Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nutrients ; 15(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36678283

RESUMEN

Background: Microbiological quality is one of the key safety standards in human milk bank (HMB) operations. We describe the profiles of bacteria in donor human milk (DHM) before and after the pasteurization of samples collected from breastfeeding women in the hospital and from the community in the first HMB in Vietnam. Methods: Data were collected between February 2017 and January 2022 from an online HMB monitoring system. First, DHM samples were cultured, and the number of colony-forming units (CFU) were counted before (n = 708) and after pasteurization (n = 1146). The gram-staining method combined with the Vitek 2 Compact system were used to identify types of organisms at the Da Nang Hospital for Women and Children's Laboratory. Passing criteria for DHM included pre-pasteurization samples had a total colony count <105 CFU/mL and post-pasteurization was <10 CFU/mL. Results: During five years of operation, Da Nang HMB had 491 donors (48.7% were hospital and the rest community donors) who donated an average amount of 14.2 L over 45 days. Of this DHM volume, 84.9% of donor samples passed the pre- and post-pasteurization microbiological tests. DHM from community donors had a higher pass rate (87.8%) compared to that from hospital donors (79.5%). Before pasteurization, 15.4% of DHM samples had a bacteria count <103 CFU/mL, 63.0% had 103-<105 CFU/mL, and 21.6% had ≥105 CFU/mL. Most of the unpasteurized DHM samples (93.0%) had microorganism growth: with one organism (16.4%), two (33.9%), three or more (43.6%). After pasteurization, 17.9% samples had a bacteria count of 1−9 CFU/mL and 7.2% had ≥10 CFU/mL. DHM samples from community donors had a lower bacterial count and number of organisms than those from hospital donors both before and after pasteurization. The highest microorganisms from unpasteurized DHM samples were Staphylococcus epidermidis (74.2%), Acinetobacter sp. (52.1%), gram-positive bacillus (51.7%), Staphylococcus coagulase-negative (15.8%), and Staphylococcus aureus (10.5%). Common microorganisms from pasteurized DHM were gram-positive bacillus (21.0%), Staphylococcus epidermidis (3.9%), and Acinetobacter sp. (0.9%). Samples from the hospital tended to have a higher contamination with those microorganisms than those from community donors. Conclusions: The majority of DHM samples in Da Nang passed microbiological testing criteria. DHM from community donors had higher pass rates than hospital donors. Corrective actions are needed to improve HMB operations and hospital microbiological quality standards, as well as general improvements in water and sanitation.


Asunto(s)
Bancos de Leche Humana , Leche Humana , Niño , Embarazo , Femenino , Humanos , Animales , Leche Humana/microbiología , Vietnam , Leche/microbiología , Pasteurización/métodos , Calostro
2.
Mol Nutr Food Res ; 67(2): e2200444, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36480309

RESUMEN

SCOPE: Supplementing Limosilactobacillus reuteri Fn041, a breast milk-derived probiotic from agricultural and pastoral areas, to maternal mice during late pregnancy and lactation prevents atopic dermatitis (AD) in offspring. This study aims to elucidate the molecular mechanism of Fn041-mediated immune regulation. METHODS AND RESULTS: Fn041 is administered prenatal and postnatal to maternal mice, and to offspring after weaning. The ears are administered with calcipotriol to induce AD. Fn041 treatment significantly alleviates ear inflammation, and reduces mast cell infiltration. Fn041 treatment upregulates and downregulates intestinal ZO-1 and Claudin-2 mRNA expression, respectively. Transcriptome analysis of Peyer's patches reveals that pathways related to DNA damage repair are activated in AD mice, which is inhibited by Fn041 treatment. Fn041 activates pathways related to retinol absorption and metabolism. Untargeted metabolomic analysis reveals that Fn041 treatment increases plasma retinol and kynurenine. Fn041 treatment does not significantly alter the overall cecal microbiota profile, only increases the relative abundances of Ligilactobacillus apodemi, Ligilactobacillus murinus, Akkermansia muciniphila, and Bacteroides thetaiotaomicron. CONCLUSIONS: Fn041 induces anti-AD immune responses directly by promoting the absorption and metabolism of retinol in Peyer's patches, and plays an indirect role by strengthening the mucosal barrier and increasing the abundance of specific anti-AD bacteria in the cecum.


Asunto(s)
Dermatitis Atópica , Limosilactobacillus reuteri , Leche Humana , Ganglios Linfáticos Agregados , Vitamina A , Animales , Femenino , Ratones , Embarazo , Dermatitis Atópica/prevención & control , Dermatitis Atópica/metabolismo , Leche Humana/microbiología , Vitamina A/metabolismo , Humanos
3.
Probiotics Antimicrob Proteins ; 15(3): 491-501, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-34671923

RESUMEN

Breast milk was long considered a sterile environment, but now it is known to harbor many bacteria that will shape the newborn microbiota. The benefits of breastfeeding to newborn health are, on some level, related to the presence of beneficial bacteria in human milk. Therefore, this study aims to investigate and isolate potential probiotics present in human milk that might be associated with improved health in infants, being potential candidates to be used in simulated human milk formula. Milk samples of 24 healthy mothers were collected at three time points: 30 min (colostrum), 5-9 days (transitional milk), and 25-30 days (mature milk) postpartum. Samples were evaluated by culturing, and the isolated bacteria were identified by MALDI-TOF MS and 16S DNA sequencing. In vitro screening for probiotics properties was performed, and the potential probiotics were mono-associated with germ-free mice to evaluate their ability to colonize the gastrointestinal tract. The microorganisms were submitted to the spray-drying process to check their viability for a potential simulated milk formula production. Seventy-seven bacteria were isolated from breast milk pertaining to four bacterial genera (Staphylococcus, Streptococcus, Leuconostoc, and Lacticaseibacillus). Four potential probiotics were selected: Lacticaseibacillus rhamnosus (n = 2) and Leuconostoc mesenteroides (n = 2). Isolates were able to colonize the gastrointestinal tract of germ-free mice and remained viable after the spray-drying process. In conclusion, breast milk harbors a unique microbiota with beneficial microorganisms that will impact the newborn gut colonization, being an essential source of probiotic candidates to be used in a formula of simulated maternal milk.


Asunto(s)
Leche Humana , Probióticos , Lactante , Femenino , Embarazo , Humanos , Animales , Ratones , Leche Humana/microbiología , Bifidobacterium/genética , Bacterias/genética , Calostro/microbiología
4.
Breastfeed Med ; 17(11): 940-946, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36378822

RESUMEN

Background: The microbial community in human milk is associated with many maternal and neonatal factors. This study aimed to investigate the effect of antibiotic exposure on the microbial community structure of colostrum. Methods: Twenty women with antibiotic treatment immediately after delivery and 10 age-matched control women were enrolled at the Guangdong Women and Children Hospital. Colostrum samples were collected within postpartum 30 hours. The V4 variable region of the bacterial 16S rRNA gene was sequenced to characterize the microbial profile using Illumina MiSeq platform. Results: Phyla Proteobacteria and Firmicutes were the predominant bacteria in colostrum samples. The core and abundant genera in colostrum included Streptococcus, Staphylococcus, and Pseudomonas. Compared with the control group, principal coordinate analysis based on the Bray-Curtis distance showed a significant difference in milk microbial community in women with antibiotic exposure, accompanied by a significantly lower alpha diversity and a different microbial ecological network. Furthermore, the relative abundances of genera Actinomyces, Anaerobacter, and Clostridium_sensu_stricto significantly decreased after antibiotic treatment. Conclusions: This study provided evidence of alterations in the colostrum microbial community with antibiotic exposure, improving our understanding of the effects of antibiotic treatment on the milk microbiome.


Asunto(s)
Calostro , Microbiota , Embarazo , Recién Nacido , Niño , Femenino , Humanos , ARN Ribosómico 16S/genética , Antibacterianos/uso terapéutico , Lactancia Materna , Leche Humana/microbiología
5.
Nutrients ; 13(12)2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34959752

RESUMEN

Intestinal colonization of the neonate is highly dependent on the term of pregnancy, the mode of delivery, the type of feeding [breast feeding or formula feeding]. Postnatal immune maturation is dependent on the intestinal microbiome implementation and composition and type of feeding is a key issue in the human gut development, the diversity of microbiome, and the intestinal function. It is well established that exclusive breastfeeding for 6 months or more has several benefits with respect to formula feeding. The composition of the new generation of infant formulas aims in mimicking HM by reproducing its beneficial effects on intestinal microbiome and on the gut associated immune system (GAIS). Several approaches have been developed currently for designing new infant formulas by the addition of bioactive ingredients such as human milk oligosaccharides (HMOs), probiotics, prebiotics [fructo-oligosaccharides (FOSs) and galacto-oligosaccharides (GOSs)], or by obtaining the so-called post-biotics also known as milk fermentation products. The aim of this article is to guide the practitioner in the understanding of these different types of Microbiota Influencing Formulas by listing and summarizing the main concepts and characteristics of these different models of enriched IFs with bioactive ingredients.


Asunto(s)
Ingestión de Alimentos/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Fórmulas Infantiles/química , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Femenino , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/microbiología , Recién Nacido , Intestinos/crecimiento & desarrollo , Intestinos/inmunología , Masculino , Leche Humana/química , Leche Humana/microbiología , Oligosacáridos/administración & dosificación , Prebióticos/administración & dosificación
6.
Sci Rep ; 11(1): 24373, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34934118

RESUMEN

Gestational Diabetes Mellitus (GDM) and obesity affect the functioning of multiple maternal systems and influence colonization of the newborn gastrointestinal through the breastmilk microbiota (BMM). It is currently unclear how GDM and obesity affect the human BMM composition. Here, we applied 16S-rRNA high-throughput sequencing to human colostrum milk to characterize BMM taxonomic changes in a cohort of 43 individuals classified in six subgroups according to mothers patho-physiological conditions (healthy control (n = 18), GDM (n = 13), or obesity (n = 12)) and newborn gender. Using various diversity indicators, including Shannon/Faith phylogenetic index and UniFrac/robust Aitchison distances, we evidenced that BMM composition was influenced by the infant gender in the obesity subgroup. In addition, the GDM group presented higher microbial diversity compared to the control group. Staphylococcus, Corynebacterium 1, Anaerococcus and Prevotella were overrepresented in colostrum from women with either obesity or GDM, compared to control samples. Finally, Rhodobacteraceae was distinct for GDM and 5 families (Bdellovibrionaceae, Halomonadaceae, Shewanellaceae, Saccharimonadales and Vibrionaceae) were distinct for obesity subgroups with an absolute effect size greater than 1 and a q-value ≤ 0.05. This study represents the first effort to describe the impact of maternal GDM and obesity on BMM.


Asunto(s)
Bacterias/genética , Calostro/microbiología , Diabetes Gestacional/microbiología , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Leche Humana/microbiología , Obesidad/microbiología , Adulto , Bacterias/clasificación , Bacterias/aislamiento & purificación , Índice de Masa Corporal , Femenino , Humanos , Recién Nacido , Masculino , Filogenia , Embarazo
7.
Nutrients ; 13(4)2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33808265

RESUMEN

Recent evidence indicates that maternal dietary intake, including dietary supplements, during pregnancy and lactation may alter the infant gut or breastmilk microbiota, with implications for health outcomes in both the mother and infant. To review the effects of maternal nutritional supplementation during pregnancy and lactation on the infant gut or breastmilk microbiota a systematic literature search was conducted. A total of 967 studies published until February 2020 were found, 31 were eligible and 29 randomized control trials were included in the qualitative synthesis. There were 23 studies that investigated the effects of probiotic supplementation, with the remaining studies investigating vitamin D, prebiotics or lipid-based nutrient supplements (LNS). The effects of maternal nutritional supplementation on the infant gut microbiota or breastmilk microbiota were examined in 21 and 12 studies, respectively. Maternal probiotic supplementation during pregnancy and lactation generally resulted in the probiotic colonization of the infant gut microbiota, and although most studies also reported alterations in the infant gut bacterial loads, there was limited evidence of effects on bacterial diversity. The data available show that maternal probiotic supplementation during pregnancy or lactation results in probiotic colonization of the breastmilk microbiota. There were no observed effects between probiotic supplementation and breastmilk bacterial counts of healthy women, however, administration of Lactobacillus probiotic to nursing women affected by mastitis was associated with significant reductions in breastmilk Staphylococcal loads. Maternal LNS supplementation during pregnancy and lactation increased bacterial diversity in the infant gut, whilst vitamin D and prebiotic supplementation did not alter either infant gut bacterial diversity or counts. Heterogeneity in study design precludes any firm conclusions on the effects of maternal nutritional supplementation during pregnancy and lactation on the infant gut or breastmilk microbiota, warranting further research.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Lactancia/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Leche Humana/microbiología , Femenino , Humanos , Lactante , Embarazo
8.
Int J Mol Sci ; 21(19)2020 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-33023062

RESUMEN

Human breast milk (HBM) is an irreplaceable source of nutrition for early infant growth and development. Breast-fed children are known to have a low prevalence and reduced risk of various diseases, such as necrotizing enterocolitis, gastroenteritis, acute lymphocytic leukemia, and acute myeloid leukemia. In recent years, HBM has been found to contain a microbiome, extracellular vesicles or exosomes, and microRNAs, as well as nutritional components and non-nutritional proteins, including immunoregulatory proteins, hormones, and growth factors. Especially, the milk-derived exosomes exert various physiological and therapeutic function in cell proliferation, inflammation, immunomodulation, and cancer, which are mainly attributed to their cargo molecules such as proteins and microRNAs. The exosomal miRNAs are protected from enzymatic digestion and acidic conditions, and play a critical role in immune regulation and cancer. In addition, the milk-derived exosomes are developed as drug carriers for delivering small molecules and siRNA to tumor sites. In this review, we examined the various components of HBM and their therapeutic potential, in particular of exosomes and microRNAs, towards cancer.


Asunto(s)
Exosomas/genética , Microbiota/genética , Leche Humana/química , Neoplasias/terapia , Exosomas/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Femenino , Humanos , MicroARNs/genética , Leche Humana/metabolismo , Leche Humana/microbiología , Neoplasias/patología , Estado Nutricional
9.
Curr Opin Infect Dis ; 33(6): 548-555, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33009143

RESUMEN

PURPOSE OF REVIEW: There has been an exponential increase in research into infant microbiome evolution, and it appears that pharyngeal microbiota are associated with clinical phenotypes (e.g. infection and asthma). Although broad consensus views are emerging, significant challenges and uncertainties remain. RECENT FINDINGS: Infant pharyngeal microbiome research is limited by low biomass, high temporal diversity and lack of agreed standards for sampling, DNA sequencing and taxonomic reporting. Analysis of amplicon sequence variants and improved cost and availability of whole-genome sequencing are promising options for improving taxonomic resolution of such studies. Infant respiratory microbiomes arise, at least in part, from maternal flora (e.g. the respiratory tract and breastmilk), and are associated with environmental and clinical factors (e.g. mode of feeding and delivery, siblings, daycare attendance, birth season and antibiotic usage). Interventional research to modify the infant pharyngeal microbiota has recently been reported, using dietary supplements. SUMMARY: Further work is needed to improve characterization of the infant pharyngeal microbiomes, including routes of bacterial acquisition, role of environmental factors and associations with disease phenotypes. Methodological standards are desirable to facilitate more reproducible, comparable research. Improved understanding may enable manipulation of infant pharyngeal microbiota to improve clinical outcomes.


Asunto(s)
Microbiota , Faringe/microbiología , Enfermedades Respiratorias/microbiología , Antibacterianos/uso terapéutico , Asma/microbiología , Bacterias/clasificación , Bacterias/genética , Ambiente , Humanos , Lactante , Recién Nacido , Infecciones/microbiología , Salud Materna , Leche Humana/microbiología , Sistema Respiratorio/microbiología , Secuenciación Completa del Genoma
10.
Nutrients ; 12(10)2020 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-33036184

RESUMEN

Preterm infants are a vulnerable population at risk of intestinal dysbiosis. The newborn microbiome is dominated by Bifidobacterium species, though abnormal microbial colonization can occur by exogenous factors such as mode of delivery, formula feeding, and exposure to antibiotics. Therefore, preterm infants are predisposed to sepsis and necrotizing enterocolitis (NEC), a fatal gastrointestinal disorder, due to an impaired intestinal barrier, immature immunity, and a dysbiotic gut microbiome. Properties of human milk serve as protection in the prevention of NEC. Human milk oligosaccharides (HMOs) and the microbiome of breast milk are immunomodulatory components that provide intestinal homeostasis through regulation of the microbiome and protection of the intestinal barrier. Enteral probiotic supplements have been trialed to evaluate their impact on establishing intestinal homeostasis. Here, we review the protective role of HMOs, probiotics, and synbiotic combinations in protecting a vulnerable population from the pathogenic features associated with necrotizing enterocolitis.


Asunto(s)
Disbiosis/microbiología , Disbiosis/prevención & control , Ingestión de Alimentos/fisiología , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Recien Nacido Prematuro , Intestinos/microbiología , Leche Humana , Oligosacáridos/administración & dosificación , Probióticos/administración & dosificación , Femenino , Homeostasis , Humanos , Recién Nacido , Masculino , Leche Humana/química , Leche Humana/microbiología , Riesgo
11.
Cell Rep Med ; 1(5): 100077, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32904427

RESUMEN

Supplementation with members of the early-life microbiota as "probiotics" is increasingly used in attempts to beneficially manipulate the preterm infant gut microbiota. We performed a large observational longitudinal study comprising two preterm groups: 101 infants orally supplemented with Bifidobacterium and Lactobacillus (Bif/Lacto) and 133 infants non-supplemented (control) matched by age, sex, and delivery method. 16S rRNA gene profiling on fecal samples (n = 592) showed a predominance of Bifidobacterium and a lower abundance of pathobionts in the Bif/Lacto group. Metabolomic analysis showed higher fecal acetate and lactate and a lower fecal pH in the Bif/Lacto group compared to the control group. Fecal acetate positively correlated with relative abundance of Bifidobacterium, consistent with the ability of the supplemented Bifidobacterium strain to metabolize human milk oligosaccharides into acetate. This study demonstrates that microbiota supplementation is associated with a Bifidobacterium-dominated preterm microbiota and gastrointestinal environment more closely resembling that of full-term infants.


Asunto(s)
Bifidobacterium/fisiología , Microbioma Gastrointestinal/fisiología , Recien Nacido Prematuro/metabolismo , Recien Nacido Prematuro/fisiología , Lactobacillus/fisiología , Metaboloma/fisiología , Bifidobacterium/genética , Lactancia Materna/métodos , Suplementos Dietéticos/microbiología , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Lactante , Recién Nacido , Lactobacillus/genética , Estudios Longitudinales , Leche Humana/microbiología , Probióticos/administración & dosificación , ARN Ribosómico 16S/genética
12.
Microbiology (Reading) ; 166(10): 966-980, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32886600

RESUMEN

Maternal milk is an important source of essential nutrients for the optimal growth of infants. Breastfeeding provides a continuous supply of beneficial bacteria to colonize the infant gastrointestinal tract (GIT) and offers health benefits for disease prevention and immunity. The purpose of this study was to isolate novel probiotic strains from the breast milk of native Pakistani mothers and to evaluate their probiotic potential. We isolated 21 strains of bacteria from the colostrum and mature milk of 20 healthy mothers, who had vaginal deliveries and were not taking antibiotics. After phenotypic and genotypic characterization, these isolates were tested for survival in the GIT using in vitro acid and bile tests. Nine strains showing good acid tolerance were assessed for their growth rate, bile resistance and ability to hydrolyze bile salts. Out of the four Lactobacillus isolates adjudged to be most promising as probiotics, three were Lactobacillus fermentum strains and one was a strain of Lactobacillus oris. This study demonstrates that human milk is a viable source of commensal bacteria beneficial to both adults and babies.


Asunto(s)
Lactobacillus/fisiología , Leche Humana/microbiología , Probióticos , Ácidos/metabolismo , Adulto , Ácidos y Sales Biliares/metabolismo , Calostro/microbiología , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Lactante , Lactobacillus/clasificación , Lactobacillus/aislamiento & purificación , Filogenia , Embarazo , Probióticos/clasificación , Probióticos/aislamiento & purificación , Adulto Joven
13.
Nutrients ; 12(9)2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32825705

RESUMEN

Inter-subject variability in human milk microbiome is well known; however, its origins and possible relationship to the mother's diet are still debated. We investigated associations between maternal nutrition, milk fatty acids composition and microbiomes in mother-infant dyads. Breast milk and infant fecal samples were collected across three time points (one week, one month and three months postpartum) from 22 mother-infant pairs. Food frequency questionnaires for the months of pregnancy and three months postpartum were collected. Milk fatty acids were analyzed by GC-MS and the microbiome in breast milk and infant feces was determined by 16S rRNA sequencing. Statistical interactions were computed using Spearman's method and corrected for multiple comparisons. We found significant negative correlation between Streptococcus relative abundance in maternal milk and intake of unsaturated fatty acids and folic acid at one month postpartum. At three months postpartum, vitamin B-12 consumption was significantly associated with a single operational taxonomic unit belonging to Streptococcus. Comparison between milk microbiome and lipid composition showed, one-month postpartum, significant negative correlation between Streptococcus relative abundance and the abundance of oleic acid. Additional correlations were detected between Staphylococcus hominis and two medium-chain saturated fatty acids. Our results reinforce the hypothesis that maternal nutrition may affect milk microbiome.


Asunto(s)
Suplementos Dietéticos , Ingestión de Alimentos/fisiología , Ácidos Grasos/análisis , Conducta Alimentaria/fisiología , Microbioma Gastrointestinal , Lactancia/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Leche Humana/metabolismo , Leche Humana/microbiología , Ácidos Grasos Insaturados/administración & dosificación , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Relaciones Madre-Hijo , Embarazo , Streptococcus , Encuestas y Cuestionarios , Vitamina B 12/administración & dosificación
14.
Gut Microbes ; 11(5): 1438-1449, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32543266

RESUMEN

Bacteria in human milk could directly seed the infant intestinal microbiota, while information about how milk microbiota develops during lactation and how geographic location, gestational hypertensive status, and maternal age influence this process is limited. Here, we collected human milk samples from mothers of term infants at the first day, 2 weeks, and 6 weeks postpartum from 117 longitudinally followed-up mothers (age: 28.7 ± 3.6 y) recruited from three cities in China. We found that milk microbial diversity and richness were the highest in colostrum but gradually decreased over lactation. Microbial composition changed across lactation and exhibited more discrete compositional patterns in 2-week and 6-week milk samples compared with colostrum samples. At phylum level, the abundance of Proteobacteria increased during lactation, while Firmicutes showed the opposite trend. At genus level, Staphylococcus, Streptococcus, Acinetobacter, Pseudomonas, and Lactobacillus were predominant in colostrum samples and showed distinct variations across lactation. Maternal geographic location was significantly associated with the milk microbiota development and the abundance of predominant genus. In addition, milk from mothers with gestational prehypertension had a different and less diverse microbial community at genus level in early lactation times, and contained less Lactobacillus in the 2-week milk samples than those from normotensive mothers. Findings of our study outlined the human milk microbial diversity and community development over lactation, and underscored the importance of maternal geographic locations and gestational hypertensive status on milk microbiota, which might have important implications in the establishment of the infant intestinal microbiota via breastfeeding.


Asunto(s)
Bacterias/clasificación , Hipertensión Inducida en el Embarazo/microbiología , Lactancia , Microbiota , Leche Humana/microbiología , Adulto , Bacterias/crecimiento & desarrollo , Calostro/microbiología , Dieta , Femenino , Firmicutes/crecimiento & desarrollo , Geografía , Humanos , Lactante , Recién Nacido , Embarazo , Proteobacteria/crecimiento & desarrollo
15.
Food Funct ; 11(6): 5607-5620, 2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32525185

RESUMEN

Constipation is a prevalent and burdensome gastrointestinal (GI) disorder that seriously affects the quality of human life. This study evaluated the effects of the P. pentosaceus B49 (from human colostrum) on loperamide (Lop)-induced constipation in mice. Mice were given P. pentosaceus B49 (5 × 109 CFU or 5 × 1010 CFU) by gavage daily for 14 days. The result shows that P. pentosaceus B49 treatment relieved constipation in mice by shortening the defecation time, increasing the GI transit rate and stool production. Compared with the constipation control group, the P. pentosaceus B49-treated groups showed decreased serum levels of inhibitory neurotransmitters (vasoactive intestinal peptide and nitric oxide), increased serum levels of excitatory neurotransmitters (acetylcholinesterase, motilin, and gastrin), and elevated cecal concentration of short chain fatty acids (SCFAs). Analysis of cecal microbiota reveals that P. pentosaceus B49 was colonized in the intestine of constipated mice, and altered the cecal microbiota by increasing beneficial SCFAs-producing bacteria (i.e., Lactobacillus, Ruminococcaceae_UCG-014, and Bacteroidales_S24-7) and decreasing potential pathogenic bacteria (i.e., Staphylococcus and Helicobacter). Moreover, transcriptome analysis of the colon tissue shows that P. pentosaceus B49 partly normalized the expression of genes related to GI peristalsis (i.e., Ache, Chrm2, Slc18a3, Grp, and Vip), water and electrolyte absorption and transport (i.e., Aqp4, Aqp8, and Atp12a), while down-regulating the expression of pro-inflammatory and pro-oncogenic genes (i.e., Lbp, Lgals2, Bcl2, Bcl2l15, Gsdmc2, and Olfm4) in constipated mice. Our findings indicate that P. pentosaceus B49 effectively relieves constipation in mice and is a promising candidate for treating constipation.


Asunto(s)
Colon/metabolismo , Calostro/microbiología , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico , Estreñimiento/microbiología , Pediococcus pentosaceus/metabolismo , Acetilcolinesterasa , Animales , Bacterias , Ácidos Grasos Volátiles/metabolismo , Heces , Gastrinas , Tránsito Gastrointestinal/efectos de los fármacos , Tránsito Gastrointestinal/fisiología , Hormonas/sangre , Humanos , Intestinos , Loperamida/efectos adversos , Masculino , Ratones , Ratones Endogámicos BALB C , Leche Humana/microbiología , Motilina , Neurotransmisores/sangre , Estrés Oxidativo , Pediococcus pentosaceus/genética , Pediococcus pentosaceus/aislamiento & purificación , Peristaltismo/genética , Probióticos/uso terapéutico , ARN Ribosómico 16S/genética , Transcriptoma
16.
PLoS One ; 15(5): e0233284, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32469943

RESUMEN

Human milk provides complete nutrition for infants and at the same time promotes the growth of specific bacteria in the infant gastrointestinal tract. Breastfeeding can often be discontinued due to mastitis which is an inflammation of the breast tissue. We isolated 18 Staphylococcus aureus strains from milk donated by healthy (n = 6), subclinical (n = 6), and mastitic (n = 6) mothers, two strains of which were VISA (Vancomycin Intermediate S. aureus). All tested strains (n = 12) were able to form biofilms. We then examined the impact of nisin A and vancomycin alone and in combination on biofilm formation and eradication of selected strains (n = 8). We observed strain-specific responses, with the combinatorial treatment at 1/4X MIC (for both singularly) significantly inhibiting biofilm formation for seven out of eight strains when compared with nisin A or vancomycin alone. None of the selected treatments were able to eradicate pre-formed biofilms. Finally, we selected two strains, namely a VISA (APC3814H) and a strong biofilm former (APC3912CM) and used confocal microscopy to evaluate the effects of the antimicrobial agents at 1X MIC on biofilm inhibition and eradication. All treatments inhibited biofilm formation of APC3814H but were ineffective in eradicating a pre-formed biofilm. Single treatments at 1X MIC against APC3912CM cells did not prevent biofilm formation whereas combination treatment caused increased death of APC3912CM cells. Finally, the combination treatment reduced the thickness of the pre-formed APC3912CM biofilm as compared with the single treatments.


Asunto(s)
Biopelículas/efectos de los fármacos , Mastitis/tratamiento farmacológico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Leche Humana/efectos de los fármacos , Nisina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Vancomicina/uso terapéutico , Antibacterianos/uso terapéutico , Biopelículas/crecimiento & desarrollo , Quimioterapia Combinada , Femenino , Humanos , Mastitis/microbiología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Leche Humana/microbiología , Infecciones Estafilocócicas/microbiología
17.
Nutrients ; 12(4)2020 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-32295092

RESUMEN

Maternal bacteria are shared with infants via breastfeeding. Prebiotics modulate the gut microbiota, promoting health benefits. We investigated whether the maternal diet supplementation with a prebiotic (fructooligosaccharides, FOS) could influence the milk microbiota. Twenty-eight lactating women received 4.5 g of fructooligosaccharides + 2 g of maltodextrin (FOS group) and twenty-five received 2 g of maltodextrin (placebo group) for 20 days. Breast-milk samples were taken before and after the intervention. The DNA from samples was used for 16S rRNA sequencing. No statistical differences between the groups were found for the bacterial genera after the intervention. However, the distances of the trajectories covered by paired samples from the beginning to the end of the supplementation were higher for the FOS group (p = 0.0007) indicating greater changes in milk microbiota compared to the control group. Linear regression models suggested that the maternal age influenced the response for FOS supplementation (p = 0.02). Interestingly, the pattern of changes to genus abundance upon supplementation was not shared between mothers. We demonstrated that manipulating the human milk microbiota through prebiotics is possible, and the maternal age can affect this response. .


Asunto(s)
Lactancia Materna , Suplementos Dietéticos , Microbioma Gastrointestinal , Edad Materna , Leche Humana/microbiología , Oligosacáridos/administración & dosificación , Polisacáridos/administración & dosificación , Prebióticos/administración & dosificación , Adolescente , Adulto , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Método Simple Ciego , Adulto Joven
18.
Nutr Clin Pract ; 35(3): 386-405, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32329544

RESUMEN

The early-life microbiome is gaining appreciation as a major influencer in human development and long-term health. Multiple factors are known to influence the initial colonization, development, and function of the neonatal gut microbiome. In addition, alterations in early-life gut microbial composition is associated with several chronic health conditions such as obesity, asthma, and allergies. In this review, we focus on both maternal and infant factors known to influence early-life gut colonization. Also reviewed is the important role of infant feeding, including evidence-based strategies for maternal and infant supplementation with the goal to protect and/or restore the infant gut microbiome.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Antibacterianos/efectos adversos , Lactancia Materna , Parto Obstétrico/métodos , Suplementos Dietéticos , Femenino , Feto/microbiología , Humanos , Lactante , Fórmulas Infantiles , Salud del Lactante , Recién Nacido , Fenómenos Fisiologicos Nutricionales Maternos , Leche Humana/química , Leche Humana/microbiología , Obesidad/complicaciones , Obesidad/microbiología , Prebióticos/administración & dosificación , Prebióticos/análisis , Embarazo , Complicaciones del Embarazo/microbiología , Probióticos/administración & dosificación
19.
Nutrients ; 12(2)2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32069822

RESUMEN

Cardiorespiratory function is not only the foremost determinant of life after premature birth, but also a major factor of long-term outcomes. However, the path from placental disconnection to nutritional autonomy is enduring and challenging for the preterm infant and, at each step, will have profound influences on respiratory physiology and disease. Fluid and energy intake, specific nutrients such as amino-acids, lipids and vitamins, and their ways of administration -parenteral or enteral-have direct implications on lung tissue composition and cellular functions, thus affect lung development and homeostasis and contributing to acute and chronic respiratory disorders. In addition, metabolomic signatures have recently emerged as biomarkers of bronchopulmonary dysplasia and other neonatal diseases, suggesting a profound implication of specific metabolites such as amino-acids, acylcarnitine and fatty acids in lung injury and repair, inflammation and immune modulation. Recent advances have highlighted the profound influence of the microbiome on many short- and long-term outcomes in the preterm infant. Lung and intestinal microbiomes are deeply intricated, and nutrition plays a prominent role in their establishment and regulation. There is an emerging evidence that human milk prevents bronchopulmonary dysplasia in premature infants, potentially through microbiome composition and/or inflammation modulation. Restoring antibiotic therapy-mediated microbiome disruption is another potentially beneficial action of human milk, which can be in part emulated by pre- and probiotics and supplements. This review will explore the many facets of the gut-lung axis and its pathophysiology in acute and chronic respiratory disorders of the prematurely born infant, and explore established and innovative nutritional approaches for prevention and treatment.


Asunto(s)
Enfermedades del Prematuro/metabolismo , Enfermedades Pulmonares/fisiopatología , Microbiota/fisiología , Nutrientes/metabolismo , Nacimiento Prematuro/fisiopatología , Femenino , Microbioma Gastrointestinal/fisiología , Humanos , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Recien Nacido Prematuro/crecimiento & desarrollo , Enfermedades del Prematuro/etiología , Enfermedades del Prematuro/microbiología , Pulmón/crecimiento & desarrollo , Pulmón/microbiología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/microbiología , Masculino , Leche Humana/microbiología , Placenta/microbiología , Embarazo , Nacimiento Prematuro/microbiología
20.
Sci Rep ; 9(1): 18653, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819085

RESUMEN

Archaeal sequences have been detected in human colostrum and milk, but no studies have determined whether living archaea are present in either of these fluids. Methanogenic archaea are neglected since they are not detected by usual molecular and culture methods. By using improved DNA detection protocols and microbial culture techniques associated with antioxidants previously developed in our center, we investigated the presence of methanogenic archaea using culture and specific Methanobrevibacter smithii and Methanobrevibacter oralis real-time PCR in human colostrum and milk. M. smithii was isolated from 3 colostrum and 5 milk (day 10) samples. M. oralis was isolated from 1 milk sample. For 2 strains, the genome was sequenced, and the rhizome was similar to that of strains previously isolated from the human mouth and gut. M. smithii was detected in the colostrum or milk of 5/13 (38%) and 37/127 (29%) mothers by culture and qPCR, respectively. The different distribution of maternal body mass index according to the detection of M. smithii suggested an association with maternal metabolic phenotype. M. oralis was not detected by molecular methods. Our results suggest that breastfeeding may contribute to the vertical transmission of these microorganisms and may be essential to seed the infant's microbiota with these neglected critical commensals from the first hour of life.


Asunto(s)
Lactancia Materna/efectos adversos , Calostro/microbiología , Methanobrevibacter/aislamiento & purificación , Leche Humana/microbiología , Animales , Índice de Masa Corporal , Crecimiento Quimioautotrófico/genética , ADN de Archaea/genética , ADN de Archaea/aislamiento & purificación , Euryarchaeota/genética , Euryarchaeota/patogenicidad , Heces/microbiología , Femenino , Humanos , Lactante , Methanobrevibacter/genética , Methanobrevibacter/patogenicidad , Microbiota/genética , Madres , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA