Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Appl Environ Microbiol ; : e0096424, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39007602

RESUMEN

Members of the mammalian gut microbiota metabolize diverse complex carbohydrates that are not digested by the host, which are collectively labeled "dietary fiber." While the enzymes and transporters that each strain uses to establish a nutrient niche in the gut are often exquisitely specific, the relationship between carbohydrate structure and microbial ecology is imperfectly understood. The present study takes advantage of recent advances in complex carbohydrate structure determination to test the effects of fiber monosaccharide composition on microbial fermentation. Fifty-five fibers with varied monosaccharide composition were fermented by a pooled feline fecal inoculum in a modified MiniBioReactor array system over a period of 72 hours. The content of the monosaccharides glucose and xylose was significantly associated with the reduction of pH during fermentation, which was also predictable from the concentrations of the short-chain fatty acids lactic acid, propionic acid, and the signaling molecule indole-3-acetic acid. Microbiome diversity and composition were also predictable from monosaccharide content and SCFA concentration. In particular, the concentrations of lactic acid and propionic acid correlated with final alpha diversity and were significantly associated with the relative abundance of several of the genera, including Lactobacillus and Dubosiella. Our results suggest that monosaccharide composition offers a generalizable method to compare any dietary fiber of interest and uncover links between diet, gut microbiota, and metabolite production. IMPORTANCE: The survival of a microbial species in the gut depends on the availability of the nutrients necessary for that species to survive. Carbohydrates in the form of non-host digestible fiber are of particular importance, and the set of genes possessed by each species for carbohydrate consumption can vary considerably. Here, differences in the monosaccharides that are the building blocks of fiber are considered for their impact on both the survival of different species of microbes and on the levels of microbial fermentation products produced. This work demonstrates that foods with similar monosaccharide content will have consistent effects on the survival of microbial species and on the production of microbial fermentation products.

2.
J Pediatr Gastroenterol Nutr ; 75(4): 535-542, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35881967

RESUMEN

OBJECTIVES: To compare the impact of two probiotic supplements on fecal microbiota and metabolites, as well as on gut inflammation in human milk-fed preterm infants. METHODS: In this single-center observational cohort study, we assessed the effects of Bifidobacterium longum subsp. infantis or Lactobacillus reuteri supplementation on the infant gut microbiota by 16S rRNA gene sequencing and fecal metabolome by 1 H nuclear magnetic resonance spectroscopy. Fecal calprotectin was measured as a marker of enteric inflammation. Aliquots of human or donor milk provided to each infant were also assessed to determine human milk oligosaccharide (HMO) content. RESULTS: As expected, each probiotic treatment was associated with increased proportions of the respective bacterial taxon. Fecal HMOs were significantly higher in L. reuteri fed babies despite similar HMO content in the milk consumed. Fecal metabolites associated with bifidobacteria fermentation products were significantly increased in B. infantis supplemented infants. Fecal calprotectin was lower in infants receiving B. infantis relative to L. reuteri ( P < 0.01, Wilcoxon rank-sum test) and was negatively associated with the microbial metabolite indole-3-lactate (ILA). CONCLUSIONS: This study demonstrates that supplementing an HMO-catabolizing Bifidobacterium probiotic results in increased microbial metabolism of milk oligosaccharides and reduced intestinal inflammation relative to a noncatabolizing Lactobacillus probiotic in human milk-fed preterm infants. In this context, Bifidobacterium may provide greater benefit in human milk-fed infants via activation of the microbiota-metabolite-immune axis.


Asunto(s)
Microbiota , Probióticos , Bifidobacterium , Bifidobacterium longum subspecies infantis/metabolismo , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Inflamación , Complejo de Antígeno L1 de Leucocito/metabolismo , Oligosacáridos/metabolismo , ARN Ribosómico 16S
3.
BMC Bioinformatics ; 21(1): 74, 2020 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-32093654

RESUMEN

BACKGROUND: Shotgun metagenomes are often assembled prior to annotation of genes which biases the functional capacity of a community towards its most abundant members. For an unbiased assessment of community function, short reads need to be mapped directly to a gene or protein database. The ability to detect genes in short read sequences is dependent on pre- and post-sequencing decisions. The objective of the current study was to determine how library size selection, read length and format, protein database, e-value threshold, and sequencing depth impact gene-centric analysis of human fecal microbiomes when using DIAMOND, an alignment tool that is up to 20,000 times faster than BLASTX. RESULTS: Using metagenomes simulated from a database of experimentally verified protein sequences, we find that read length, e-value threshold, and the choice of protein database dramatically impact detection of a known target, with best performance achieved with longer reads, stricter e-value thresholds, and a custom database. Using publicly available metagenomes, we evaluated library size selection, paired end read strategy, and sequencing depth. Longer read lengths were acheivable by merging paired ends when the sequencing library was size-selected to enable overlaps. When paired ends could not be merged, a congruent strategy in which both ends are independently mapped was acceptable. Sequencing depths of 5 million merged reads minimized the error of abundance estimates of specific target genes, including an antimicrobial resistance gene. CONCLUSIONS: Shotgun metagenomes of DNA extracted from human fecal samples sequenced using the Illumina platform should be size-selected to enable merging of paired end reads and should be sequenced in the PE150 format with a minimum sequencing depth of 5 million merge-able reads to enable detection of specific target genes. Expecting the merged reads to be 180-250 bp in length, the appropriate e-value threshold for DIAMOND would then need to be more strict than the default. Accurate and interpretable results for specific hypotheses will be best obtained using small databases customized for the research question.


Asunto(s)
Metagenómica/métodos , Análisis de Secuencia de ADN/métodos , Bases de Datos de Proteínas , Heces/microbiología , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metagenoma , Análisis de Secuencia de Proteína
4.
J Nutr ; 149(6): 1075-1088, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31006815

RESUMEN

BACKGROUND: Infancy is a crucial period for establishing the intestinal microbiome. This process may be influenced by vitamin A (VA) status because VA affects intestinal immunity and epithelial integrity, factors that can, in turn, modulate microbiome development. OBJECTIVES: The aim of this study was to determine if neonatal VA supplementation (VAS) affected the abundance of Bifidobacterium, a beneficial commensal, or of Proteobacteria, a phylum containing enteric pathogens, in early (6-15 wk) or late (2 y) infancy. Secondary objectives were to determine if VAS affected the abundance of other bacterial taxa, and to determine if VA status assessed by measuring plasma retinol was associated with bacterial abundance. METHODS: Three hundred and six Bangladeshi infants were randomized by sex and birthweight status (above/below median) to receive 1 VA dose (50,000 IU) or placebo within 48 h of birth. Relative abundance at the genus level and above was assessed by 16S rRNA gene sequencing. A terminal restriction fragment-length polymorphism assay was used to identify Bifidobacterium species and subspecies at 6 wk. RESULTS: Linear regression showed that Bifidobacterium abundance in early infancy was lower in boys (median, 1st/3rd quartiles; 0.67, 0.52/0.78) than girls (0.73, 0.60/0.80; P = 0.003) but that boys receiving VAS (0.69, 0.55/0.78) had higher abundance than boys receiving placebo (0.65, 0.44/0.77; P = 0.039). However this difference was not seen in girls (VAS 0.71, 0.54/0.80; placebo 0.75, 0.63/0.81; P = 0.25). VAS did not affect Proteobacteria abundance. Sex-specific associations were also seen for VA status, including positive associations of plasma retinol with Actinobacteria (the phylum containing Bifidobacterium) and Akkermansia, another commensal with possible health benefits, for girls in late infancy. CONCLUSIONS: Better VA status in infancy may influence health both in infancy and later in life by promoting the establishment of a healthy microbiota. This postulated effect of VA may differ between boys and girls. This trial was registered at clinicaltrials.gov as NCT02027610.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Vitamina A/administración & dosificación , Bangladesh , Bifidobacterium/efectos de los fármacos , Bifidobacterium/aislamiento & purificación , Preescolar , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Estudios Longitudinales , Masculino , Estado Nutricional , Proteobacteria/efectos de los fármacos , Proteobacteria/aislamiento & purificación , Vitamina A/sangre
6.
J Pediatr ; 165(1): 23-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24529620

RESUMEN

OBJECTIVES: To determine the impact of empiric ampicillin and gentamicin use in the first week of life on microbial colonization and diversity in preterm infants. STUDY DESIGN: The 16s ribosomal DNA community profiling was used to compare the microbiota of 74 infants born ≤32 weeks gestational age by degree of antibiotic use in the first week of life. The degree of antibiotic use was classified as 0 days, 1-4 days, and 5-7 days of antibiotic administration. All of the antibiotic use was empiric, defined as treatment based solely on clinical suspicion of infection without a positive culture result. RESULTS: Infants who received 5-7 days of empiric antimicrobial agents in the first week had increased relative abundance of Enterobacter (P = .016) and lower bacterial diversity in the second and third weeks of life. Infants receiving early antibiotics also experienced more cases of necrotizing enterocolitis, sepsis, or death than those not exposed to antibiotics. CONCLUSIONS: Early empiric antibiotics have sustained effects on the intestinal microbiota of preterm infants. Intestinal dysbiosis in this population has been found to be associated with elevated risk of necrotizing enterocolitis, sepsis, or death.


Asunto(s)
Antibacterianos/uso terapéutico , Enterobacter/efectos de los fármacos , Recien Nacido Prematuro , Intestinos/microbiología , Microbiota/efectos de los fármacos , Ampicilina/efectos adversos , Ampicilina/uso terapéutico , Antibacterianos/efectos adversos , Biodiversidad , Estudios de Cohortes , Dermatoglifia del ADN , ADN Ribosómico/genética , Femenino , Gentamicinas/efectos adversos , Gentamicinas/uso terapéutico , Edad Gestacional , Humanos , Recién Nacido , Estudios Longitudinales , Masculino , Ohio , ARN Ribosómico 16S/genética
7.
Cell Host Microbe ; 32(5): 637-638, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723603

RESUMEN

Gut bacteria are thought to contribute to neurodevelopmental disorders, but whether they are causal or predictive of disease remains unclear. In a prospective longitudinal study of thousands of children, Ahrens et al. generate evidence for the role of the gut microbiome in neurodevelopmental disorders while highlighting important open questions.


Asunto(s)
Microbioma Gastrointestinal , Trastornos del Neurodesarrollo , Humanos , Niño , Estudios Longitudinales , Bacterias/genética , Estudios Prospectivos
8.
Breastfeed Med ; 19(2): 98-108, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38271575

RESUMEN

Background and Objectives: Few U.S. women meet the public health recommendations to exclusively breastfeed for 6 months and continue breastfeeding for at least 1-2 years. We compared prenatally collected demographic, health, and breastfeeding support/intention variables to examine how these factors intersect to predict meeting breastfeeding recommendations. Methods: PREVAIL, a CDC-funded birth cohort in Cincinnati, OH, was approved by the IRB at CDC, Cincinnati Children's Hospital, and the hospitals where enrollment (third trimester, 2017-2018) occurred. The prenatal questionnaire captured sociodemographics, pre-pregnancy weight and height, breastfeeding environment, and breastfeeding intention, while health factors were obtained from obstetrical records. Body mass index (BMI) (kg/m2) was categorized as healthy (18.5-24.9), overweight (25-29.9), obesity 1 (30-34.9), and obesity 2+ (≥35). Mothers self-reported date of exclusive and any breastfeeding cessation through quarterly postnatal questionnaires. Random forest was used for variable selection, cross-validated in multivariable logistic models. Results: Analysis included n = 237 mothers with BMI ≥18.5. Random forest identified BMI category, prenatal intention, and insurance type as the most important predictors of meeting breastfeeding recommendations. The resulting logistic models explained >40% of the variance with an area under the curve of ≥0.89 for both recommendations. More than 73% of the risk of not meeting breastfeeding recommendations was attributable to having an elevated BMI or lacking strong breastfeeding intention. Conclusions: In PREVAIL, maternal BMI and prenatal intention explained most risks of not meeting breastfeeding exclusivity and duration recommendations. Our findings suggest efforts to improve breastfeeding exclusivity and duration should focus on strengthening prenatal breastfeeding intention and identifying effective interventions for supporting breastfeeding among mothers with higher BMI.


Asunto(s)
Cohorte de Nacimiento , Lactancia Materna , Embarazo , Niño , Femenino , Humanos , Intención , Obesidad/epidemiología , Obesidad/prevención & control , Madres
9.
J Am Nutr Assoc ; : 1-7, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38560824

RESUMEN

OBJECTIVE: Obesity as assessed by body mass index (BMI) is associated with increased risk of chronic disease. Health fatalism, defined as the belief that health outcomes are outside of one's control, is also associated with chronic disease risk. The purpose of this cross-sectional study was to understand the relationship between health fatalism and BMI in healthy adults. Secondary outcomes assessed the relationships between health fatalism and diet quality and health fatalism and physical activity. METHOD: Healthy individuals aged 18 to 65 years were recruited via ResearchMatch, electronic mailing lists, and social media. Participants completed online questionnaires on demographic characteristics, diet quality, physical activity, and degree of health fatalism. Regression models were used to assess the primary and secondary outcomes. For the primary outcome, the model of health fatalism (predictor) and BMI (outcome) was also adjusted for diet quality, physical activity, and demographic characteristics. RESULTS: Participants (n = 496) were 38.7 ± 14.3 years old and primarily female (76%) and White (81%), with a BMI of 25.1 ± 5.2 kg/m2. Most participants had a college or post-college education (74%), stated that they always had sufficient income to live comfortably (90%), and were moderately to highly active (91%). There was no relationship between health fatalism and BMI (p > 0.05) or health fatalism and physical activity (p > 0.05); however, there was a significant relationship between health fatalism and diet quality (beta coefficient: -0.046; 95% confidence interval, -0.086 to -0.0058; p = 0.025), such that a higher degree of fatalism predicted a slight decrease in diet quality. CONCLUSIONS: Although health fatalism did not predict BMI in this population, fatalistic beliefs were associated with poorer diet quality.

10.
Imeta ; 3(2): e169, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38882494

RESUMEN

The infant gut microbiome is increasingly recognized as a reservoir of antibiotic resistance genes, yet the assembly of gut resistome in infants and its influencing factors remain largely unknown. We characterized resistome in 4132 metagenomes from 963 infants in six countries and 4285 resistance genes were observed. The inherent resistome pattern of healthy infants (N = 272) could be distinguished by two stages: a multicompound resistance phase (Months 0-7) and a tetracycline-mupirocin-ß-lactam-dominant phase (Months 8-14). Microbial taxonomy explained 40.7% of the gut resistome of healthy infants, with Escherichia (25.5%) harboring the most resistance genes. In a further analysis with all available infants (N = 963), we found age was the strongest influencer on the resistome and was negatively correlated with the overall resistance during the first 3 years (p < 0.001). Using a random-forest approach, a set of 34 resistance genes could be used to predict age (R 2 = 68.0%). Leveraging microbial host inference analyses, we inferred the age-dependent assembly of infant resistome was a result of shifts in the gut microbiome, primarily driven by changes in taxa that disproportionately harbor resistance genes across taxa (e.g., Escherichia coli more frequently harbored resistance genes than other taxa). We performed metagenomic functional profiling and metagenomic assembled genome analyses whose results indicate that the development of gut resistome was driven by changes in microbial carbohydrate metabolism, with an increasing need for carbohydrate-active enzymes from Bacteroidota and a decreasing need for Pseudomonadota during infancy. Importantly, we observed increased acquired resistance genes over time, which was related to increased horizontal gene transfer in the developing infant gut microbiome. In summary, infant age was negatively correlated with antimicrobial resistance gene levels, reflecting a composition shift in the gut microbiome, likely driven by the changing need for microbial carbohydrate metabolism during early life.

11.
Microbiome ; 11(1): 194, 2023 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-37635250

RESUMEN

BACKGROUND: Bifidobacteria represent an important gut commensal in humans, particularly during initial microbiome assembly in the first year of life. Enrichment of Bifidobacterium is mediated though the utilization of human milk oligosaccharides (HMOs), as several human-adapted species have dedicated genomic loci for transport and metabolism of these glycans. This results in the release of fermentation products into the gut lumen which may offer physiological benefits to the host. Synbiotic pairing of probiotic species with a cognate prebiotic delivers a competitive advantage, as the prebiotic provides a nutrient niche. METHODS: To determine the fitness advantage and metabolic characteristics of an HMO-catabolizing Bifidobacterium strain in the presence or absence of 2'-fucosyllactose (2'-FL), conventionally colonized mice were gavaged with either Bifidobacterium pseudocatenulatum MP80 (B.p. MP80) (as the probiotic) or saline during the first 3 days of the experiment and received water or water containing 2'-FL (as the prebiotic) throughout the study. RESULTS: 16S rRNA gene sequencing revealed that mice provided only B.p. MP80 were observed to have a similar microbiota composition as control mice throughout the experiment with a consistently low proportion of Bifidobacteriaceae present. Using 1H NMR spectroscopy, similar metabolic profiles of gut luminal contents and serum were observed between the control and B.p. MP80 group. Conversely, synbiotic supplemented mice exhibited dramatic shifts in their community structure across time with an overall increased, yet variable, proportion of Bifidobacteriaceae following oral inoculation. Parsing the synbiotic group into high and moderate bifidobacterial persistence based on the median proportion of Bifidobacteriaceae, significant differences in gut microbial diversity and metabolite profiles were observed. Notably, metabolites associated with the fermentation of 2'-FL by bifidobacteria were significantly greater in mice with a high proportion of Bifidobacteriaceae in the gut suggesting metabolite production scales with population density. Moreover, 1,2-propanediol, a fucose fermentation product, was only observed in the liver and brain of mice harboring high proportions of Bifidobacteriaceae. CONCLUSIONS: This study reinforces that the colonization of the gut with a commensal microorganism does not guarantee a specific functional output. Video Abstract.


Asunto(s)
Actinobacteria , Bifidobacterium pseudocatenulatum , Simbióticos , Humanos , Animales , Ratones , ARN Ribosómico 16S/genética , Leche Humana , Oligosacáridos , Bifidobacterium , Prebióticos
12.
Nutrients ; 15(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37299550

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of death caused by gastrointestinal disease in preterm infants. Major risk factors include prematurity, formula feeding, and gut microbial colonization. Microbes have been linked to NEC, yet there is no evidence of causal species, and select probiotics have been shown to reduce NEC incidence in infants. In this study, we evaluated the effect of the probiotic Bifidobacterium longum subsp. infantis (BL. infantis), alone and in combination with a human milk oligosaccharide (HMO)-sialylactose (3'SL)-on the microbiome, and the incidence of NEC in preterm piglets fed an infant formula diet. We studied 50 preterm piglets randomized between 5 treatments: (1) Preterm infant formula, (2) Donor human milk (DHM), (3) Infant formula + 3'SL, (4) Infant formula + BL. infantis, and (5) Infant formula and BL. infantis + 3'SL. NEC incidence and severity were assessed through the evaluation of tissue from all the segments of the GI tract. The gut microbiota composition was assessed both daily and terminally through 16S and whole-genome sequencing (WGS) of rectal stool samples and intestinal contents. Dietary BL. infantis and 3'SL supplementation had no effect, yet DHM significantly reduced the incidence of NEC. The abundance of BL. infantis in the gut contents negatively correlated with disease severity. Clostridium sensu stricto 1 and Clostridium perfringens were significantly more abundant in NEC and positively correlated with disease severity. Our results suggest that pre- and probiotics are not sufficient for protection from NEC in an exclusively formula-based diet. The results highlight the differences in microbial species positively associated with both diet and NEC incidence.


Asunto(s)
Enterocolitis Necrotizante , Microbioma Gastrointestinal , Probióticos , Animales , Humanos , Bifidobacterium longum subspecies infantis , Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/etiología , Incidencia , Leche Humana , Porcinos
13.
J Perinatol ; 42(11): 1446-1452, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35840710

RESUMEN

OBJECTIVE: Feeding tubes harbor microbial contaminants; studies to date have not explored differences between orogastric (OG) and nasogastric (NG) tube biofilms. We sought to extend a previous analysis by comparing bacterial colonization by location (OG v NG) and by evaluating clinical factors that may affect tube bacterial populations. STUDY DESIGN: The pharyngeal segments of 41 infant feeding tubes (14 OG and 27 NG) from 41 infants were analyzed by next generation 16 S rRNA sequencing on the MiSeq platform. RESULTS: At the phylum level, Proteobacteria had the highest relative abundance of both OG and NG tubes. At the genus/species level, nine taxa differed significantly between OG and NG tubes. Alpha and beta diversity analyses showed significant differences between OG and NG tubes with relatively little contribution from clinical factors. CONCLUSION: The route of feeding tube insertion (oral vs nasal) had a greater impact on bacterial colonization than the assessed clinical factors.


Asunto(s)
Nutrición Enteral , Unidades de Cuidado Intensivo Neonatal , Lactante , Recién Nacido , Humanos , Intubación Gastrointestinal , Bacterias/genética , Nariz
14.
mBio ; 13(3): e0010122, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35536006

RESUMEN

Antimicrobial resistance (AMR) represents a significant source of morbidity and mortality worldwide, with expectations that AMR-associated consequences will continue to worsen throughout the coming decades. Since resistance to antibiotics is encoded in the microbiome, interventions aimed at altering the taxonomic composition of the gut might allow us to prophylactically engineer microbiomes that harbor fewer antibiotic resistant genes (ARGs). Diet is one method of intervention, and yet little is known about the association between diet and antimicrobial resistance. To address this knowledge gap, we examined diet using the food frequency questionnaire (FFQ; habitual diet) and 24-h dietary recalls (Automated Self-Administered 24-h [ASA24®] tool) coupled with an analysis of the microbiome using shotgun metagenome sequencing in 290 healthy adult participants of the United States Department of Agriculture (USDA) Nutritional Phenotyping Study. We found that aminoglycosides were the most abundant and prevalent mechanism of AMR in these healthy adults and that aminoglycoside-O-phosphotransferases (aph3-dprime) correlated negatively with total calories and soluble fiber intake. Individuals in the lowest quartile of ARGs (low-ARG) consumed significantly more fiber in their diets than medium- and high-ARG individuals, which was concomitant with increased abundances of obligate anaerobes, especially from the family Clostridiaceae, in their gut microbiota. Finally, we applied machine learning to examine 387 dietary, physiological, and lifestyle features for associations with antimicrobial resistance, finding that increased phylogenetic diversity of diet was associated with low-ARG individuals. These data suggest diet may be a potential method for reducing the burden of AMR. IMPORTANCE Antimicrobial resistance (AMR) represents a considerable burden to health care systems, with the public health community largely in consensus that AMR will be a major cause of death worldwide in the coming decades. Humans carry antibiotic resistance in the microbes that live in and on us, collectively known as the human microbiome. Diet is a powerful method for shaping the human gut microbiome and may be a tractable method for lessening antibiotic resistance, and yet little is known about the relationship between diet and AMR. We examined this relationship in healthy individuals who contained various abundances of antibiotic resistance genes and found that individuals who consumed diverse diets that were high in fiber and low in animal protein had fewer antibiotic resistance genes. Dietary interventions may be useful for lessening the burden of antimicrobial resistance and might ultimately motivate dietary guidelines which will consider how nutrition can reduce the impact of infectious disease.


Asunto(s)
Antibacterianos , Microbioma Gastrointestinal , Animales , Antibacterianos/farmacología , Dieta , Fibras de la Dieta , Farmacorresistencia Bacteriana/genética , Humanos , Filogenia
15.
Nutrients ; 14(7)2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35406036

RESUMEN

Bifidobacterium species are beneficial and dominant members of the breastfed infant gut microbiome; however, their health benefits are partially species-dependent. Here, we characterize the species and subspecies of Bifidobacterium in breastfed infants around the world to consider the potential impact of a historic dietary shift on the disappearance of B. longum subsp. infantis in some populations. Across populations, three distinct patterns of Bifidobacterium colonization emerged: (1) The dominance of Bifidobacterium longum subspecies infantis, (2) prevalent Bifidobacterium of multiple species, and (3) the frequent absence of any Bifidobacterium. These patterns appear related to a country's history of breastfeeding, with infants in countries with historically high rates of long-duration breastfeeding more likely to be colonized by B. longum subspecies infantis compared with infants in countries with histories of shorter-duration breastfeeding. In addition, the timing of infant colonization with B. longum subsp. infantis is consistent with horizontal transmission of this subspecies, rather than the vertical transmission previously reported for other Bifidobacterium species. These findings highlight the need to consider historical and cultural influences on the prevalence of gut commensals and the need to understand epidemiological transmission patterns of Bifidobacterium and other major commensals.


Asunto(s)
Bifidobacterium longum , Microbioma Gastrointestinal , Bifidobacterium , Lactancia Materna , Estudios Transversales , Femenino , Humanos , Lactante
16.
PLoS One ; 16(10): e0258308, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34648525

RESUMEN

The ongoing COVID-19 pandemic is causing significant morbidity and mortality across the US. In this ecological study, we identified county-level variables associated with the COVID-19 case-fatality rate (CFR) using publicly available datasets and a negative binomial generalized linear model. Variables associated with decreased CFR included a greater number of hospitals per 10,000 people, banning religious gatherings, a higher percentage of people living in mobile homes, and a higher percentage of uninsured people. Variables associated with increased CFR included a higher percentage of the population over age 65, a higher percentage of Black or African Americans, a higher asthma prevalence, and a greater number of hospitals in a county. By identifying factors that are associated with COVID-19 CFR in US counties, we hope to help officials target public health interventions and healthcare resources to locations that are at increased risk of COVID-19 fatalities.


Asunto(s)
COVID-19/mortalidad , Factores de Edad , Estudios Transversales , Femenino , Humanos , Masculino , Modelos Teóricos , Pandemias , Pronóstico , Factores de Riesgo , Estados Unidos/epidemiología
17.
Gut Microbes ; 13(1): 1986666, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34705611

RESUMEN

Understanding how exogenous microbes stably colonize the animal gut is essential to reveal mechanisms of action and tailor effective probiotic treatments. Bifidobacterium species are naturally enriched in the gastrointestinal tract of breast-fed infants. Human milk oligosaccharides (HMOs) are associated with this enrichment. However, direct mechanistic proof of the importance of HMOs in this colonization is lacking given milk contains additional factors that impact the gut microbiota. This study examined mice supplemented with the HMO 2'fucosyllactose (2'FL) together with a 2'FL-consuming strain, Bifidobacterium pseudocatenulatum MP80. 2'FL supplementation creates a niche for high levels of B.p. MP80 persistence, similar to Bifidobacterium levels seen in breast-fed infants. This synergism impacted gut microbiota composition, activated anti-inflammatory pathways and protected against chemically-induced colitis. These results demonstrate that bacterial-milk glycan interactions alone drive enrichment of beneficial Bifidobacterium and provide a model for tunable colonization thus facilitating insight into mechanisms of health promotion by bifidobacteriain neonates.


Asunto(s)
Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Colitis/prevención & control , Leche Humana/metabolismo , Oligosacáridos/metabolismo , Animales , Lactancia Materna , Colitis/metabolismo , Colitis/microbiología , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
18.
Nutrients ; 13(3)2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804415

RESUMEN

Breastfeeding is the gold standard for feeding infants because of its long-term benefits to health and development, but most infants in the United States are not exclusively breastfed in the first six months. We enrolled 24 infants who were either exclusively breastfed or supplemented with formula by the age of one month. We collected diet information, stool samples for evaluation of microbiotas by 16S rRNA sequencing, and blood samples for assessment of immune development by flow cytometry from birth to 6 months of age. We further typed the Bifidobacterium strains in stool samples whose 16S rRNA sequencing showed the presence of Bifidobacteriaceae. Supplementation with formula during breastfeeding transiently changed the composition of the gut microbiome, but the impact dissipated by six months of age. For example, Bifidobacterium longum, a bacterial species highly correlated with human milk consumption, was found to be significantly different only at 1 month of age but not at later time points. No immunologic differences were found to be associated with supplementation, including the development of T-cell subsets, B cells, or monocytes. These data suggest that early formula supplementation, given in addition to breast milk, has minimal lasting impact on the gut microbiome or immunity.


Asunto(s)
Suplementos Dietéticos/microbiología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/microbiología , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Lactancia Materna/métodos , Encuestas sobre Dietas , Heces/microbiología , Femenino , Humanos , Sistema Inmunológico/microbiología , Lactante , Recién Nacido , Masculino , ARN Ribosómico 16S/aislamiento & purificación , Estados Unidos
19.
medRxiv ; 2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33655256

RESUMEN

The ongoing COVID-19 pandemic is causing significant morbidity and mortality across the US. In this ecological study, we identified county-level variables associated with the COVID-19 case-fatality rate (CFR) using publicly available datasets and a negative binomial generalized linear model. Variables associated with decreased CFR included a greater number of hospitals per 10,000 people, banning religious gatherings, a higher percentage of people living in mobile homes, and a higher percentage of uninsured people. Variables associated with increased CFR included a higher percentage of the population over age 65, a higher percentage of Black or African Americans, a higher asthma prevalence, and a greater number of hospitals in a county. By identifying factors that are associated with COVID-19 CFR in US counties, we hope to help officials target public health interventions and healthcare resources to locations that are at increased risk of COVID-19 fatalities.

20.
Cell Host Microbe ; 26(2): 147-148, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31415742

RESUMEN

Human milk feeding is associated with lower rates of necrotizing enterocolitis (NEC), but an understanding of mechanism is lacking. In recent work, Gopalakrishna et al. report that human infants who develop NEC first experience an increase in Enterobacteriaceae in the portion of the microbiota not bound to IgA.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Inmunoglobulina A/uso terapéutico , Leche Humana/inmunología , Animales , Disbiosis/prevención & control , Enterobacteriaceae/patogenicidad , Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/microbiología , Humanos , Lactante , Recién Nacido , Madres , Tacto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA