Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Gut Microbes ; 15(2): 2283913, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38010080

RESUMO

The aim of this study was to obtain insight into the composition and function of the deviant gut microbiome throughout infancy in children born moderately and late preterm and their response to microbiome modulation. We characterized the longitudinal development of the gut microbiome from birth to the age of 12 months by metagenomic sequencing in 43 moderate and late preterm children participating in a randomized, controlled trial (ClinicalTrials.gov/no.NCT00167700) assessing the impact of a probiotic (Lactobacillus rhamnosus GG, ATCC 53,103, currently Lacticaseibacillus rhamnosus GG) and a prebiotic (galacto-oligosaccharide and polydextrose mixture, 1:1) intervention as compared to a placebo administered from 3 to 60 days of life. In addition, 9 full-term, vaginally delivered, breast-fed infants, who remained healthy long-term were included as references. Significant differences in taxonomy, but not in functional potential, were found when comparing the gut microbiome composition of preterm and full-term infants during the first month of life. However, the gut microbiome of preterm infants resembled that of full-term infants by 6 months age. Probiotic and prebiotic treatments were found to mitigate the shift in the microbiome of preterm infants by accelerating Bifidobacteria-dominated gut microbiome in beta diversity analysis. This study provides intriguing information regarding the establishment of the gut microbiome in children born moderately and late preterm, representing the majority of children born preterm. Specific pro- and prebiotics may reverse the proinflammatory gut microbiome composition during the vulnerable period, when the microbiome is low in resilience and susceptible to environmental exposure and simultaneously promotes immunological and metabolic maturation.


Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Probióticos , Lactente , Criança , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Microbioma Gastrointestinal/fisiologia , Prebióticos , Aleitamento Materno
2.
Gut Microbes ; 15(1): 2234656, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37469006

RESUMO

BACKGROUND: Past decades have witnessed a decrease in environmental biodiversity. We hypothesized a similar decrease in indigenous gut microbiota diversity, which may have contributed to the obesity epidemic. OBJECTIVE: To investigate the changes in the composition and function of the gut microbiota in pregnant women over a period of 20 years. STUDY DESIGN: Altogether 124 pregnant women (41 overweight and matched 83 normal weight) pregnant in 1997, 2007 or 2017 were included in the study. The gut microbiota composition was assessed from fecal samples obtained at 32 weeks of gestation by 16S rRNA gene sequencing. Fecal short chain fatty acid (SCFA) profiles were measured by gas chromatography mass spectrometry (GC-MS). RESULTS: Distinct gut microbiota profiles were detected in pregnant women from 1997, 2007 and 2017 (PERMANOVA Bray-Curtis R2 = 0.029, p = 0.001). The women pregnant in 1997 exhibited significantly higher microbiota richness and diversity as compared to those pregnant in 2007 and 2017. The total concentration of fecal SCFAs was significantly higher in the pregnant women in 1997 compared to those in 2007 and 2017. Significant differences in gut microbiota composition between normal weight and overweight women were manifest in 1997 but not in 2007 or 2017. CONCLUSIONS: The decrease in intestinal microbiota richness and diversity over two decades occurred in parallel with the decline in biodiversity in our natural surroundings. It appears that the gut microbiota of pregnant women has changed over time to a composition typical for overweight individuals.


The composition of the indigenous gut microbiota was investigated in pregnant women from three different time periods (1997, 2007 and 2017) in the same geographical and cultural area in Southwest Finland. Distinct gut microbiota profiles were evident in the women from the different time periods. The women pregnant in 1997 exhibited significantly higher microbiota richness and diversity as compared to the pregnant women from 2007 to 2017. The cause of the loss of gut microbiota richness and diversity over time remains obscure, since no major changes in the population, dietary practices or antibiotic use occurred in the area during the course of the study periods. Gut microbiota composition has been suggested to play a causal role in the development of overweight and obesity. In line with this notion, significant differences in the gut microbiota composition between normal weight and overweight were detectable in women pregnant in 1997. However, no such differences were manifest in women pregnant in 2007 or 2017 and the gut microbiota of these individuals resembled that of overweight pregnant women from 1997. The results of the study provide direct evidence for a decline in gut microbiota diversity over time in the same geographical area and the same population. It furthermore appears that the gut microbiota of pregnant women has changed over time to a composition typical for overweight individuals. The gut microbiota profiles may thus provide insight into the development and intergenerational transfer of overweight.


Assuntos
Microbioma Gastrointestinal , Sobrepeso , Humanos , Feminino , Gravidez , Gestantes , RNA Ribossômico 16S/genética , Obesidade , Fezes
3.
Pediatr Res ; 94(4): 1480-1487, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37020105

RESUMO

BACKGROUND: Preterm children with their aberrant gut microbiota and susceptibility to infections and inflammation constitute a considerable target group for probiotic therapy to generate the age-appropriate healthy microbiota. METHODS: 68 preterm neonates were randomized into five intervention groups: Beginning from the median age of 3 days, 13 children received Lactobacillus rhamnosus GG (LGG) directly orally, and 17 via the lactating mother. 14 children received LGG with Bifidobacterium lactis Bb-12 (Bb12) orally, and 10 via the lactating mother. 14 children received placebo. The children's faecal microbiota was assessed at the age of 7 days by 16S rRNA gene sequencing. RESULTS: The gut microbiota compositions of the children directly receiving the probiotic combination (LGG + Bb12) were significantly different from those of the children receiving the other intervention modes or placebo (p = 0.0012; PERMANOVA), the distinction being due to an increase in the relative abundance of Bifidobacterium animalis (P < 0.00010; ANCOM-BC), and the order Lactobacillales (P = 0.020; ANCOM-BC). CONCLUSION: The connection between aberrant primary gut microbiota and a heightened risk of infectious and non-communicable diseases invites effective microbiota modulation. We show that the direct, early, and brief probiotic intervention of LGG + Bb12 109 CFU each, is sufficient to modulate the gut microbiota of the preterm neonate. IMPACT: Preterm children have a higher risk of several health problems partly due to their aberrant gut microbiota. More research is needed to find a safe probiotic intervention to modify the gut microbiota of preterm children. The maternal administration route via breast milk might be safer for the newborn. In our study, the early and direct administration of the probiotic combination Lactobacillus rhamnosus GG with Bifidobacterium lactis Bb-12 increased the proportion of bifidobacteria in the preterm children's gut at the age of 7 days, but the maternal administration route was not as effective.


Assuntos
Bifidobacterium animalis , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Probióticos , Recém-Nascido , Criança , Feminino , Humanos , Lactação , RNA Ribossômico 16S/genética , Bifidobacterium animalis/genética , Mães
4.
Gut ; 72(5): 918-928, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36627187

RESUMO

OBJECTIVE: Gestational diabetes mellitus (GDM) is a condition in which women without diabetes are diagnosed with glucose intolerance during pregnancy, typically in the second or third trimester. Early diagnosis, along with a better understanding of its pathophysiology during the first trimester of pregnancy, may be effective in reducing incidence and associated short-term and long-term morbidities. DESIGN: We comprehensively profiled the gut microbiome, metabolome, inflammatory cytokines, nutrition and clinical records of 394 women during the first trimester of pregnancy, before GDM diagnosis. We then built a model that can predict GDM onset weeks before it is typically diagnosed. Further, we demonstrated the role of the microbiome in disease using faecal microbiota transplant (FMT) of first trimester samples from pregnant women across three unique cohorts. RESULTS: We found elevated levels of proinflammatory cytokines in women who later developed GDM, decreased faecal short-chain fatty acids and altered microbiome. We next confirmed that differences in GDM-associated microbial composition during the first trimester drove inflammation and insulin resistance more than 10 weeks prior to GDM diagnosis using FMT experiments. Following these observations, we used a machine learning approach to predict GDM based on first trimester clinical, microbial and inflammatory markers with high accuracy. CONCLUSION: GDM onset can be identified in the first trimester of pregnancy, earlier than currently accepted. Furthermore, the gut microbiome appears to play a role in inflammation-induced GDM pathogenesis, with interleukin-6 as a potential contributor to pathogenesis. Potential GDM markers, including microbiota, can serve as targets for early diagnostics and therapeutic intervention leading to prevention.


Assuntos
Diabetes Gestacional , Microbiota , Gravidez , Feminino , Humanos , Diabetes Gestacional/diagnóstico , Terceiro Trimestre da Gravidez , Inflamação , Citocinas
5.
Acta Paediatr ; 112(1): 115-121, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35989564

RESUMO

AIM: We search revision of risk determinants of the ongoing allergy epidemic. METHODS: Children numbering 433 born to mothers with allergic disease or sensitisation were selected from the three ongoing probiotic intervention trials for this case-control study. Children who developed atopic eczema or food allergy, had positive skinprick test results or had been prescribed inhaled corticosteroids by the age of 2 years were identified as cases (n = 231), while children without allergic manifestations were the healthy controls (n = 202). The data on early environmental exposures were collected from prospectively documented study records. The statistical analyses were adjusted for potential confounders. RESULTS: Determinants associated with the increased risk of atopic eczema were lower maternal prepregnancy BMI (aOR 0.15, 95% CI: 0.037-0.54) and maternal intrapartum antibiotic treatment (aOR 2.21, 95% CI 1.20-4.10), the latter also linked to obstructive respiratory symptoms (aOR 3.87, 95% CI 1.07-14.06). The risk of allergic sensitisation was associated with lower maternal prepegnancy BMI (aOR 0.18, 95% CI 0.43-0.79) and intrapartum antibiotic treatment (aOR 2.13, 95% CI 1.07-4.22). CONCLUSION: Based on our demonstrations, interventions such as personalised diets, can be optimised for specific subgroups and definite risk periods.


Assuntos
Predisposição Genética para Doença , Hipersensibilidade , Criança , Feminino , Humanos , Pré-Escolar , Estudos de Casos e Controles , Projetos de Pesquisa , Mães , Hipersensibilidade/epidemiologia
6.
Pediatr Res ; 91(7): 1804-1811, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34349229

RESUMO

BACKGROUND: Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS: Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS: Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS: The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT: The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.


Assuntos
Microbioma Gastrointestinal , Nascimento Prematuro , Feminino , Humanos , Doença Iatrogênica , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Gravidez , RNA Ribossômico 16S/genética
7.
Neonatology ; 119(1): 93-102, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34808634

RESUMO

OBJECTIVE: The aim of the study was to investigate the impact of intrapartum antibiotic treatment (IAT) on the compositional development of gut microbiota in healthy term infants. STUDY DESIGN: A case-control study of 24 infants exposed to and 24 matched infants not exposed to IAT was conducted. All subjects were born by vaginal delivery at term and breastfed. None of the infants received antibiotics during the immediate neonatal period. Fecal samples were obtained at the ages of 1 and 6 months. The composition of the intestinal microbiota was assessed by 16S rRNA gene sequencing. RESULTS: IAT was associated with reduced microbial richness but not diversity at 1 month of age. Furthermore, the relative abundances of Clostridiaceae and Erysipelotrichaceae were significantly altered in infants exposed to IAT as compared to nonexposed infants at 1 month of age. The observed deviations in gut microbiota composition between infants exposed and not exposed to IAT diminished by the age of 6 months. CONCLUSIONS: IAT is associated with short-term perturbations in the gut microbiota development in healthy term, vaginally delivered, breastfed infants. The composition of the gut microbiota is mostly restored by the age of 6 months.


Assuntos
Microbioma Gastrointestinal , Antibacterianos , Estudos de Casos e Controles , Fezes , Feminino , Humanos , Lactente , Recém-Nascido , RNA Ribossômico 16S/genética
8.
Cell Rep Med ; 2(11): 100447, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34841294

RESUMO

Preterm birth may result in adverse health outcomes. Very preterm infants typically exhibit postnatal growth restriction, metabolic disturbances, and exaggerated inflammatory responses. We investigated the differences in the meconium microbiota composition between very preterm (<32 weeks), moderately preterm (32-37 weeks), and term (>37 weeks) human neonates by 16S rRNA gene sequencing. Human meconium microbiota transplants to germ-free mice were conducted to investigate whether the meconium microbiota is causally related to the preterm infant phenotype in an experimental model. Our results indicate that very preterm birth is associated with a distinct meconium microbiota composition. Fecal microbiota transplant of very preterm infant meconium results in impaired growth, altered intestinal immune function, and metabolic parameters as compared to term infant meconium transplants in germ-free mice. This finding suggests that measures aiming to minimize the long-term adverse consequences of very preterm birth should be commenced during pregnancy or directly after birth.


Assuntos
Transplante de Microbiota Fecal , Vida Livre de Germes , Crescimento e Desenvolvimento , Recém-Nascido Prematuro/fisiologia , Inflamação/patologia , Mecônio/microbiologia , Metabolismo , Animais , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Hormônios/metabolismo , Humanos , Recém-Nascido , Inflamação/genética , Masculino , Metabolismo/genética , Camundongos , Aumento de Peso
9.
Microorganisms ; 9(3)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800493

RESUMO

The development of the infant gut microbiota is initiated during pregnancy and continued through early life and childhood, guided by the immediate environment of the child. Our aim was to characterize the shared microbiota between dogs and children as well as to determine whether introduction to dogs of a dog-specific probiotic combination modifies the transfer process. We studied 31 children from allergic families with pet dog(s) and 18 control families without a dog. Altogether 37 dogs were randomized for a 4-week period in a double-blind design to receive canine-derived probiotic product containing a mixture of L. fermentum, L. plantarum, and L. rhamnosus, or placebo. Fecal samples from children and dogs were taken before and after the treatment. Distinctive gut microbiota composition was observed in children with dogs compared to those without a dog, characterized by higher abundance of Bacteroides and short-chain fatty acid producing bacteria such as Ruminococcus and Lachnospiraceae. Probiotic intervention in dogs had an impact on the composition of the gut microbiota in both dogs and children, characterized by a reduction in Bacteroides. We provide evidence for a direct effect of home environment and household pets on children microbiota and document that modification of dog microbiota by specific probiotics is reflected in children's microbiota.

10.
Nat Commun ; 12(1): 443, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33500411

RESUMO

Exposure to antibiotics in the first days of life is thought to affect various physiological aspects of neonatal development. Here, we investigate the long-term impact of antibiotic treatment in the neonatal period and early childhood on child growth in an unselected birth cohort of 12,422 children born at full term. We find significant attenuation of weight and height gain during the first 6 years of life after neonatal antibiotic exposure in boys, but not in girls, after adjusting for potential confounders. In contrast, antibiotic use after the neonatal period but during the first 6 years of life is associated with significantly higher body mass index throughout the study period in both boys and girls. Neonatal antibiotic exposure is associated with significant differences in the gut microbiome, particularly in decreased abundance and diversity of fecal Bifidobacteria until 2 years of age. Finally, we demonstrate that fecal microbiota transplant from antibiotic-exposed children to germ-free male, but not female, mice results in significant growth impairment. Thus, we conclude that neonatal antibiotic exposure is associated with a long-term gut microbiome perturbation and may result in reduced growth in boys during the first six years of life while antibiotic use later in childhood is associated with increased body mass index.


Assuntos
Antibacterianos/efeitos adversos , Infecções Bacterianas/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Transtornos do Crescimento/induzido quimicamente , Animais , Estatura/efeitos dos fármacos , Estatura/fisiologia , Índice de Massa Corporal , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Criança , Pré-Escolar , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Seguimentos , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Transtornos do Crescimento/microbiologia , Transtornos do Crescimento/fisiopatologia , Humanos , Recém-Nascido , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Gravidez , Fatores de Risco , Fatores Sexuais
11.
Nutrients ; 12(1)2020 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-31936487

RESUMO

Preterm delivery complications are the primary cause of death among children under the age of five. Preventive strategies include the use of pasteurized donor human milk (DHM), its fortification with human milk fortifiers (protein supplements), and supplementation with probiotics. Our aim was to examine the impact of DHM and fortified DHM (FDHM) on the mucus adhesion properties of two widely used probiotics. The study covered two forms of human milk fortifier, liquid and powdered, with or without probiotics and storage at 4 °C for 24 h. To test the adhesion properties of the probiotic strains, DHM+probiotics and FDHM+probiotics were prepared and added to immobilized mucus isolated from the stool of healthy Finnish infants. The probiotic adhesion was then measured by liquid scintillation. Our results suggest that addition of liquid or powdered human milk fortifier in donor human milk had no impact on probiotic adhesion. In addition, given the increased adhesion of probiotics suspended in buffer, other matrices should be further studied. These factors need to be considered when designing future intervention strategies using probiotics in preterm infants.


Assuntos
Aderência Bacteriana/fisiologia , Leite Humano , Probióticos , Bifidobacterium/fisiologia , Fezes/química , Fezes/microbiologia , Feminino , Humanos , Lactente , Recém-Nascido , Lacticaseibacillus rhamnosus/fisiologia , Bancos de Leite Humano , Leite Humano/química , Leite Humano/microbiologia , Leite Humano/fisiologia , Muco/química
12.
Annu Rev Nutr ; 39: 267-290, 2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31433738

RESUMO

According to the developmental origins of health and disease hypothesis, our health is determined by events experienced in utero and during early infancy. Indeed, both our prenatal and postnatal nutrition conditions have an impact on the initial architecture and activity of our microbiota. Recent evidence has underlined the importance of the composition of the early gut microbiota in relation to malnutrition, whether it be undernutrition or overnutrition, that is, in terms of both stunted and overweight development. It remains unclear how early microbial contact is linked to the risk of disease, as well as whether alterations in the microbiome underlie the pathogenesis of malnutrition or are merely the end result of it, which indicates that thequestion of causality must urgently be answered. This review provides information on the complex interaction between the microbiota and nutrition during the first 1,000 days of life, taking into account the impact of both undernutrition and overnutrition on the microbiota and on infants' health outcomes in the short- and long-term.


Assuntos
Microbioma Gastrointestinal , Transtornos da Nutrição do Lactente , Feminino , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Gravidez , Fenômenos Fisiológicos da Nutrição Pré-Natal
13.
Nestle Nutr Inst Workshop Ser ; 90: 151-162, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30865983

RESUMO

Recent demonstrations link clinical conditions, phenotypes alternating from inflammatory bowel disease, obesity, and allergic diseases to neurodevelopmental disorders, to aberrant gut microbiota composition. This has led to a growing interest in host-microbe crosstalk, characterizing the healthy microbiome and modifying its deviations at an early age. The rationale arises from the recognition of the intimate interrelationship between diet, immune system, and microbiome and the origins of human diseases. Before satisfactory preventive measures can be put in practice, important questions remain to be solved. First, we need more profound understanding of the complex mechanisms underlying these heterogeneous manifestations of immune-mediated and microbiome-associated chronic conditions. Second, long-term follow-up studies are required to determine whether the changes in the microbiome underlie the pathogenesis of noncommunicable diseases or are merely end results thereof, confronting the question of causality. This uncertainty notwithstanding, the complex and bidirectional interrelationship of the diet and the gut microbiota is becoming evident. Early exposures by the enteral route induce dynamic adaptive modifications in the microbiota composition and activity, which may carry long-term clinical impacts. Microbiota changes, again, control energy acquisition and storage and may contribute to gut immunological milieu; high-energy Western diets alter the microenvironment of the gut leading to propagation of the inflammatory tone and perturbation of gut barrier function and thereby to systemic low-grade inflammation. On this basis, rigorous clinical intervention studies, providing the ultimate answers to these questions, need accurate characterization of the immediate environment of the child, in particular the early nutrition. The model of early nutrition for future studies is the healthy breastfed infant that remains healthy in the long term. Scientific interest is currently extending from the duration of breastfeeding to the composition of breast milk, which shows marked variation according to the mother's immunological and metabolic health, antibiotic use, and mode of delivery. Human milk, rich in bioactive compounds, including health-promoting microbes and their optimal growth factors, human milk oligosaccharides, continues to afford tools to study diet-microbiota interactions for research aiming at reducing the risk of noncommunicable diseases.


Assuntos
Microbioma Gastrointestinal/fisiologia , Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Aleitamento Materno , Dieta , Trato Gastrointestinal/fisiologia , Nível de Saúde , Humanos , Lactente , Recém-Nascido , Mucosa Intestinal/fisiologia , Leite Humano/química , Leite Humano/microbiologia , Leite Humano/fisiologia
14.
Appl Environ Microbiol ; 85(9)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824446

RESUMO

Recent studies report the presence of fungal species in breast milk of healthy mothers, suggesting a potential role in infant mycobiome development. In the present work, we aimed to determine whether the healthy human breast milk mycobiota is influenced by geographical location and mode of delivery, as well as to investigate its interaction with bacterial profiles in the same samples. A total of 80 mature breast milk samples from 4 different countries were analyzed by Illumina sequencing of the internal transcribed spacer 1 (ITS1) region, joining the 18S and 5.8S regions of the fungal rRNA region. Basidiomycota and Ascomycota were found to be the dominant phyla, with Malassezia and Davidiella being the most prevalent genera across countries. A core formed by Malassezia, Davidiella, Sistotrema, and Penicillium was shared in the milk samples from the different origins, although specific shifts in mycobiome composition were associated with geographic location and delivery mode. The presence of fungi in the breast milk samples was further confirmed by culture and isolate characterization, and fungal loads were estimated by quantitative PCR (qPCR) targeting the fungal ITS1 region. Cooccurrence network analysis of bacteria and fungi showed complex interactions that were influenced by geographical location, mode of delivery, maternal age, and pregestational body mass index. The presence of a breast milk mycobiome was confirmed in all samples analyzed, regardless of the geographic origin.IMPORTANCE During recent years, human breast milk has been documented as a potential source of bacteria for the newborn. Recently, we have reported the presence of fungi in breast milk from healthy mothers. It is well known that environmental and perinatal factors can affect milk bacteria; however, the impact on milk fungi is still unknown. The current report describes fungal communities (mycobiota) in breast milk samples across different geographic locations and the influence of the mode of delivery. We also provide novel insights on bacterium-fungus interactions, taking into account environmental and perinatal factors. We identified a core of four genera shared across locations, consisting of Malassezia, Davidiella, Sistotrema, and Penicillium, which have been reported to be present in the infant gut. Our data confirm the presence of fungi in breast milk across continents and support the potential role of breast milk in the initial seeding of fungal species in the infant gut.


Assuntos
Fenômenos Fisiológicos Bacterianos , Fungos/isolamento & purificação , Leite Humano/microbiologia , Micobioma , Adulto , China , Feminino , Finlândia , Geografia , Humanos , RNA Fúngico/análise , África do Sul , Espanha
15.
Front Nutr ; 6: 4, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30778389

RESUMO

The mode of delivery has been suggested to modulate the bacterial composition of breast milk but the impact of intrapartum antibiotic use on the milk microbiota is currently not known. The aim of this study was to analyze the effects of the mode of the delivery and intrapartum antibiotic administration on the microbial composition of breast milk. Breast milk samples were collected from 84 healthy mothers 1 month after the delivery. In total, 61 mothers had delivered vaginally, 23 of which had received intrapartum antibiotics, 13 women had delivered with non-elective cesarean section, 7 of which had received antibiotics, and 10 mothers had delivered with elective cesarean section without intrapartum antibiotic treatment. Both mode of delivery and intrapartum antibiotic exposure were significantly associated with changes in the milk microbial composition as assessed by analysis of similarities (ANOSIM) test (p = 0.001). The mode of delivery had a more profound effect on the milk microbiota composition as compared to intrapartum antibiotic exposure. Although the clinical significance of breast milk microbiota is currently poorly understood, this study shows that cesarean section delivery has an independent effect on breast milk microbiota composition. The dysbiosis observed in infants born by cesarean section delivery may be aggravated by the aberrant breast milk microbiota.

16.
Eur J Nutr ; 58(1): 367-377, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29299736

RESUMO

PURPOSE: Dietary supplementation with probiotics during pregnancy has been suggested to decrease the risk for obesity in women after delivery and to minimize excessive weight gain in their children. Epigenetic DNA methylation has been proposed to impact on gene activity, thereby providing a plausible molecular mechanism for a broad range of biological processes and diseases. This pilot study aimed to evaluate whether probiotic supplementation during pregnancy could modify the DNA methylation status of the promoters of obesity and weight gain-related genes in mothers and their children. METHODS: A sample of 15 pregnant women was taken from a prospective, randomized mother and infant nutrition and probiotic study. Seven women received the probiotic supplementation and eight served as controls. The women's and their children's DNA methylation status of obesity (623 genes) and weight gain-related (433) gene promoters were analyzed from blood samples at the mean of 9.8 months (range 6.1-12.7 months) postpartum. RESULTS: Probiotic supplementation led to significantly decreased levels of DNA methylation in 37 gene promoters and increased levels of DNA methylation in one gene promoter in women. In their children, 68 gene promoters were significantly affected consistently with a lower level of DNA methylation in the probiotic group. CONCLUSIONS: On the basis of our pilot study, we suggest that probiotic supplementation during pregnancy may affect the DNA methylation status of certain promoters of obesity and weight gain-related genes both in mothers and their children, thereby providing a potential mechanism for long-lasting health effects.


Assuntos
Metilação de DNA/efeitos dos fármacos , Suplementos Nutricionais , Obesidade/genética , Obesidade/metabolismo , Probióticos/farmacologia , Adulto , Feminino , Finlândia , Humanos , Lactente , Recém-Nascido , Masculino , Mães , Projetos Piloto , Gravidez , Probióticos/metabolismo , Estudos Prospectivos
17.
J Pediatr Gastroenterol Nutr ; 68(2): 272-277, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30289820

RESUMO

BACKGROUND AND OBJECTIVES: Early microbial colonization has a key impact on infant health through nutritional, immunological, and metabolic programming. The origin of child snoring is multifactorial and complex, and may thereby also generate long-term health problems. The link between child snoring and gut microbes remains unclear, although indirect evidence exists regarding this relationship. This study aimed to characterize the connection between gut microbiota and child snoring. METHODS: In a prospective, observational CHILD-SLEEP birth cohort study, gut microbiota in a subcohort of 43 of these children at 2 years of life was profiled with 16S ribosomal RNA gene amplicon sequencing. RESULTS: A higher abundance of the Proteobacteria phylum, the Enterobacteriaceae family, and Erysipelotrichaceae family, as well as a higher ratio of Firmicutes to Bacteroidetes were detected in snorers as compared to controls. Furthermore, snorers showed significantly lower microbial diversity and richness than non-snorers. CONCLUSIONS: The snoring children manifest different gut microbiota as compared with healthy children. Considering that snoring and sleep disorders can be a source of long-term consequences, including cardiovascular, metabolic, immunological, neurocognitive and behavioral consequences, our results proposes early microbiota as a new treatment target.


Assuntos
Disbiose/microbiologia , Microbioma Gastrointestinal/genética , Ronco/microbiologia , Pré-Escolar , Disbiose/complicações , Feminino , Humanos , Lactente , Masculino , Estudos Prospectivos , RNA Ribossômico 16S
18.
Pediatr Res ; 85(1): 72-78, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30237571

RESUMO

BACKGROUND AND OBJECTIVE: We investigated the associations of maternal diet and serum fatty acids during pregnancy and in early infancy on infantile neurodevelopment. METHODS: Pattern-reversal visual evoked potentials (pVEP) as depictors of central nervous system maturation were recorded from 56 children when they were 2 years old. Maternal nutrient intakes were calculated from food diaries and fish consumption from questionnaires collected during pregnancy. Serum phospholipid fatty acids were determined by gas chromatography in late pregnancy and from infants at 1 month of age. RESULTS: The children of the women who consumed fish three or more times per week during the last trimester of pregnancy had a higher pVEP component P100 amplitude for 60' (mean 23.4, SD 8.1) and 30' (mean 20.4, SD 6.7) of arcminute check sizes compared to those who consumed fish 0-2 times per week (mean 15.0, SD 4.8, p = 0.023, adjusted for birth weight and gender p = 0.058 and mean 13.4, SD 2.0, respectively, p = 0.028, adjusted p = 0.072). Maternal and child serum phospholipid fatty acids correlated with child pVEP measurements. CONCLUSION: The results of this small-scale study suggest that fish consumption during pregnancy and perinatal serum fatty acid status may associate with neurodevelopment within visual system during infancy.


Assuntos
Desenvolvimento Infantil , Fenômenos Fisiológicos da Nutrição Infantil , Potenciais Evocados Visuais , Ácidos Graxos/administração & dosagem , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Alimentos Marinhos , Vias Visuais/crescimento & desenvolvimento , Adulto , Fatores Etários , Pré-Escolar , Ácidos Graxos/sangue , Feminino , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Masculino , Valor Nutritivo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Recomendações Nutricionais , Adulto Jovem
19.
Front Pediatr ; 6: 270, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30333965

RESUMO

Human milk oligosaccharides (HMOs) are complex carbohydrates that are highly abundant in and, in their complexity, unique to human milk. Accumulating evidence indicates that exposure to HMOs in the postnatal period affects immediate as well as long-term infant health and development. However, studies reported in the 1970s showed that HMOs already appear in maternal urine and blood during pregnancy and as early as the first trimester. In this pilot study we aimed to determine whether or not HMOs also appear in amniotic fluid. We enrolled women during pregnancy and collected their urine and amniotic fluid at birth as well as their milk 4 days postpartum. We analyzed the samples by high-performance liquid chromatography (HPLC) and mass spectrometry and identified several HMOs including 2'-fucosyllactose, 3-fucosyllactose, difucosyllactose, and 6'-sialyllactose to be present in different relative abundancies in all three tissues. This is the first report that HMOs appear in amniotic fluid and that the fetus is already exposed to HMOs in utero, warranting future research to investigate the immediate and long-term implications on fetal and infant health and development.

20.
Nutrients ; 10(10)2018 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-30248972

RESUMO

The composition of human breast milk is highly variable, and it can be influenced by genetics, diet, lifestyle, and other environmental factors. This study aimed to investigate the impact of geographical location and mode of delivery on the nuclear magnetic resonance spectroscopy (NMR) metabolic profile of breast milk and its relationship with the milk microbiome. Human milk metabolic and microbiota profiles were determined using NMR and 16S rRNA gene sequencing, respectively, in 79 healthy women from Finland, Spain, South Africa, and China. Up to 68 metabolites, including amino acids, oligosaccharides, and fatty acid-associated metabolites, were identified in the milk NMR spectra. The metabolite profiles showed significant differences between geographical locations, with significant differences (p < 0.05) in the levels of galactose, lacto-N-fucopentaose III, lacto-N-fucopentaose I and 2-fucosyllactose, 3-fucosyllactose, lacto-N-difucohexaose II, lacto-N-fucopentaose III, 2-hydroxybutyrate, 3-hydroxybutyrate, proline, N-acetyl lysine, methyl-histidine, dimethylamine, kynurenine, urea, creatine and creatine phosphate, formate, lactate, acetate, phosphocholine, acetylcholine, LDL, VLDL, ethanolamine, riboflavin, hippurate, spermidine, spermine and uridine. Additionally, the effect of caesarean section on milk metabolome was dependent on the geographical region. Specific interrelations between human milk metabolites and microbiota were also identified. Proteobacteria, Actinobacteria, and Bacilli were most significantly associated with the milk metabolites, being either positively or negatively correlated depending on the metabolite. Our results reveal specific milk metabolomic profiles across geographical locations and also highlight the potential interactions between human milk's metabolites and microbes.


Assuntos
Bactérias/crescimento & desenvolvimento , Metaboloma , Microbiota , Leite Humano/metabolismo , Gravidez/etnologia , Actinobacteria/genética , Actinobacteria/crescimento & desenvolvimento , Adulto , Aminoácidos/metabolismo , Bacillus/genética , Bacillus/crescimento & desenvolvimento , Bactérias/genética , Ácidos Carboxílicos/metabolismo , China , LDL-Colesterol/metabolismo , Parto Obstétrico , Feminino , Finlândia , Humanos , Metabolômica/métodos , Microbiota/genética , Leite Humano/microbiologia , Oligossacarídeos/metabolismo , Gravidez/fisiologia , Proteobactérias/genética , Proteobactérias/crescimento & desenvolvimento , Riboflavina/metabolismo , África do Sul , Espanha , Ureia/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...