Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
Methods ; 149: 31-41, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30102990

RESUMO

The study of complex microbial communities has become a major research focus as mounting evidence suggests the pivotal role microbial communities play in host health and disease. Microbial communities of the gastrointestinal tract, known as the gut microbiota, have been implicated in aiding the host with vitamin biosynthesis, regulation of host energy metabolism, immune system development, and resistance to pathogen invasion. Conversely, disruptions of the gut microbiota have been linked to host morbidity, including the development of inflammatory diseases, metabolic disorders, increased cardiovascular risk, and increased risk of infectious diseases. However, studying the gut microbiota in humans and animals is challenging, as many microorganisms are fastidious with unique nutritional or environmental requirements that are often not met using conventional culture techniques. Bioreactors provide a unique solution to overcome some of the limitations of conventional culture techniques. Bioreactors have been used to propagate and establish complex microbial communities in vitro by recapitulating the physiological conditions found in the GI tract. These systems further our understanding of microbial physiology and facilitate our understanding of the impact of medications and xenobiotics on microbial communities. Here, we review the versatility and breadth of bioreactor systems that are currently available and how they are being used to study faecal and defined microbial communities. Bioreactors provide a unique opportunity to study complex microbial interactions and perturbations in vitro in a controlled environment without confounding biotic and abiotic variables.


Assuntos
Reatores Biológicos/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Preparações Farmacêuticas/administração & dosagem , Antibacterianos/farmacologia , Gastroenteropatias/metabolismo , Gastroenteropatias/microbiologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos
5.
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
6.
Dev Psychobiol ; 61(5): 739-751, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30460694

RESUMO

Dysbiosis of the gut microbiome in preterm infants predisposes the neonate to various major morbidities including neonatal necrotizing enterocolitis and sepsis in the neonatal intensive care unit, and adverse neurological outcomes later in life. There are parallel early developmental windows for the gut microbiota and the nervous system during prenatal to postnatal of life. Therefore, preterm infants represent a unique population in which optimization of initial colonization and microbiota development can affect brain development and enhance neurological outcomes. In this review, we will first discuss the factors affecting the assembly of neonatal gut microbiota and the contribution of dysbiosis in preterm infants to neuroinflammation and neurodevelopmental disorders. We then will discuss the emerging pathways connecting the gut microbiome and brain development. Further we will discuss the significance of current models for alteration of the gut microbiome (including humanized gnotobiotic models and exposure to antibiotics) to brain development and functions. Understanding the role of early optimization of the microbiome in brain development is of paramount importance for developing microbiome-targeted therapies and protecting infants from prematurity-related neurodevelopmental diseases.


Assuntos
Encéfalo/crescimento & desenvolvimento , Microbioma Gastrointestinal/fisiologia , Recém-Nascido Prematuro/crescimento & desenvolvimento , Humanos , Lactente , Recém-Nascido
7.
Pediatr Res ; 82(5): 829-838, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28665922

RESUMO

BackgroundThe preterm infant gut microbiota is vulnerable to different biotic and abiotic factors. Although the development of this microbiota has been extensively studied, the mobilome-i.e. the mobile genetic elements (MGEs) in the gut microbiota-has not been considered. Therefore, the aim of this study was to investigate the association of the mobilome with birth weight and hospital location in the preterm infant gut microbiota.MethodsThe data set consists of fecal samples from 62 preterm infants with and without necrotizing enterocolitis (NEC) from three different hospitals. We analyzed the gut microbiome by using 16S rRNA amplicon sequencing, shot-gun metagenome sequencing, and quantitative PCR. Predictive models and other data analyses were performed using MATLAB and QIIME.ResultSThe microbiota composition was significantly different between NEC-positive and NEC-negative infants and significantly different between hospitals. An operational taxanomic unit (OTU) showed strong positive and negative correlation with NEC and birth weight, respectively, whereas none showed significance for mode of delivery. Metagenome analyses revealed high levels of conjugative plasmids with MGEs and virulence genes. Results from quantitative PCR showed that the plasmid signature genes were significantly different between hospitals and in NEC-positive infants.ConclusionOur results point toward an association of the mobilome with hospital location in preterm infants.


Assuntos
Peso ao Nascer , DNA Bacteriano/genética , Enterocolite Necrosante/microbiologia , Microbioma Gastrointestinal/genética , Trato Gastrointestinal/microbiologia , Recém-Nascido Prematuro , Recém-Nascido de muito Baixo Peso , Sequências Repetitivas Dispersas , Nascimento Prematuro/microbiologia , Estudos de Casos e Controles , Biologia Computacional , Bases de Dados Genéticas , Enterocolite Necrosante/diagnóstico , Enterocolite Necrosante/epidemiologia , Fezes/microbiologia , Feminino , Genoma Bacteriano , Idade Gestacional , Humanos , Recém-Nascido , Masculino , Metagenoma , Metagenômica/métodos , Nascimento Prematuro/diagnóstico , Nascimento Prematuro/epidemiologia , Ribotipagem , Estados Unidos/epidemiologia
8.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G521-32, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27492329

RESUMO

Development of the infant small intestine is influenced by bacterial colonization. To promote establishment of optimal microbial communities in preterm infants, knowledge of the beneficial functions of the early gut microbiota on intestinal development is needed. The purpose of this study was to investigate the impact of early preterm infant microbiota on host gut development using a gnotobiotic mouse model. Histological assessment of intestinal development was performed. The differentiation of four epithelial cell lineages (enterocytes, goblet cells, Paneth cells, enteroendocrine cells) and tight junction (TJ) formation was examined. Using weight gain as a surrogate marker for health, we found that early microbiota from a preterm infant with normal weight gain (MPI-H) induced increased villus height and crypt depth, increased cell proliferation, increased numbers of goblet cells and Paneth cells, and enhanced TJs compared with the changes induced by early microbiota from a poor weight gain preterm infant (MPI-L). Laser capture microdissection (LCM) plus qRT-PCR further revealed, in MPI-H mice, a higher expression of stem cell marker Lgr5 and Paneth cell markers Lyz1 and Cryptdin5 in crypt populations, along with higher expression of the goblet cell and mature enterocyte marker Muc3 in villus populations. In contrast, MPI-L microbiota failed to induce the aforementioned changes and presented intestinal characteristics comparable to a germ-free host. Our data demonstrate that microbial communities have differential effects on intestinal development. Future studies to identify pioneer settlers in neonatal microbial communities necessary to induce maturation may provide new insights for preterm infant microbial ecosystem therapeutics.


Assuntos
Mucosa Intestinal/microbiologia , Microbiota/genética , Animais , Vida Livre de Germes , Humanos , Camundongos , Camundongos Endogâmicos C57BL
9.
Gut ; 64(7): 1082-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25080448

RESUMO

OBJECTIVE: Vitamin D and the vitamin D receptor (VDR) appear to be important immunological regulators of inflammatory bowel diseases (IBD). Defective autophagy has also been implicated in IBD, where interestingly, polymorphisms of genes such as ATG16L1 have been associated with increased risk. Although vitamin D, the microbiome and autophagy are all involved in pathogenesis of IBD, it remains unclear whether these processes are related or function independently. DESIGN: We investigated the effects and mechanisms of intestinal epithelial VDR in healthy and inflamed states using cell culture models, a conditional VDR knockout mouse model (VDR(ΔIEC)), colitis models and human samples. RESULTS: Absence of intestinal epithelial VDR affects microbial assemblage and increases susceptibility to dextran sulfate sodium-induced colitis. Intestinal epithelial VDR downregulates expressions of ATG16L1 and lysozyme, and impairs antimicrobial function of Paneth cells. Gain and loss-of-function assays showed that VDR levels regulate ATG16L1 and lysozyme at the transcriptional and translational levels. Moreover, low levels of intestinal epithelial VDR correlated with reduced ATG16L1 and representation by intestinal Bacteroides in patients with IBD. Administration of the butyrate (a fermentation product of gut microbes) increases intestinal VDR expression and suppresses inflammation in a colitis model. CONCLUSIONS: Our study demonstrates fundamental relationship between VDR, autophagy and gut microbial assemblage that is essential for maintaining intestinal homeostasis, but also in contributing to the pathophysiology of IBD. These insights can be leveraged to define therapeutic targets for restoring VDR expression and function.


Assuntos
Autofagia/fisiologia , Colite/fisiopatologia , Mucosa Intestinal/metabolismo , Receptores de Calcitriol/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Colite/imunologia , Regulação para Baixo/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Imunoprecipitação , Mucosa Intestinal/imunologia , Camundongos Endogâmicos , Camundongos Knockout , Pessoa de Meia-Idade , Celulas de Paneth/metabolismo , Receptores de Calcitriol/imunologia
10.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G341-9, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26159695

RESUMO

Low expression of vitamin D receptor (VDR) and dysfunction of vitamin D/VDR signaling are reported in patients with inflammatory bowel disease (IBD); therefore, restoration of VDR function to control inflammation in IBD is desirable. Probiotics have been used in the treatment of IBD. However, the role of probiotics in the modulation of VDR signaling to effectively reduce inflammation is unknown. We identified a novel role of probiotics in activating VDR activity, thus inhibiting inflammation, using cell models and VDR knockout mice. We found that the probiotics Lactobacillus rhamnosus strain GG (LGG) and Lactobacillus plantarum (LP) increased VDR protein expression in both mouse and human intestinal epithelial cells. Using the VDR luciferase reporter vector, we detected increased transcriptional activity of VDR after probiotic treatment. Probiotics increased the expression of the VDR target genes, such as antimicrobial peptide cathelicidin, at the transcriptional level. Furthermore, the role of probiotics in regulating VDR signaling was tested in vivo using a Salmonella-colitis model in VDR knockout mice. Probiotic treatment conferred physiological and histologic protection from Salmonella-induced colitis in VDR(+/+) mice, whereas probiotics had no effects in the VDR(-/-) mice. Probiotic treatment also enhanced numbers of Paneth cells, which secrete AMPs for host defense. These data indicate that the VDR pathway is required for probiotic protection in colitis. Understanding how probiotics enhance VDR signaling and inhibit inflammation will allow probiotics to be used effectively, resulting in innovative approaches to the prevention and treatment of chronic inflammation.


Assuntos
Colite Ulcerativa/metabolismo , Microbiota , Probióticos/farmacologia , Receptores de Calcitriol/metabolismo , Animais , Colite Ulcerativa/microbiologia , Colite Ulcerativa/prevenção & controle , Feminino , Células HCT116 , Humanos , Lactobacillus plantarum , Lacticaseibacillus rhamnosus , Camundongos , Camundongos Endogâmicos C57BL , Celulas de Paneth/efeitos dos fármacos , Celulas de Paneth/metabolismo , Probióticos/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Calcitriol/genética
11.
J Biol Chem ; 288(48): 34757-66, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24129565

RESUMO

Inflammatory immune responses play an important role in mucosal homeostasis and gut diseases. Nuclear factor κB (NF-κB), central to the proinflammatory cascade, is activated in necrotizing enterocolitis (NEC), a devastating condition of intestinal injury with extensive inflammation in premature infants. TGF-ß is a strong immune suppressor and a factor in breast milk, which has been shown to be protective against NEC. In an NEC animal model, oral administration of the isoform TGF-ß1 activated the downstream effector Smad2 in intestine and significantly reduced NEC incidence. In addition, TGF-ß1 suppressed NF-κB activation, maintained levels of the NF-κB inhibitor IκBα in the intestinal epithelium, and systemically decreased serum levels of IL-6 and IFN-γ. The immature human fetal intestinal epithelial cell line H4 was used as a reductionistic model of the immature enterocyte to investigate mechanism. TGF-ß1 pretreatment inhibited the TNF-α-induced IκBα phosphorylation that targets the IκBα protein for degradation and inhibited NF-κB activation. Chromatin immunoprecipitation (ChIP) assays demonstrated decreased NF-κB binding to the promoters of IL-6, IL-8, and IκBα in response to TNF-α with TGF-ß1 pretreatment. These TGF-ß1 effects appear to be mediated through the canonical Smad pathway as silencing of the TGF-ß central mediator Smad4 resulted in loss of the TGF-ß1 effects. Thus, TGF-ß1 is capable of eliciting anti-inflammatory effects by inhibiting NF-κB specifically in the intestinal epithelium as well as by decreasing systemic IL-6 and IFN-γ levels. Oral administration of TGF-ß1 therefore can potentially be used to protect against gastrointestinal diseases.


Assuntos
Gastroenteropatias/tratamento farmacológico , Inflamação/metabolismo , NF-kappa B/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/administração & dosagem , Administração Oral , Animais , Linhagem Celular , Gastroenteropatias/metabolismo , Gastroenteropatias/patologia , Humanos , Imunidade Inata/genética , Inflamação/tratamento farmacológico , Inflamação/genética , Interferon gama/metabolismo , Interleucina-6/metabolismo , Interleucina-8 , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Regiões Promotoras Genéticas , Ratos , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
12.
NMR Biomed ; 27(3): 272-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24318809

RESUMO

Neonatal necrotizing enterocolitis (NEC) is a poorly understood life-threatening illness afflicting premature infants. Research is hampered by the absence of a suitable method to monitor disease progression noninvasively. The primary goal of this research was to test in vivo MRI methods for the noninvasive early detection and staging of inflammation in the ileum of an infant rat model of NEC. Neonatal rats were delivered by cesarean section at embryonic stage of day 20 after the beginning of pregnancy and stressed with formula feeding, hypoxia and bacterial colonization to induce NEC. Naturally born and dam-fed neonatal rats were used as healthy controls. In vivo MRI studies were performed using a Bruker 9.4-T scanner to obtain high-resolution anatomical MR images using both gradient echo and spin echo sequences, pixel-by-pixel T2 maps using a multi-slice-multi-echo sequence, and maps of the apparent diffusion coefficient (ADC) of water using a spin echo sequence, to assess the degree of ileal damage. Pups were sacrificed at the end of the MRI experiment on day 2 or 4 for histology. T2 measured by MRI was increased significantly in the ileal regions of pups with NEC by histology (106.3 ± 6.1 ms) compared with experimentally stressed pups without NEC (85.2 ± 6.8 ms) and nonstressed, control rat pups (64.9 ± 2.3 ms). ADC values measured by diffusion-weighted MRI were also increased in the ileal regions of pups with NEC by histology [(1.98 ± 0.15) × 10(-3) mm(2)/s] compared with experimentally stressed pups without NEC [(1.43 ± 0.16) × 10(-3) mm(2)/s] and nonstressed control pups [(1.10 ± 0.06) × 10(-3) mm(2)/s]. Both T2 and ADC values between these groups were found to be significantly different (p < 0.03). The correlation of MRI results with histologic images of the excised ileal tissue samples strongly suggests that MRI can noninvasively identify NEC and assess intestinal injury prior to clinical symptoms in a physiologic rat pup model of NEC.


Assuntos
Enterocolite Necrosante/diagnóstico , Imageamento por Ressonância Magnética , Animais , Difusão , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Feminino , Intestinos/patologia , Gravidez , Ratos , Ratos Sprague-Dawley , Marcadores de Spin
13.
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
14.
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
15.
Cell Host Microbe ; 32(7): 1192-1206.e5, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38955186

RESUMO

The impact of gestational diabetes mellitus (GDM) on maternal or infant microbiome trajectory remains poorly understood. Utilizing large-scale longitudinal fecal samples from 264 mother-baby dyads, we present the gut microbiome trajectory of the mothers throughout pregnancy and infants during the first year of life. GDM mothers had a distinct microbiome diversity and composition during the gestation period. GDM leaves fingerprints on the infant's gut microbiome, which are confounded by delivery mode. Further, Clostridium species positively correlate with a larger head circumference at month 12 in male offspring but not females. The gut microbiome of GDM mothers with male fetuses displays depleted gut-brain modules, including acetate synthesis I and degradation and glutamate synthesis II. The gut microbiome of female infants of GDM mothers has higher histamine degradation and dopamine degradation. Together, our integrative analysis indicates that GDM affects maternal and infant gut composition, which is associated with sexually dimorphic infant head growth.


Assuntos
Diabetes Gestacional , Fezes , Microbioma Gastrointestinal , Feminino , Humanos , Diabetes Gestacional/microbiologia , Gravidez , Masculino , Lactente , Fezes/microbiologia , Cabeça/microbiologia , Adulto , Recém-Nascido , Clostridium/crescimento & desenvolvimento , Efeitos Tardios da Exposição Pré-Natal/microbiologia
16.
Dig Dis Sci ; 58(3): 668-77, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23329012

RESUMO

BACKGROUND: Lubiprostone has been used to treat constipation through its effects to stimulate Cl(-) secretion, resulting in water and electrolyte secretion. AIM: Potential associated changes in intestinal mucus and the colonizing bacteria (microbiome) have not been studied. As mucus obstructions may play a role in cystic fibrosis, the hypothesis that lubiprostone alters intestinal mucus and the microbiome was investigated. METHODS: Ion transport studies were performed ex vivo. For mucus and microbiome studies, mice were gavaged daily with lubiprostone or vehicle. Mucin from intestinal sections was analyzed in Carnoy's fixed tissues stained with Alcian blue. Microbiome composition was analyzed by 16S rRNA gene-based sequencing. RESULTS: Lubiprostone stimulated short circuit current in all mouse intestinal segments after both serosal and mucosal additions, albeit at lower concentrations in the latter. Current was Cl-dependent and blocked by mucosal diphenylcarboxylic acid, serosal bumetanide, and serosal Ba(++). The CFTR inhibitor CFTRinh172 had a marginal effect. Mucus near epithelial cells (inner layer mucus) was not present in the small intestine of any mice. Proximal colon inner mucus layer was thicker in ∆F/∆F compared with +/∆F and +/+ mice. Lubiprostone decreased inner mucus layer thickness in both proximal and distal colon of all mice. Furthermore, lubiprostone altered the intestinal microbiome by increasing abundance of Lactobacillus and Alistipes. CONCLUSIONS: Lubiprostone activates non-CFTR Cl(-) secretion and alters the colonic inner mucus layer, which is associated with changes in the composition of the enteric microbiome.


Assuntos
Alprostadil/análogos & derivados , Bactérias/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/microbiologia , Alprostadil/farmacologia , Animais , Ânions/metabolismo , Bactérias/classificação , Bactérias/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Lubiprostona , Camundongos , Camundongos Endogâmicos CFTR , Filogenia , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo
17.
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.

18.
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
19.
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.

20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA