Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Sci Adv ; 10(15): eadj0400, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38598636

RESUMO

Despite the recognized gut-brain axis link, natural variations in microbial profiles between patients hinder definition of normal abundance ranges, confounding the impact of dysbiosis on infant neurodevelopment. We infer a digital twin of the infant microbiome, forecasting ecosystem trajectories from a few initial observations. Using 16S ribosomal RNA profiles from 88 preterm infants (398 fecal samples and 32,942 abundance estimates for 91 microbial classes), the model (Q-net) predicts abundance dynamics with R2 = 0.69. Contrasting the fit to Q-nets of typical versus suboptimal development, we can reliably estimate individual deficit risk (Mδ) and identify infants achieving poor future head circumference growth with ≈76% area under the receiver operator characteristic curve, 95% ± 1.8% positive predictive value at 98% specificity at 30 weeks postmenstrual age. We find that early transplantation might mitigate risk for ≈45.2% of the cohort, with potentially negative effects from incorrect supplementation. Q-nets are generative artificial intelligence models for ecosystem dynamics, with broad potential applications.


Assuntos
Microbioma Gastrointestinal , Microbiota , Lactente , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Inteligência Artificial , Microbioma Gastrointestinal/genética , Fezes
2.
Gut Microbes ; 16(1): 2298697, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38303501

RESUMO

The early life gut microbiome affects the developing brain, and therefore may serve as a target to support neurodevelopment of children living in stressful and under-resourced environments, such as Black youth living on the South Side of Chicago, for whom we observe racial disparities in health. Microbiome compositions/functions key to multiple neurodevelopmental facets have not been studied in Black children, a vulnerable population due to racial disparities in health; thus, a subsample of Black infants living in urban, low-income neighborhoods whose mothers participated in a prenatal nutrition study were recruited for testing associations between composition and function of the gut microbiome (16S rRNA gene sequencing, shotgun metagenomics, and targeted metabolomics of fecal samples) and neurodevelopment (developmental testing, maternal report of temperament, and observed stress regulation). Two microbiome community types, defined by high Lachnospiraceae or Enterobacteriaceae abundance, were discovered in this cohort from 16S rRNA gene sequencing analysis; the Enterobacteriaceae-dominant community type was significantly negatively associated with cognition and language scores, specifically in male children. Vitamin B12 biosynthesis emerged as a key microbiome function from shotgun metagenomics sequencing analysis, showing positive associations with all measured developmental skills (i.e., cognition, language, motor, surgency, effortful control, and observed stress regulation). Blautia spp. also were identified as substantial contributors of important microbiome functions, including vitamin B12 biosynthesis and related vitamin B12-dependent microbiome functions, anti-inflammatory microbial surface antigens, competitive mechanisms against pathobionts, and production of antioxidants. The results are promising with respect to the potential for exploring therapeutic candidates, such as vitamin B12 nutritional or Blautia spp. probiotic supplementation, to support the neurodevelopment of infants at risk for experiencing racial disparities in health.


Assuntos
Microbioma Gastrointestinal , Vitamina B 12 , Lactente , Criança , Gravidez , Feminino , Adolescente , Humanos , Masculino , RNA Ribossômico 16S/genética , Microbioma Gastrointestinal/genética , Encéfalo , Vitaminas
3.
Microorganisms ; 11(5)2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37317106

RESUMO

Necrotizing enterocolitis (NEC) is the leading basis for gastrointestinal morbidity and poses a significant risk for neurodevelopmental impairment (NDI) in preterm infants. Aberrant bacterial colonization preceding NEC contributes to the pathogenesis of NEC, and we have demonstrated that immature microbiota in preterm infants negatively impacts neurodevelopment and neurological outcomes. In this study, we tested the hypothesis that microbial communities before the onset of NEC drive NDI. Using our humanized gnotobiotic model in which human infant microbial samples were gavaged to pregnant germ-free C57BL/6J dams, we compared the effects of the microbiota from preterm infants who went on to develop NEC (MNEC) to the microbiota from healthy term infants (MTERM) on brain development and neurological outcomes in offspring mice. Immunohistochemical studies demonstrated that MNEC mice had significantly decreased occludin and ZO-1 expression compared to MTERM mice and increased ileal inflammation marked by the increased nuclear phospho-p65 of NFκB expression, revealing that microbial communities from patients who developed NEC had a negative effect on ileal barrier development and homeostasis. In open field and elevated plus maze tests, MNEC mice had worse mobility and were more anxious than MTERM mice. In cued fear conditioning tests, MNEC mice had worse contextual memory than MTERM mice. MRI revealed that MNEC mice had decreased myelination in major white and grey matter structures and lower fractional anisotropy values in white matter areas, demonstrating delayed brain maturation and organization. MNEC also altered the metabolic profiles, especially carnitine, phosphocholine, and bile acid analogs in the brain. Our data demonstrated numerous significant differences in gut maturity, brain metabolic profiles, brain maturation and organization, and behaviors between MTERM and MNEC mice. Our study suggests that the microbiome before the onset of NEC has negative impacts on brain development and neurological outcomes and can be a prospective target to improve long-term developmental outcomes.

4.
Biomolecules ; 13(5)2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37238716

RESUMO

Vitamin D signaling via the Vitamin D Receptor (VDR) has been shown to protect against intestinal inflammation. Previous studies have also reported the mutual interactions of intestinal VDR and the microbiome, indicating a potential role of probiotics in modulating VDR expression. In preterm infants, although probiotics have been shown to reduce the incidence of necrotizing enterocolitis (NEC), they are not currently recommended by the FDA due to potential risks in this population. No previous studies have delved into the effect of maternally administered probiotics on intestinal VDR expression in early life. Using an infancy mouse model, we found that young mice exposed to maternally administered probiotics (SPF/LB) maintained higher colonic VDR expression than our unexposed mice (SPF) in the face of a systemic inflammatory stimulus. These findings indicate a potential role for microbiome-modulating therapies in preventing diseases such as NEC through the enhancement of VDR signaling.


Assuntos
Enterocolite Necrosante , Probióticos , Recém-Nascido , Humanos , Animais , Camundongos , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Recém-Nascido Prematuro , Intestinos , Enterocolite Necrosante/prevenção & controle , Enterocolite Necrosante/metabolismo , Probióticos/farmacologia , Probióticos/uso terapêutico
5.
Microorganisms ; 11(4)2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37110458

RESUMO

Necrotizing Enterocolitis (NEC) is characterized by an inflammation of intestinal tissue that primarily affects premature infants. It is the most common and devastating gastrointestinal morbidity of prematurity, but beyond intestinal morbidity, this condition has also been associated with an increased risk of neurodevelopmental delays that persist beyond infancy. Prematurity, enteral feeding, bacterial colonization, and prolonged exposure to antibiotics are all risk factors that predispose preterm infants to NEC. Interestingly, these factors are all also associated with the gut microbiome. However, whether or not there is a connection between the microbiome and the risk of neurodevelopmental delays in infants after NEC is still an emerging area of research. Furthermore, how microbes in the gut could impact a distant organ such as the brain is also poorly understood. In this review, we discuss the current understanding of NEC and the role of the gut microbiome-brain axis in neurodevelopmental outcomes after NEC. Understanding the potential role of the microbiome in neurodevelopmental outcomes is important as the microbiome is modifiable and thus offers the hope of improved therapeutic options. We highlight the progress and limitations in this field. Insights into the gut microbiome-brain axis may offer potential therapeutic approaches to improve the long-term outcomes of premature infants.

6.
Gut Microbes ; 15(1): 2178800, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36799469

RESUMO

Maternal immune activation (MIA) derived from late gestational infection such as seen in chorioamnionitis poses a significantly increased risk for neurodevelopmental deficits in the offspring. Manipulating early microbiota through maternal probiotic supplementation has been shown to be an effective means to improve outcomes; however, the mechanisms remain unclear. In this study, we demonstrated that MIA modeled by exposing pregnant dams to lipopolysaccharide (LPS) induced an underdevelopment of the blood vessels, an increase in permeability and astrogliosis of the blood-brain barrier (BBB) at prewean age. The BBB developmental and functional deficits early in life impaired spatial learning later in life. Maternal Limosilactobacillus reuteri (L. reuteri) supplementation starting at birth rescued the BBB underdevelopment and dysfunction-associated cognitive function. Maternal L. reuteri-mediated alterations in ß-diversity of the microbial community and metabolic responses in the offspring provide mechanisms and potential targets for promoting BBB integrity and long-term neurodevelopmental outcomes.


Assuntos
Microbioma Gastrointestinal , Limosilactobacillus reuteri , Efeitos Tardios da Exposição Pré-Natal , Feminino , Recém-Nascido , Gravidez , Humanos , Barreira Hematoencefálica/metabolismo , Lipopolissacarídeos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/metabolismo
7.
Semin Perinatol ; 47(1): 151694, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36572620

RESUMO

Necrotizing enterocolitis (NEC) poses a significant risk for neurodevelopmental impairment in extremely preterm infants. The gut microbiota shapes the development of the gut, immune system, and the brain; and dysbiosis drive neonatal morbidities including NEC. In this chapter, we delineate a gut-brain axis linking gut microbiota to the adverse neurological outcomes in NEC patients. We propose that in NEC, immaturity of the microbiome along with aberrant gut microbiota-driven immaturity of the gut barrier and immune system can lead to effects including systemic inflammation and circulating microbial mediators. This nexus of gut microbiota-driven systemic effects further interacts with a likewise underdeveloped blood-brain barrier to regulate neuroinflammation and neurodevelopment. Targeting deviant gut-brain axis signaling presents an opportunity to improve the neurodevelopmental outcomes of NEC patients.


Assuntos
Enterocolite Necrosante , Microbioma Gastrointestinal , Doenças do Recém-Nascido , Microbiota , Recém-Nascido , Lactente , Humanos , Eixo Encéfalo-Intestino , Lactente Extremamente Prematuro
8.
Nat Microbiol ; 7(10): 1506-1507, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36163499
9.
Sci Rep ; 12(1): 3310, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35228616

RESUMO

Interventions to mitigate long-term neurodevelopmental deficits such as memory and learning impairment in preterm infants are warranted. Manipulation of the gut microbiome affects host behaviors. In this study we determined whether early maturation of the infant microbiome is associated with neurodevelopment outcomes. Germ free mice colonized at birth with human preterm infant microbiomes from infants of advancing post menstrual age (PMA) demonstrated an increase in bacterial diversity and a shift in dominance of taxa mimicking the human preterm microbiome development pattern. These characteristics along with changes in a number of metabolites as the microbiome matured influenced associative learning and memory but not locomotor ability, anxiety-like behaviors, or social interaction in adult mice. As a regulator of learning and memory, brain glial cell-derived neurotrophic factor increased with advancing PMA and was also associated with better performance in associative learning and memory in adult mice. We conclude that maturation of the microbiome in early life of preterm infants primes adult associative memory and learning ability. Our findings suggest a critical window of early intervention to affect maturation of the preterm infant microbiome and ultimately improve neurodevelopmental outcomes.


Assuntos
Microbioma Gastrointestinal , Microbiota , Animais , Bactérias , Encéfalo , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Camundongos
10.
Epidemiology ; 33(1): 131-140, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34561347

RESUMO

RATIONALE: Asthma and obesity often co-occur. It has been hypothesized that asthma may contribute to childhood obesity onset. OBJECTIVES: To determine if childhood asthma is associated with incident obesity and examine the role of asthma medication in this association. METHODS: We studied 8,716 children between ages 6 and 18.5 years who were nonobese at study entry participating in 18 US cohorts of the Environmental influences on Child Health Outcomes program (among 7,299 children with complete covariate data mean [SD] study entry age = 7.2 [1.6] years and follow up = 5.3 [3.1] years). MEASUREMENTS AND MAIN RESULTS: We defined asthma based on caregiver report of provider diagnosis. Incident obesity was defined as the first documented body mass index ≥95th percentile for age and sex following asthma status ascertainment. Over the study period, 26% of children had an asthma diagnosis and 11% developed obesity. Cox proportional hazards models with sex-specific baseline hazards were fitted to assess the association of asthma diagnosis with obesity incidence. Children with asthma had a 23% (95% confidence intervals [CI] = 4, 44) higher risk for subsequently developing obesity compared with those without asthma. A novel mediation analysis was also conducted to decompose the total asthma effect on obesity into pathways mediated and not mediated by asthma medication use. Use of asthma medication attenuated the total estimated effect of asthma on obesity by 64% (excess hazard ratios = 0.64; 95% CI = -1.05, -0.23). CONCLUSIONS: This nationwide study supports the hypothesis that childhood asthma is associated with later risk of obesity. Asthma medication may reduce this association and merits further investigation as a potential strategy for obesity prevention among children with asthma.


Assuntos
Asma , Obesidade Infantil , Adolescente , Asma/epidemiologia , Índice de Massa Corporal , Criança , Feminino , Humanos , Incidência , Masculino , Obesidade Infantil/complicações , Obesidade Infantil/epidemiologia , Modelos de Riscos Proporcionais , Fatores de Risco
11.
Gut Microbes ; 13(1): 1997560, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34839801

RESUMO

The early life microbiome plays critical roles in host development, shaping long-term outcomes including brain functioning. It is not known which initial infant colonizers elicit optimal neurodevelopment; thus, this study investigated the association between gut microbiome succession from the first week of life and head circumference growth (HCG), the earliest validated marker for neurodevelopment. Fecal samples were collected weekly from a preterm infant cohort during their neonatal intensive care unit stay and subjected to 16S rRNA gene sequencing for evaluating gut microbiome composition, in conjunction with clinical data and head circumference measurements. Preterm infants with suboptimal HCG trajectories had a depletion in the abundance/prevalence of Bacteroidota and Lachnospiraceae, independent of morbidity and caloric restriction. The severity of gut microbiome depletion matched the timing of significant HCG pattern separation between study groups at 30-week postmenstrual age demonstrating a potential mediating relationship resultant from clinical practices. Consideration of the clinical variables indicated that optimal infant microbiome succession is primarily driven by dispersal limitation (i.e., delivery mode) and secondarily by habitat filtering (i.e., antibiotics and enteral feeding). Bacteroidota and Lachnospiraceae are known core taxa of the adult microbiome, with roles in dietary glycan foraging, beneficial metabolite production and immunity, and our work provides evidence that their integration into the gut microbiome needs to occur early for optimal neurodevelopment.


Assuntos
Bacteroidetes/fisiologia , Desenvolvimento Infantil/fisiologia , Clostridiales/fisiologia , Microbioma Gastrointestinal/fisiologia , Antibacterianos/uso terapêutico , Bacteroidetes/isolamento & purificação , Clostridiales/isolamento & purificação , Parto Obstétrico , Nutrição Enteral , Fezes/microbiologia , Feminino , Cabeça/crescimento & desenvolvimento , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Masculino
12.
Trends Mol Med ; 27(12): 1175-1186, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34518093

RESUMO

Despite improvements in survival for very low birthweight (VLBW) premature infants, there continues to be significant morbidity for these infants at remarkable cost to the healthcare system. Concurrent development of the preterm infant intestine alongside the gut microbiome in the clinical setting rather than in the protected in utero environment where it would usually occur creates significant vulnerabilities for the infant's immature intestine and immune system, resulting in devastating illness and neurological injury. However, the microbiome also has the capacity to promote healthy development. Studies of parallel gut microbiome and preterm infant development have given key insight into the impact of the microbiome on intestinal as well as neural development and may provide potential therapeutic targets to prevent preterm infant morbidities.


Assuntos
Microbioma Gastrointestinal , Saúde do Lactente , Criança , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro
13.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161260

RESUMO

Individuals who are minoritized as a result of race, sexual identity, gender, or socioeconomic status experience a higher prevalence of many diseases. Understanding the biological processes that cause and maintain these socially driven health inequities is essential for addressing them. The gut microbiome is strongly shaped by host environments and affects host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities. Nevertheless, few studies have directly integrated the gut microbiome into investigations of health inequities. Here, we argue that accounting for host-gut microbe interactions will improve understanding and management of health inequities, and that health policy must begin to consider the microbiome as an important pathway linking environments to population health.


Assuntos
Microbioma Gastrointestinal , Disparidades nos Níveis de Saúde , Doença , Saúde , Humanos , Saúde Mental , Publicações
14.
Gastroenterology ; 160(2): 495-506, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33307032

RESUMO

The composition of the intestinal microbiome affects health from the prenatal period throughout childhood, and many diseases have been associated with dysbiosis. The gut microbiome is constantly changing, from birth throughout adulthood, and several variables affect its development and content. Features of the intestinal microbiota can affect development of the brain, immune system, and lungs, as well as body growth. We review the development of the gut microbiome, proponents of dysbiosis, and interactions of the microbiota with other organs. The gut microbiome should be thought of as an organ system that has important effects on childhood development. Dysbiosis has been associated with diseases in children and adults, including autism, attention deficit hyperactivity disorder, asthma, and allergies.


Assuntos
Desenvolvimento Infantil/fisiologia , Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Adolescente , Peso Corporal/fisiologia , Sistema Nervoso Central/crescimento & desenvolvimento , Criança , Pré-Escolar , Disbiose/microbiologia , Meio Ambiente , Feminino , Saúde , Nível de Saúde , Humanos , Sistema Imunitário/crescimento & desenvolvimento , Sistema Imunitário/fisiopatologia , Lactente , Recém-Nascido , Pulmão/crescimento & desenvolvimento , Pulmão/fisiologia , Pulmão/fisiopatologia
16.
J AAPOS ; 24(4): 236-238, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32707176

RESUMO

In this study, 6 infants with type 1 retinopathy of prematurity (ROP) were compared with 4 high-risk preterm neonates without any ROP but similar baseline neonatal comorbidities. The infants with type-1 ROP showed significant enrichment of Enterobacteriaceae at 28 weeks' postmenstrual age. Several metabolic pathways, including several amino acid metabolism pathways, were enriched in gut microbiota of infants without ROP. Based on these findings, we posit a possible association between early gut microbiome profile and ROP pathogenesis. Furthermore, it is possible that absence of Enterobacteriaceae overabundance, in addition to enrichment of amino acid biosynthesis pathways, may protect against severe ROP in high-risk preterm infants.


Assuntos
Microbioma Gastrointestinal , Retinopatia da Prematuridade , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Retinopatia da Prematuridade/prevenção & controle
17.
Sci Rep ; 10(1): 8178, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32424168

RESUMO

Neonatal morbidities are associated with long term neurological deficits in life and have also been associated with dysbiosis. We tested whether optimizing the neonate's microbiome through maternal probiotic supplementation can improve offspring's neurodevelopmental outcomes. Maternal LB supplementation, carried out by giving Lactobacillus acidophilus and Bifidobacterium infantis (LB) to pregnant C57/BL6J mice daily from E16 to weaning, significantly suppressed postnatal peripheral proinflammatory insult-induced systemic inflammation and normalized compromised blood-brain barrier permeability and tight junction protein expression in the offspring at pre-weaned age. Maternal LB exposure also regulated markers associated with leukocyte transendothelial migration, extracellular matrix injury and neuroinflammation. The suppressed neuroinflammation by maternal LB supplementation was associated with reduced astrocyte/microglia activation and downregulation of the transcriptional regulators CEBPD and IκBα. Furthermore, maternal LB supplementation promoted neuronal and oligodendrocyte progenitor cell development. Our study demonstrates the efficacy of maternal LB supplementation in modulating systemic and central nervous system inflammation as well as promoting neural/oligodendrocyte progenitor development in the offspring. This evidence suggests that maternal probiotic supplementation may be a safe and effective strategy to improve neurological outcomes in the offspring.


Assuntos
Encéfalo/crescimento & desenvolvimento , Doenças do Recém-Nascido/prevenção & controle , Probióticos/administração & dosagem , Substâncias Protetoras/administração & dosagem , Animais , Animais Recém-Nascidos , Bifidobacterium longum subspecies infantis/fisiologia , Encéfalo/imunologia , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proteína delta de Ligação ao Facilitador CCAAT/imunologia , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/genética , Doenças do Recém-Nascido/imunologia , Lactobacillus acidophilus/fisiologia , Masculino , Herança Materna/efeitos dos fármacos , Camundongos , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/imunologia , Gravidez
18.
Sci Rep ; 10(1): 6692, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317678

RESUMO

Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of incompletely understood pathophysiology predominantly affecting premature infants. While NEC is associated with microbial invasion of intestinal tissues, and mucus modulates interactions between microbes and underlying tissues, variations in mucus barrier properties with NEC-associated risk factors have not been investigated. This study explored differences in mucus composition (total protein, DNA, mucin content, sialic acid, and immunoregulatory proteins), as well as structural and transport properties, assessed by tracking of particles and bacteria (E. coli and E. cloacae) with developmental age and exposure to NEC stressors in Sprague Dawley rats. Early developmental age (5 day old) was characterized by a more permeable mucus layer relative to 21 day old pups, suggesting immaturity may contribute to exposure of the epithelium to microbes. Exposure to NEC stressors was associated with reduced mucus permeability, which may aid in survival. Feeding with breastmilk as opposed to formula reduces incidence of NEC. Thus, NEC-stressed (N-S) rat pups were orally dosed with breastmilk components lysozyme (N-S-LYS) or docosahexaenoic acid (N-S-DHA). N-S-LYS and N-S-DHA pups had a less permeable mucus barrier relative to N-S pups, which suggests the potential of these factors to strengthen the mucus barrier and thus protect against disease.


Assuntos
Envelhecimento/patologia , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/uso terapêutico , Enterocolite Necrosante/tratamento farmacológico , Muco/metabolismo , Muramidase/administração & dosagem , Muramidase/uso terapêutico , Estresse Fisiológico , Administração Oral , Animais , DNA/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Enterobacter cloacae/fisiologia , Enterocolite Necrosante/microbiologia , Escherichia coli/fisiologia , Fucose/metabolismo , Íleo/patologia , Íleo/ultraestrutura , Imunoglobulina G/metabolismo , Mucinas/metabolismo , Muco/efeitos dos fármacos , Muramidase/farmacologia , Ácido N-Acetilneuramínico/metabolismo , Permeabilidade , Polietilenoglicóis/química , Ratos Sprague-Dawley , Estresse Fisiológico/efeitos dos fármacos
19.
Clin Perinatol ; 46(1): 29-38, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30771817

RESUMO

Necrotizing enterocolitis is a major cause of mortality and morbidity in the preterm infant population. The gut microbiome is of particular interest in research surrounding necrotizing enterocolitis, because variations in the intestinal microbiota seem to correlate with the risk of inflammation and disease. Recent advances in non-culture-based genomic sequencing have also allowed for more intricate analyses of the intestinal microbiome. Its evolution seems to be influenced by intrauterine and extrauterine factors, ranging from antenatal antibiotic exposure to type of enteral feeds. Ultimately, these alterations in the gut microbiome have the potential to result in devastating diseases like necrotizing enterocolitis.


Assuntos
Antibacterianos/uso terapêutico , Cesárea , Disbiose/microbiologia , Nutrição Enteral , Enterocolite Necrosante/microbiologia , Microbioma Gastrointestinal , Fórmulas Infantis , Leite Humano , Parto Obstétrico , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Placenta/microbiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal
20.
Adv Exp Med Biol ; 1125: 25-36, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30680646

RESUMO

Bacterial colonization patterns in preterm infants differ from those of their term counterparts due to maternal microbial diversity, delivery mode, feeding methods, antibiotic use, and exposure to commensal microbiota and pathogens in the neonatal intensive care unit (NICU). Early gut microbiome dysbiosis predisposes neonates to necrotizing enterocolitis (NEC), a devastating intestinal disease with high morbidity and mortality. Though mechanisms of NEC pathogenesis are not fully understood, the microbiome is a promising therapy target for prevention and treatment. Direct administration of probiotics to preterm infants has been shown to reduce the incidence of NEC, but is not without risk. The immature immune systems of preterm infants leave them vulnerable to even beneficial bacteria. Further research is required to investigate both short-term and long-term effects of probiotic administration to preterm infants. Other methods of altering the preterm infant microbiome must also be considered, including breastfeeding, prebiotics, and targeting the maternal microbiome.


Assuntos
Disbiose/fisiopatologia , Enterocolite Necrosante/microbiologia , Microbioma Gastrointestinal , Doenças do Prematuro/microbiologia , Recém-Nascido Prematuro , Probióticos/uso terapêutico , Humanos , Recém-Nascido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...