Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Microbiome ; 12(1): 22, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326891

RESUMEN

BACKGROUND: The gut microbiome undergoes primary ecological succession over the course of early life before achieving ecosystem stability around 3 years of age. These maturational patterns have been well-characterized for bacteria, but limited descriptions exist for other microbiota members, such as fungi. Further, our current understanding of the prevalence of different patterns of bacterial and fungal microbiome maturation and how inter-kingdom dynamics influence early-life microbiome establishment is limited. RESULTS: We examined individual shifts in bacterial and fungal alpha diversity from 3 to 12 months of age in 100 infants from the CHILD Cohort Study. We identified divergent patterns of gut bacterial or fungal microbiome maturation in over 40% of infants, which were characterized by differences in community composition, inter-kingdom dynamics, and microbe-derived metabolites in urine, suggestive of alterations in the timing of ecosystem transitions. Known microbiome-modifying factors, such as formula feeding and delivery by C-section, were associated with atypical bacterial, but not fungal, microbiome maturation patterns. Instead, fungal microbiome maturation was influenced by prenatal exposure to artificially sweetened beverages and the bacterial microbiome, emphasizing the importance of inter-kingdom dynamics in early-life colonization patterns. CONCLUSIONS: These findings highlight the ecological and environmental factors underlying atypical patterns of microbiome maturation in infants, and the need to incorporate multi-kingdom and individual-level perspectives in microbiome research to improve our understandings of gut microbiome maturation patterns in early life and how they relate to host health. Video Abstract.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Micobioma , Humanos , Lactante , Estudios de Cohortes , Edulcorantes , Bacterias/genética
2.
Open Forum Infect Dis ; 10(5): ofad195, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37180590

RESUMEN

Interactions between the microbiome and medical therapies are distinct and bidirectional. The existing term "pharmacomicrobiomics" describes the effects of the microbiome on drug distribution, metabolism, efficacy, and toxicity. We propose that the term "pharmacoecology" be used to describe the effects that drugs and other medical interventions such as probiotics have on microbiome composition and function. We suggest that the terms are complementary but distinct and that both are potentially important when assessing drug safety and efficacy as well as drug-microbiome interactions. As a proof of principle, we describe the ways in which these concepts apply to antimicrobial and non-antimicrobial medications.

3.
FASEB J ; 37(4): e22882, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36943402

RESUMEN

Physical inactivity is one of the leading causes of chronic metabolic disease including obesity. Increasing physical activity (PA) has been shown to improve cardiometabolic and musculoskeletal health and to be associated with a distinct gut microbiota composition in trained athletes. However, the impact of PA on the gut microbiota is inconclusive for individuals performing PA in their day-to-day life. This study examined the role of PA and hand-grip strength on gut microbiome composition in middle-aged adults (40-65 years, n = 350) with normal (18.5-24.9 kg/m2 ) and overweight (25-29.9 kg/m2 ) body mass index (BMI). PA was recorded using the International Physical Activity Questionnaire, and hand-grip strength was measured using a dynamometer. Serum samples were assessed for lipidomics while DNA was extracted from fecal samples for microbiome analysis. Overweight participants showed a higher concentration of triacylglycerols, and lower concentrations of cholesteryl esters, sphingomyelin, and lyso-phosphotidylcholine lipids (p < .05) compared with those with normal BMI. Additionally, overweight participants had a lower abundance of the Oscillibacter genus (p < .05). The impact of PA duration on the gut microbiome was BMI dependent. In normal but not overweight participants, high PA duration showed greater relative abundance of commensal taxa such as Actinobacteria and Proteobacteria phyla, as well as Collinsella and Prevotella genera (p < .05). Furthermore, in males with normal BMI, a stronger grip strength was associated with a higher relative abundance of Faecalibacterium and F. prausnitzii (p < .05) compared with lower grip strength. Taken together, data suggest that BMI plays a significant role in modeling PA-induced changes in gut microbiota.


Asunto(s)
Índice de Masa Corporal , Ejercicio Físico , Microbioma Gastrointestinal , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ejercicio Físico/fisiología , Obesidad/microbiología , Sobrepeso/microbiología , Fuerza de la Mano
4.
Adv Sci (Weinh) ; 10(15): e2206615, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36995043

RESUMEN

The widespread accessibility of commercial/clinically-viable electrochemical diagnostic systems for rapid quantification of viral proteins demands translational/preclinical investigations. Here, Covid-Sense (CoVSense) antigen testing platform; an all-in-one electrochemical nano-immunosensor for sample-to-result, self-validated, and accurate quantification of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid (N)-proteins in clinical examinations is developed. The platform's sensing strips benefit from a highly-sensitive, nanostructured surface, created through the incorporation of carboxyl-functionalized graphene nanosheets, and poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS) conductive polymers, enhancing the overall conductivity of the system. The nanoengineered surface chemistry allows for compatible direct assembly of bioreceptor molecules. CoVSense offers an inexpensive (<$2 kit) and fast/digital response (<10 min), measured using a customized hand-held reader (<$25), enabling data-driven outbreak management. The sensor shows 95% clinical sensitivity and 100% specificity (Ct<25), and overall sensitivity of 91% for combined symptomatic/asymptomatic cohort with wildtype SARS-CoV-2 or B.1.1.7 variant (N = 105, nasal/throat samples). The sensor correlates the N-protein levels to viral load, detecting high Ct values of ≈35, with no sample preparation steps, while outperforming the commercial rapid antigen tests. The current translational technology fills the gap in the workflow of rapid, point-of-care, and accurate diagnosis of COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/diagnóstico , Sensibilidad y Especificidad , Nucleocápside , Antígenos
5.
Cell Rep Med ; 4(2): 100928, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36736319

RESUMEN

Unlike the bacterial microbiome, the role of early-life gut fungi in host metabolism and childhood obesity development remains poorly characterized. To address this, we investigate the relationship between the gut mycobiome of 100 infants from the Canadian Healthy Infant Longitudinal Development (CHILD) Cohort Study and body mass index Z scores (BMIz) in the first 5 years of life. An increase in fungal richness during the first year of life is linked to parental and infant BMI. The relationship between richness pattern and early-life BMIz is modified by maternal BMI, maternal diet, infant antibiotic exposure, and bacterial beta diversity. Further, the abundances of Saccharomyces, Rhodotorula, and Malassezia are differentially associated with early-life BMIz. Using structural equation modeling, we determine that the mycobiome's contribution to BMIz is likely mediated by the bacterial microbiome. This demonstrates that mycobiome maturation and infant growth trajectories are distinctly linked, advocating for inclusion of fungi in larger pediatric microbiome studies.


Asunto(s)
Microbioma Gastrointestinal , Micobioma , Obesidad Infantil , Humanos , Lactante , Niño , Índice de Masa Corporal , Estudios de Cohortes , Canadá
6.
Adv Sci (Weinh) ; 10(7): e2204171, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36461733

RESUMEN

Stress affects cognition, behavior, and physiology, leading to lasting physical and mental illness. The ability to detect and measure stress, however, is poor. Increased circulating cortisol during stress is mirrored by cortisol release from sweat glands, providing an opportunity to use it as an external biomarker for monitoring internal emotional state. Despite the attempts at using wearable sensors for monitoring sweat cortisol, there is a lack of reliable wearable sweat collection devices that preserve the concentration and integrity of sweat biomolecules corresponding to stress levels. Here, a flexible, self-powered, evaporation-free, bubble-free, surfactant-free, and scalable capillary microfluidic device, MicroSweat, is fabricated to reliably collect human sweat from different body locations. Cortisol levels are detected corresponding to severe stress ranging from 25 to 125 ng mL-1 averaged across multiple body regions and 100-1000 ng mL-1 from the axilla. A positive nonlinear correlation exists between cortisol concentration and stress levels quantified using the perceived stress scale (PSS). Moreover, owing to the sweat variation in response to environmental effects and physiological differences, the longitudinal and personalized profile of sweat cortisol is acquired, for the first time, for various body locations. The obtained sweat cortisol data is crucial for analyzing human stress in personalized and clinical healthcare sectors.


Asunto(s)
Sudor , Dispositivos Electrónicos Vestibles , Humanos , Microfluídica , Hidrocortisona , Glándulas Sudoríparas
7.
Cell Host Microbe ; 30(5): 696-711.e5, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35550672

RESUMEN

Probiotics are increasingly administered to premature infants to prevent necrotizing enterocolitis and neonatal sepsis. However, their effects on gut microbiome assembly and immunity are poorly understood. Using a randomized intervention trial in extremely premature infants, we tested the effects of a probiotic product containing four strains of Bifidobacterium species autochthonous to the infant gut and one Lacticaseibacillus strain on the compositional and functional trajectory of microbiome. Daily administration of the mixture accelerated the transition into a mature, term-like microbiome with higher stability and species interconnectivity. Besides infant age, Bifidobacterium strains and stool metabolites were the best predictors of microbiome maturation, and structural equation modeling confirmed probiotics as a major determinant for the trajectory of microbiome assembly. Bifidobacterium-driven microbiome maturation was also linked to an anti-inflammatory intestinal immune milieu. This demonstrates that Bifidobacterium strains are ecosystem engineers that lead to an acceleration of microbiome maturation and immunological consequences in extremely premature infants.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Bifidobacterium , Ecosistema , Humanos , Lactante , Recien Nacido Extremadamente Prematuro , Recién Nacido , Inflamación
8.
Pediatr Res ; 92(6): 1663-1670, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35314794

RESUMEN

BACKGROUND: Effects of probiotics on intestinal microbiota and feeding tolerance remain unclear in extremely low-birth-weight (ELBW) infants. METHODS: ELBW infants were randomly assigned to receive probiotics or no intervention. Stool samples were collected prior to, 2 and 4 weeks after initiation, and 2 weeks after probiotics cessation for infants in the probiotics group, and at matched postnatal age time points for infants in the control group. RESULTS: Of the 102 infants assessed for eligibility, sixty-two were included. Infants who received probiotics reached full enteral feeds sooner (Mean difference (MD) -1.8; 95% CI:-3.7 to -0.01 day), had a tendency toward lower incidence of hematochezia before hospital discharge (22.6% vs 3.2%; P = 0.053), and were less likely to require extensively hydrolyzed- or amino acids-based formulas to alleviate signs of cow's milk protein intolerance in the first 6 months of life (19.4% vs 51.6%; P = 0.008). Infants on probiotics were more likely to receive wide-spectrum antibiotics (64.5% vs 32.2%; P = 0.01). Multi-strain probiotics resulted in significant increase in fecal Bifidobacterium (P < 0.001) and Lactobacillus (P = 0.005), and marked reduction in fecal candida abundance (P = 0.04). CONCLUSION: Probiotics sustained intestinal Bifidobacterium and reduced time to achieve full enteral feeds in extremely preterm infants. Probiotics might improve tolerance for cow's milk protein supplements. CLINICAL TRIAL REGISTRATION: This trial has been registered at www. CLINICALTRIALS: gov (identifier NCT03422562). IMPACT: Probiotics may help extremely preterm infants achieve full enteral feeds sooner. Probiotics may improve tolerance for cow's milk protein supplements. Multi-strain probiotics can sustain intestinal Bifidobacterium and Lactobacillus until hospital discharge.


Asunto(s)
Recien Nacido Extremadamente Prematuro , Probióticos , Recién Nacido , Humanos , Femenino , Animales , Bovinos , Suplementos Dietéticos , Probióticos/uso terapéutico , Recien Nacido con Peso al Nacer Extremadamente Bajo , Proteínas de la Leche
9.
Trends Microbiol ; 30(8): 710-721, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35190251

RESUMEN

There is a growing interest to understand if and how the gut microbiome is causally linked to the pathogenesis and/or progression of diseases. While in vitro cell line models are commonly used for studying specific aspects of the host-microbe interaction, gnotobiotic murine models are considered the preferred platform for studying causality in microbiome research. Nevertheless, findings from animal studies provide limited opportunity for delineating various areas of interest to the human gut microbiome research. Gut-on-chips are biomimetics recapitulating intestinal physiology which enable investigation of bidirectional effects of the host and microbiome. We posit that they could advance causal and ecological gut microbiome research in three major areas: (i) diet-microbiome and drug-microbiome interaction; (ii) microbiome-targeted therapeutics pharmacoecology; and (iii) mechanistic studies of gut microbiome and microbiome-targeted intervention in extraintestinal pathologies.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Dieta , Microbioma Gastrointestinal/fisiología , Interacciones Microbiota-Huesped , Humanos , Ratones
10.
Adv Exp Med Biol ; 1318: 575-604, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33973200

RESUMEN

The disease 2019 (COVID-19) made a public health emergency in early 2020. Despite attempts for the development of therapeutic modalities, there is no effective treatment yet. Therefore, preventive measures in various settings could help reduce the burden of disease. In this chapter, the transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing COVID-19, non-pharmaceutical approaches at individual and population level, chemoprevention, immunoprevention, preventive measures in different healthcare settings and other professions, special considerations in high-risk groups, and the role of organizations to hamper the psychosocial effects will be discussed.


Asunto(s)
COVID-19 , Vacunas contra el Cáncer , Atención a la Salud , Humanos , Inmunoterapia , SARS-CoV-2
11.
Microbiome ; 9(1): 41, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568231

RESUMEN

BACKGROUND: Quality control including assessment of batch variabilities and confirmation of repeatability and reproducibility are integral component of high throughput omics studies including microbiome research. Batch effects can mask true biological results and/or result in irreproducible conclusions and interpretations. Low biomass samples in microbiome research are prone to reagent contamination; yet, quality control procedures for low biomass samples in large-scale microbiome studies are not well established. RESULTS: In this study, we have proposed a framework for an in-depth step-by-step approach to address this gap. The framework consists of three independent stages: (1) verification of sequencing accuracy by assessing technical repeatability and reproducibility of the results using mock communities and biological controls; (2) contaminant removal and batch variability correction by applying a two-tier strategy using statistical algorithms (e.g. decontam) followed by comparison of the data structure between batches; and (3) corroborating the repeatability and reproducibility of microbiome composition and downstream statistical analysis. Using this approach on the milk microbiota data from the CHILD Cohort generated in two batches (extracted and sequenced in 2016 and 2019), we were able to identify potential reagent contaminants that were missed with standard algorithms and substantially reduce contaminant-induced batch variability. Additionally, we confirmed the repeatability and reproducibility of our results in each batch before merging them for downstream analysis. CONCLUSION: This study provides important insight to advance quality control efforts in low biomass microbiome research. Within-study quality control that takes advantage of the data structure (i.e. differential prevalence of contaminants between batches) would enhance the overall reliability and reproducibility of research in this field. Video abstract.


Asunto(s)
Microbiota , Leche Humana/microbiología , Adulto , Animales , Preescolar , Femenino , Humanos , Lactante , Microbiota/genética , Reproducibilidad de los Resultados
12.
FEMS Microbiol Lett ; 368(3)2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33417698

RESUMEN

Previous human milk studies have confirmed the existence of a highly diverse bacterial community using culture-independent and targeted culture-dependent techniques. However, culture-enriched molecular profiling of milk microbiota has not been done. Additionally, the impact of storage conditions and milk fractionation on microbiota composition is not understood. In this feasibility study, we optimized and applied culture-enriched molecular profiling to study culturable milk microbiota in eight milk samples collected from mothers of infants admitted to a neonatal intensive care unit. Fresh samples were immediately plated or stored at -80°C for 2 weeks (short-term frozen). Long-term samples were stored at -20°C for >6 months. Samples were cultured using 10 different culture media and incubated both aerobically and anaerobically. We successfully isolated major milk bacteria, including Streptococcus, Staphylococcus and Bifidobacterium, from fresh milk samples, but were unable to culture any bacteria from the long-term frozen samples. Short-term freezing shifted the composition of viable milk bacteria from the original composition in fresh samples. Nevertheless, the inter-individual variability of milk microbiota composition was observed even after short-term storage. There was no major difference in the overall milk microbiota composition between milk fractions in this feasibility study. This is among the first studies on culture-enriched molecular profiling of the milk microbiota demonstrating the effect of storage and fractionation on milk microbiota composition.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , Biodiversidad , Microbiología de Alimentos , Leche Humana/microbiología , Bacterias/clasificación , Bacterias/genética , Técnicas de Cultivo , Humanos , ARN Ribosómico 16S/genética
13.
Nutrients ; 14(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35011027

RESUMEN

Necrotizing enterocolitis (NEC) is a significant cause of mortality and morbidity in preterm infants. The pathogenesis of NEC is not completely understood; however, intestinal immaturity and excessive immunoreactivity of intestinal mucosa to intraluminal microbes and nutrients appear to have critical roles. Dietary fats are not only the main source of energy for preterm infants, but also exert potent effects on intestinal development, intestinal microbial colonization, immune function, and inflammatory response. Preterm infants have a relatively low capacity to digest and absorb triglyceride fat. Fat may thereby accumulate in the ileum and contribute to the development of NEC by inducing oxidative stress and inflammation. Some fat components, such as long-chain polyunsaturated fatty acids (LC-PUFAs), also exert immunomodulatory roles during the early postnatal period when the immune system is rapidly developing. LC-PUFAs may have the ability to modulate the inflammatory process of NEC, particularly when the balance between n3 and n6 LC-PUFAs derivatives is maintained. Supplementation with n3 LC-PUFAs alone may have limited effect on NEC prevention. In this review, we describe how various fatty acids play different roles in the pathogenesis of NEC in preterm infants.


Asunto(s)
Grasas de la Dieta , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/prevención & control , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Recien Nacido Prematuro , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/análisis , Grasas de la Dieta/metabolismo , Digestión/fisiología , Enterocolitis Necrotizante/inmunología , Ácidos Grasos/análisis , Ácidos Grasos/química , Humanos , Fórmulas Infantiles/análisis , Recién Nacido , Inflamación , Leche Humana/química , Estrés Oxidativo
14.
BMC Microbiol ; 20(1): 290, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32948144

RESUMEN

BACKGROUND: In recent years, the microbiome field has undergone a shift from clustering-based methods of operational taxonomic unit (OTU) designation based on sequence similarity to denoising algorithms that identify exact amplicon sequence variants (ASVs), and methods to identify contaminating bacterial DNA sequences from low biomass samples have been developed. Although these methods improve accuracy when analyzing mock communities, their impact on real samples and downstream analysis of biological associations is less clear. RESULTS: Here, we re-processed our recently published milk microbiota data using Qiime1 to identify OTUs, and Qiime2 to identify ASVs, with or without contaminant removal using decontam. Qiime2 resolved the mock community more accurately, primarily because Qiime1 failed to detect Lactobacillus. Qiime2 also considerably reduced the average number of ASVs detected in human milk samples (364 ± 145 OTUs vs. 170 ± 73 ASVs, p < 0.001). Compared to the richness, the estimated diversity measures had a similar range using both methods albeit statistically different (inverse Simpson index: 14.3 ± 8.5 vs. 15.6 ± 8.7, p = 0.031) and there was strong consistency and agreement for the relative abundances of the most abundant bacterial taxa, including Staphylococcaceae and Streptococcaceae. One notable exception was Oxalobacteriaceae, which was overrepresented using Qiime1 regardless of contaminant removal. Downstream statistical analyses were not impacted by the choice of algorithm in terms of the direction, strength, and significance of associations of host factors with bacterial diversity and overall community composition. CONCLUSION: Overall, the biological observations and conclusions were robust to the choice of the sequencing processing methods and contaminant removal.


Asunto(s)
Algoritmos , ADN Bacteriano/genética , Microbiota/genética , Leche Humana/microbiología , ARN Ribosómico 16S/genética , Actinobacteria/clasificación , Actinobacteria/genética , Actinobacteria/aislamiento & purificación , Técnicas de Tipificación Bacteriana , Bacteroidetes/clasificación , Bacteroidetes/genética , Bacteroidetes/aislamiento & purificación , Contaminación de ADN , Enterobacteriaceae/clasificación , Enterobacteriaceae/genética , Enterobacteriaceae/aislamiento & purificación , Femenino , Firmicutes/clasificación , Firmicutes/genética , Firmicutes/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Filogenia , Proteobacteria/clasificación , Proteobacteria/genética , Proteobacteria/aislamiento & purificación , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/estadística & datos numéricos
15.
Front Oncol ; 10: 1029, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850307

RESUMEN

Background: Alcohol consumption is an established risk factor for colorectal cancer (CRC). Identifying cofactor(s) that modulate the effect of alcohol on colon inflammation and carcinogenesis could help risk stratification for CRC. Disruption of circadian rhythm by light/dark shift promotes alcohol-induced colonic inflammation and cancer. More recently, we found that abnormal food timing causes circadian rhythm disruption and promotes alcohol associated colon carcinogenesis. In this study, we examined the interaction of wrong-time feeding (WTF) and alcohol on CRC-related pathways, in relation to changes in microbial community structure. Methods: Polyposis mice (TS4Cre ×cAPC Δ468) underwent four conditions: alcohol or water and feeding during the light (wrong-time fed/WTF) or during the dark (right-time fed). Colonic cecum mucosal gene expression was analyzed by RNA-seq. Microbiota 16S ribosomal RNA sequencing analysis was used to examine colonic feces. Modeling was used to estimate the extent of the gene expression changes that could be related to the changes in the colonic microbial composition. Results: The circadian rhythm pathway was the most altered pathway by the WTF treatment, indicating that WTF is disruptive to the colonic circadian rhythm. Pathway analysis revealed interaction of WTF with alcohol in dysregulating pathways related to colon carcinogenesis. Similarly, the interaction of alcohol and WTF was detected at multiple parameters of the colonic microbiota including α and ß diversity, as well as the community structure. Our modeling revealed that almost a third of total gene alterations induced by our treatments could be related to alterations in the abundance of the microbial taxa. Conclusion: These data support the promoting effect of abnormal food timing alcohol-associated CRC-related pathways in the colon and suggest colon dysbiosis as a targetable mechanism.

16.
Cell Host Microbe ; 28(2): 285-297.e4, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32652062

RESUMEN

Gut microbiota play a critical role in infant health. It is now accepted that breastmilk contains live bacteria from endogenous and exogenous sources, but it remains unclear whether these bacteria transfer to the infant gut and whether this process is influenced by breastmilk feeding practices. Here, we show that certain bacteria, including Streptococcus spp. and Veillonella dispar, co-occur in mothers' milk and their infants' stool, and co-occurrence is reduced when infants receive pumped breastmilk. The relative abundances of commonly shared species are positively correlated between breastmilk and stool. Overall, gut microbiota composition is strongly associated with breastfeeding exclusivity and duration but not breastmilk feeding mode (nursing versus pumping). Moreover, breastmilk bacteria contributed to overall gut microbiota variation to a similar extent as other modifiers of the infant microbiome, such as birth mode. These results provide evidence that breastmilk may transfer bacteria to the infant gut and influence microbiota development.


Asunto(s)
Lactancia Materna/métodos , Microbioma Gastrointestinal/fisiología , Leche Humana/microbiología , Streptococcus/aislamiento & purificación , Veillonella/aislamiento & purificación , Extracción de Leche Materna/métodos , Estudios de Cohortes , Heces/microbiología , Conducta Alimentaria , Femenino , Humanos , Lactante , ARN Ribosómico 16S/genética , Streptococcus/clasificación
17.
BMC Microbiol ; 20(1): 146, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32503420

RESUMEN

BACKGROUND: Fungi constitute an important yet frequently neglected component of the human microbiota with a possible role in health and disease. Fungi and bacteria colonise the infant gastrointestinal tract in parallel, yet most infant microbiome studies have ignored fungi. Milk is a source of diverse and viable bacteria, but few studies have assessed the diversity of fungi in human milk. RESULTS: Here we profiled mycobiota in milk from 271 mothers in the CHILD birth cohort and detected fungi in 58 (21.4%). Samples containing detectable fungi were dominated by Candida, Alternaria, and Rhodotorula, and had lower concentrations of two human milk oligosaccharides (disialyllacto-N-tetraose and lacto-N-hexaose). The presence of milk fungi was associated with multiple outdoor environmental features (city, population density, and season), maternal atopy, and early-life antibiotic exposure. In addition, despite a strong positive correlation between bacterial and fungal richness, there was a co-exclusion pattern between the most abundant fungus (Candida) and most of the core bacterial genera. CONCLUSION: We profiled human milk mycobiota in a well-characterised cohort of mother-infant dyads and provide evidence of possible host-environment interactions in fungal inoculation. Further research is required to establish the role of breastfeeding in delivering fungi to the developing infant, and to assess the health impact of the milk microbiota in its entirety, including both bacterial and fungal components.


Asunto(s)
Hongos/clasificación , Leche Humana/microbiología , Oligosacáridos/análisis , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos , Lactancia Materna , Estudios de Cohortes , ADN de Hongos/genética , ADN Ribosómico/genética , Femenino , Hongos/genética , Hongos/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Microbiota , Leche Humana/química , Madres , Factores de Riesgo
18.
Gut Microbes ; 12(1): 1667722, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-31684806

RESUMEN

Human milk contains a diverse community of bacteria. The growing appreciation of commensal microbes and increasing availability of high-throughput technology has set the stage for a theory-driven approach to the study of milk microbiota, and translation of this knowledge to improve maternal and child health. We recently profiled the milk microbiota of healthy Canadian mothers and applied theory-driven causal modeling, finding that mode of breast milk feeding (nursing directly at the breast vs. pumping and feeding breast milk from a bottle) was significantly associated with milk microbiota composition. This observation could reflect an increased exposure to pumps and/or a decreased exposure to the infant mouth. Either way, it provides evidence for the retrograde mechanism of milk inoculation. Here, we discuss the implications of this research and related controversies, and raise new questions about the origins and function of milk bacteria.


Asunto(s)
Bacterias/aislamiento & purificación , Alimentación con Biberón/métodos , Lactancia Materna/métodos , Leche Humana/microbiología , Bacterias/clasificación , Extracción de Leche Materna/métodos , Canadá , Humanos , Microbiota/fisiología , Madres
19.
BMC Microbiol ; 19(1): 145, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31253096

RESUMEN

BACKGROUND: Fecal samples are currently the most commonly studied proxy for gut microbiota. The gold standard of sample handling and storage for microbiota analysis is maintaining the cold chain during sample transfer and immediate storage at - 80 °C. Gut microbiota studies in large-scale, population-based cohorts require a feasible sample collection protocol. We compared the effect of three different storage methods and mock shipment: immediate freezing at - 80 °C, in 95% ethanol stored at room temperature (RT) for 48 h, and on blood collection card stored at RT for 48 h, on the measured composition of fecal microbiota of eight healthy, female volunteers by sequencing the V4 region of the 16S rRNA gene on an Illumina MiSeq. RESULTS: Shared operational taxonomic units (OTUs) between different methods were 68 and 3% for OTUs > 0.01 and < 0.01% mean relative abundance within each group, respectively. α and ß-diversity measures were not significantly impacted by different storage methods. With the exception of Actinobacteria, fecal microbiota profiles at the phylum level were not significantly affected by the storage method. Actinobacteria was significantly higher in samples collected on card compared to immediate freezing (1.6 ± 1.1% vs. 0.4 ± 0.2%, p = 0.005) mainly driven by expansion of Actinobacteria relative abundance in fecal samples stored on card in two individuals. There was no statistically significant difference at lower taxonomic levels tested. CONCLUSION: Consistent results of the microbiota composition and structure for different storage methods were observed. Fecal collection on card could be a suitable alternative to immediate freezing for fecal microbiota analysis using 16S rRNA gene amplicon sequencing.


Asunto(s)
Biodiversidad , Heces/microbiología , Microbioma Gastrointestinal , Manejo de Especímenes/métodos , Manejo de Especímenes/normas , Bacterias/clasificación , Bacterias/genética , Estudios de Cohortes , Femenino , Congelación , Microbioma Gastrointestinal/genética , Humanos , Proyectos Piloto , ARN Ribosómico 16S/genética
20.
Front Nutr ; 6: 58, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31157227

RESUMEN

Background: Human milk contains many bioactive components that are typically studied in isolation, including bacteria. We performed an integrated analysis of human milk oligosaccharides and fatty acids to explore their associations with milk microbiota. Methods: We studied a sub-sample of 393 mothers in the CHILD birth cohort. Milk was collected at 3-4 months postpartum. Microbiota was analyzed by 16S rRNA gene V4 sequencing. Oligosaccharides and fatty acids were analyzed by rapid high-throughput high performance and gas liquid chromatography, respectively. Dimension reduction was performed with principal component analysis for oligosaccharides and fatty acids. Center log-ratio transformation was applied to all three components. Associations between components were assessed using Spearman rank correlation, network visualization, multivariable linear regression, redundancy analysis, and structural equation modeling. P-values were adjusted for multiple comparisons. Key covariates were considered, including fucosyltransferase-2 (FUT2) secretor status of mother and infant, method of feeding (direct breastfeeding or pumped breast milk), and maternal fish oil supplement use. Results: Overall, correlations were strongest between milk components of the same type. For example, FUT2-dependent HMOs were positively correlated with each other, and Staphylococcus was negatively correlated with other core taxa. Some associations were also observed between components of different types. Using redundancy analysis and structural equation modeling, the overall milk fatty acid profile was significantly associated with milk microbiota composition. In addition, some individual fatty acids [22:6n3 (docosahexaenoic acid), 22:5n3, 20:5n3, 17:0, 18:0] and oligosaccharides (fucosyl-lacto-N-hexaose, lacto-N-hexaose, lacto-N-fucopentaose I) were associated with microbiota α diversity, while others (C18:0, 3'-sialyllactose, disialyl-lacto-N-tetraose) were associated with overall microbiota composition. Only a few significant associations between individual HMOs and microbiota were observed; notably, among mothers using breast pumps, Bifidobacterium prevalence was associated with lower abundances of disialyl-lacto-N-hexaose. Additionally, among non-secretor mothers, Staphylococcus was positively correlated with sialylated HMOs. Conclusion: Using multiple approaches to integrate and analyse milk microbiota, oligosaccharides, and fatty acids, we observed several associations between different milk components and microbiota, some of which were modified by secretor status and/or breastfeeding practices. Additional research is needed to further validate and mechanistically characterize these associations and determine their relevance to infant gut and respiratory microbiota development and health.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...