium difficile. Low abundance of potential pathogens was detected by 16S profiling and confirmed by qPCR. Incidence and dominance of skin and breast milk associated microbes were increased in the gut microbiome of breastfed infants compared to formula-fed infants. The approaches in this study indicate that microbiota development of breastfed and formula-fed infants proceeds according to similar developmental stages with microbiota signatures that include stage-specific species.


Subject(s)
Breast Feeding , Feces/microbiology , Infant Formula , Intestines/microbiology , Microbiota/physiology , Bacteria/classification , Bacteria/genetics , Female , Humans , Infant , Infant, Newborn , Male , Microbiota/genetics , Milk, Human/microbiology , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
17.
PLoS One ; 12(9): e0183509, 2017.
Article in English | MEDLINE | ID: mdl-28863139

ABSTRACT

BACKGROUND: Microorganisms in the human intestine (i.e. the gut microbiome) have an increasingly recognized impact on human health, including brain functioning. Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder associated with abnormalities in dopamine neurotransmission and deficits in reward processing and its underlying neuro-circuitry including the ventral striatum. The microbiome might contribute to ADHD etiology via the gut-brain axis. In this pilot study, we investigated potential differences in the microbiome between ADHD cases and undiagnosed controls, as well as its relation to neural reward processing. METHODS: We used 16S rRNA marker gene sequencing (16S) to identify bacterial taxa and their predicted gene functions in 19 ADHD and 77 control participants. Using functional magnetic resonance imaging (fMRI), we interrogated the effect of observed microbiome differences in neural reward responses in a subset of 28 participants, independent of diagnosis. RESULTS: For the first time, we describe gut microbial makeup of adolescents and adults diagnosed with ADHD. We found that the relative abundance of several bacterial taxa differed between cases and controls, albeit marginally significant. A nominal increase in the Bifidobacterium genus was observed in ADHD cases. In a hypothesis-driven approach, we found that the observed increase was linked to significantly enhanced 16S-based predicted bacterial gene functionality encoding cyclohexadienyl dehydratase in cases relative to controls. This enzyme is involved in the synthesis of phenylalanine, a precursor of dopamine. Increased relative abundance of this functionality was significantly associated with decreased ventral striatal fMRI responses during reward anticipation, independent of ADHD diagnosis and age. CONCLUSIONS: Our results show increases in gut microbiome predicted function of dopamine precursor synthesis between ADHD cases and controls. This increase in microbiome function relates to decreased neural responses to reward anticipation. Decreased neural reward anticipation constitutes one of the hallmarks of ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/complications , Attention Deficit Disorder with Hyperactivity/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Reward , Adolescent , Adult , Bifidobacterium/isolation & purification , Cohort Studies , Female , Gastrointestinal Diseases/complications , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Models, Neurological , Pilot Projects , Prephenate Dehydratase/metabolism , RNA, Ribosomal, 16S/genetics , Treatment Outcome , Young Adult
18.
Mol Nutr Food Res ; 61(1)2017 01.
Article in English | MEDLINE | ID: mdl-27129739

ABSTRACT

SCOPE: The long-lasting consequences of nutritional programming during the early phase of life have become increasingly evident. The effects of maternal nutrition on the developing intestine are still underexplored. METHODS AND RESULTS: In this study, we observed (1) altered microbiota composition of the colonic luminal content, and (2) differential gene expression in the intestinal wall in 2-week-old mouse pups born from dams exposed to a Western-style (WS) diet during the perinatal period. A sexually dimorphic effect was found for the differentially expressed genes in the offspring of WS diet-exposed dams but no differences between male and female pups were found for the microbiota composition. Integrative analysis of the microbiota and gene expression data revealed that the maternal WS diet independently affected gene expression and microbiota composition. However, the abundance of bacterial families not affected by the WS diet (Bacteroidaceae, Porphyromonadaceae, and Lachnospiraceae) correlated with the expression of genes playing a key role in intestinal development and functioning (e.g. Pitx2 and Ace2). CONCLUSION: Our data reveal that maternal consumption of a WS diet during the perinatal period alters both gene expression and microbiota composition in the intestinal tract of 2-week-old offspring.


Subject(s)
Diet, Western/adverse effects , Gastrointestinal Microbiome/drug effects , Gene Expression , Intestine, Small/physiology , Maternal Exposure , Animals , Animals, Newborn , Colon/physiology , Female , Gastrointestinal Microbiome/genetics , Lactation , Male , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S , Sex Factors
19.
Med Hypotheses ; 64(1): 64-8, 2005.
Article in English | MEDLINE | ID: mdl-15533613

ABSTRACT

In order to reduce ammonia production by urease-positive bacteria Solga recently hypothesised (S.F. Solga, Probiotics can treat hepatic encephalopathy, Medical Hypotheses 2003; 61: 307-13), that probiotics are new therapeutics for hepatic encephalopathy (HE), and that they may replace antibiotics and lactulose. This influenced our view of the effect of antibiotics, prebiotics, e.g., lactulose, and probiotics on intestinal bacteria in the treatment of HE. Intestinal ammonia arises from aminoacids after bacterial de-amination and not from urea making urease-positive bacteria irrelevant. Antibiotics are not preferred in the treatment of HE, since ammonia-producing antibiotic-resistant bacteria may survive and replace ammonia-producing antibiotic-susceptible bacteria. Intestinal prebiotics are carbohydrate-like compounds, such as lactulose and resistant starch, that beneficially affects host's health in a different manner than normal food. In the small bowel prebiotics are not absorbed and digested, but are fermented in the colon by colonic bacteria. Fermentation of prebiotics yields lactic, acetic and butyric acids, as well as gas especially hydrogen (H2). The massive H2 volumes cause rapid intestinal hurry and thus massive amounts of colonic bacteria, not only urease-positive bacteria, but also deaminating bacteria, are removed and intestinal uptake of toxic bacterial metabolites, e.g., ammonia, reduced. As living non-pathogenic micro-organisms, probiotics beneficially affect the host's health by fermenting non-absorbed sugars, especially in the small bowel. Thus, they reduce the substrate of the other bacteria, and simultaneously they create a surplus of fermentation products which may affect the non-probiotic flora. Regarding the fermentation products (lactic acid, ethanol, acetic acid and CO2) five groups of probiotic micro-organisms are known. It is argued that probiotic, CO2-producing (facultatively) heterolactic lactobacilli, i.e., lactobacilli, that produce both lactic acid and CO2 from sugars, such as glucose, are preferred in the treatment of HE. Our ideas concur with the practice guidelines regarding HE as formulated by Blei, Cordoba and the Practice Parameters Committee of the American College of Gastroenterology, and does not alter the final conclusion of Solga as regards the beneficial use in future treatment of HE.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Hepatic Encephalopathy/diet therapy , Hepatic Encephalopathy/microbiology , Intestines/microbiology , Lactulose/therapeutic use , Probiotics/therapeutic use , Ammonia/metabolism , Clinical Trials as Topic , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Models, Biological , Treatment Outcome
20.
BMC Surg ; 4: 12, 2004 Sep 29.
Article in English | MEDLINE | ID: mdl-15456517

ABSTRACT

BACKGROUND: Infectious complications are the major cause of death in acute pancreatitis. Small bowel bacterial overgrowth and subsequent bacterial translocation are held responsible for the vast majority of these infections. Goal of this study is to determine whether selected probiotics are capable of preventing infectious complications without the disadvantages of antibiotic prophylaxis; antibiotic resistance and fungal overgrowth. METHODS/DESIGN: PROPATRIA is a double-blind, placebo-controlled randomised multicenter trial in which 200 patients will be randomly allocated to a multispecies probiotic preparation (Ecologic 641) or placebo. The study is performed in all 8 Dutch University Hospitals and 7 non-University hospitals. The study-product is administered twice daily through a nasojejunal tube for 28 days or until discharge. Patients eligible for randomisation are adult patients with a first onset of predicted severe acute pancreatitis: Imrie criteria 3 or more, CRP 150 mg/L or more, APACHE II score 8 or more. Exclusion criteria are post-ERCP pancreatitis, malignancy, infection/sepsis caused by a second disease, intra-operative diagnosis of pancreatitis and use of probiotics during the study. Administration of the study product is started within 72 hours after onset of abdominal pain. The primary endpoint is the total number of infectious complications. Secondary endpoints are mortality, necrosectomy, antibiotic resistance, hospital stay and adverse events. To demonstrate that probiotic prophylaxis reduces the proportion of patients with infectious complications from 50% to 30%, with alpha 0,05 and power 80%, a total sample size of 200 patients was calculated. CONCLUSION: The PROPATRIA study is aimed to show a reduction in infectious complications due to early enteral use of multispecies probiotics in severe acute pancreatitis.


Subject(s)
Pancreatitis/prevention & control , Probiotics/therapeutic use , Randomized Controlled Trials as Topic/methods , Acute Disease , Adult , Double-Blind Method , Follow-Up Studies , Humans , Lactobacillus plantarum , Multicenter Studies as Topic , Necrosis/prevention & control , Pancreas/pathology , Research Design , Sample Size
SELECTION OF CITATIONS
SEARCH DETAIL