Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Semin Perinatol ; 47(6): 151818, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37775366

RESUMO

Premature births account for over 10% of live births worldwide. Bronchopulmonary dysplasia (BPD) represents a severe sequela in neonates born very prematurely and remains the most common chronic neonatal lung disease, often leading to serious adverse consequences in adulthood. Nutrition plays a crucial role in lung development and repair. Ongoing research has primarily focused on the pathogenesis and prevention of BPD in preterm birth. However, infants with established BPD need specialist medical care that persists throughout their hospitalization and continues after discharge. This manuscript aims to highlight the impact of growth and nutrition on BPD and highlight research gaps to provide direction for future studies. Protective practices include ensuring adequate early energy delivery through parenteral nutrition and enteral feedings while carefully monitoring total fluid intake and the use of breast milk over formula. These nutritional strategies remain the same for infants with established BPD with the addition of limiting the use of diuretics and steroids; but if employed, monitoring carefully without compromising total energy delivery. Functional nutrient supplements with a potential protective role against BPD are revisited, despite the limited evidence of their efficacy, including vitamins, trace elements, zinc, lipids, and sphingolipids. Planning post-intensive care and outpatient longitudinal nutrition support is critical in caring for an infant with established BPD.


Assuntos
Displasia Broncopulmonar , Estado Nutricional , Nascimento Prematuro , Feminino , Humanos , Lactente , Recém-Nascido , Displasia Broncopulmonar/prevenção & controle , Displasia Broncopulmonar/etiologia , Nutrição Enteral , Leite Humano
2.
Clin Perinatol ; 50(3): 575-589, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37536765

RESUMO

Intravenous lipid emulsions (ILEs) are a source of nonprotein calories and fatty acids and help promote growth in preterm infants and infants with intestinal failure. An ILE dose and oil source determines its fatty acid, phytosterol, and vitamin E delivery. These factors play a role in the infant's risk for essential fatty acid deficiency and cholestasis, and help modulate inflammation, immunity, and organ development. This article reviews different ILEs and their constituents and their relationship with neonatal health.


Assuntos
Colestase , Emulsões Gordurosas Intravenosas , Lactente , Recém-Nascido , Humanos , Emulsões Gordurosas Intravenosas/uso terapêutico , Recém-Nascido Prematuro , Unidades de Terapia Intensiva Neonatal , Óleos de Peixe , Óleo de Soja , Nutrição Parenteral
4.
Clin Perinatol ; 49(2): 381-391, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35659092

RESUMO

Fatty acids are critical bioactives for fetal and neonatal development. Premature delivery and current nutritional strategies pose several challenges in restoring fatty acid balance in the preterm infant. The impact on fatty acid balance and outcomes using lipid emulsions, enteral nutrition, and enteral supplements are reviewed, including a summary of the most recent large clinical trials of enteral fatty acid supplementation for the preterm infant. Research gaps remain in successfully implementing nutritional strategies to optimize fatty acid status in preterm infants.


Assuntos
Ácidos Graxos , Recém-Nascido Prematuro , Nutrição Enteral , Ácidos Graxos Insaturados/uso terapêutico , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido
5.
J Clin Endocrinol Metab ; 106(6): 1793-1803, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33544860

RESUMO

CONTEXT: Human milk contains hormones that regulate metabolism. Extrauterine growth restriction remains common among preterm infants, but the effect of ingesting milk hormones on preterm infant growth is poorly understood. OBJECTIVE: To quantify associations of longitudinal exposure to leptin, adiponectin, and insulin in milk with physical growth of preterm infants. DESIGN/METHODS: In 50 preterm neonates (median gestational age 29.4 weeks), we sampled maternal milk on day-of-life 7, 14, 21, and 28 and measured hormone levels in whole milk by ELISA. Milk leptin levels were available for a subset of 18 infants. We calculated milk hormone doses by multiplying the hormone level by the milk volume ingested on each day and estimated the area under the curve (AUC) to reflect longitudinal exposure. We analyzed associations of milk hormone exposure with growth outcomes in generalized estimated equations. MAIN OUTCOME MEASURES: Weight gain velocity and z-scores in weight, length, head circumference, and body mass index at 36 weeks' postmenstrual age (PMA). RESULTS: Higher leptin intake was associated with greater weight gain (2.17g/kg/day [95% CI, 1.31, 3.02]) and weight z-score at 36 weeks' PMA (0.30 [0.08, 0.53] higher z-score per tertile). Higher adiponectin intake was associated with greater length z-score (0.41 [0.13, 0.69]), however, this association was nullified after adjustment of protein and calorie intake. Higher adiponectin was associated with smaller head circumference z-score (-0.36 [-0.64, -0.07]). Insulin was not associated with growth outcomes. CONCLUSIONS: Milk leptin and adiponectin exposures may affect growth of preterm infants. The long-term effects of milk hormones warrant further investigation.


Assuntos
Desenvolvimento Infantil/fisiologia , Ingestão de Alimentos/fisiologia , Hormônios/administração & dosagem , Leite Humano/fisiologia , Adiponectina/administração & dosagem , Adiponectina/metabolismo , Estudos de Coortes , Feminino , Hormônios/metabolismo , Humanos , Lactente , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Recém-Nascido Prematuro/crescimento & desenvolvimento , Leptina/administração & dosagem , Leptina/metabolismo , Estudos Longitudinais , Masculino , Massachusetts , Leite Humano/química , Leite Humano/metabolismo , Aumento de Peso/fisiologia
6.
J Acad Nutr Diet ; 121(11): 2287-2300.e12, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33358688

RESUMO

Adequate protein intake by very-low-birth-weight preterm infants (≤1,500 g at birth) is essential to optimize growth and development. The estimated needs for this population are the highest of all humans, however, the recommended intake has varied greatly over the past several years. A literature search was conducted in PubMed, Embase, CINAHL (Cumulative Index to Nursing and Allied Health Literature), and Cochrane Central databases to identify randomized controlled trials evaluating the effect of prescribed protein intake and identified outcomes. Articles were screened by 2 reviewers, risk of bias was assessed, data were synthesized quantitatively and narratively, and each outcome was separately graded for certainty of evidence. The literature search retrieved 25,384 articles and 2 trials were included in final analysis. No trials were identified that evaluated effect of protein amount on morbidities or mortality. Moderate certainty evidence found a significant difference in weight gain when protein intake of greater than 3.5 g/kg/day from preterm infant formula was compared with lower intakes. Low-certainty evidence found no evidence of effect of protein intake of 2.6 vs 3.1 vs 3.8 g/kg/day on length, head circumference, skinfold measurements, or mid-arm circumference. Low-certainty evidence found some improvement in development measures when higher protein intake of 3.8 vs 3.1 vs 2.6 g/kg/day were compared. Low-certainty evidence found no significant difference in bone mineral content when these protein intakes were compared. No studies were identified that compared protein intake greater than 4.0 g/kg/day. This systematic review found that protein intake between 3.5 and 4.0 g/kg/day promotes weight gain and improved development.


Assuntos
Proteínas Alimentares/administração & dosagem , Nutrição Enteral/métodos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido Prematuro/crescimento & desenvolvimento , Recém-Nascido de muito Baixo Peso/crescimento & desenvolvimento , Ingestão de Alimentos/fisiologia , Feminino , Humanos , Fórmulas Infantis/análise , Recém-Nascido , Masculino , Avaliação de Resultados em Cuidados de Saúde , Ensaios Clínicos Controlados Aleatórios como Assunto , Aumento de Peso
7.
Nutrients ; 12(2)2020 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-32092925

RESUMO

Human breast milk is the optimal source of nutrition for infant growth and development. Breast milk fats and their downstream derivatives of fatty acids and fatty acid-derived terminal mediators not only provide an energy source but also are important regulators of development, immune function, and metabolism. The composition of the lipids and fatty acids determines the nutritional and physicochemical properties of human milk fat. Essential fatty acids, including long-chain polyunsaturated fatty acids (LCPUFAs) and specialized pro-resolving mediators, are critical for growth, organogenesis, and regulation of inflammation. Combined data including in vitro, in vivo, and human cohort studies support the beneficial effects of human breast milk in intestinal development and in reducing the risk of intestinal injury. Human milk has been shown to reduce the occurrence of necrotizing enterocolitis (NEC), a common gastrointestinal disease in preterm infants. Preterm infants fed human breast milk are less likely to develop NEC compared to preterm infants receiving infant formula. Intestinal development and its physiological functions are highly adaptive to changes in nutritional status influencing the susceptibility towards intestinal injury in response to pathological challenges. In this review, we focus on lipids and fatty acids present in breast milk and their impact on neonatal gut development and the risk of disease.


Assuntos
Gorduras na Dieta/administração & dosagem , Ácidos Graxos Essenciais/administração & dosagem , Intestinos/crescimento & desenvolvimento , Lipídeos/administração & dosagem , Leite Humano/química , Enterocolite Necrosante/prevenção & controle , Feminino , Humanos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Recém-Nascido Prematuro/crescimento & desenvolvimento , Masculino , Estado Nutricional
8.
Nutrients ; 12(1)2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31906339

RESUMO

After birth, preterm infants are deficient in arachidonic acid (ARA), docosahexaenoic acid (DHA), and antioxidants, increasing their risk of oxidative stress-related pathologies. The principal aim was to evaluate if supplementation with long-chain polyunsaturated fatty acids (LCPUFAs) improves antioxidant defenses. In total, 21 preterm infants were supplemented with ARA and DHA in a 2:1 ratio (ARA:DHA-S) or with medium-chain triglycerides (MCT-S). Plasma n-3 and n-6 LCPUFAs were measured at birth, postnatal day 28, and 36 weeks of postmenstrual age (36 WPA) by gas chromatography-mass spectroscopy. Plasma antioxidants (glutathione (GSH), catalase, and thiols) and oxidative damage biomarkers (malondialdehyde (MDA), carbonyls) were analyzed at the same time points by spectrophotometry, and scores of antioxidant status (Antiox-S) and oxidative damage (Proxy-S) were calculated. At 36 WPA, linoleic acid (LA) and dihomo--linolenic acid (DGLA) were decreased in ARA:DHA-S compared to the MCT-S group (LA: ARA:DHA-S = 18.54 1.68, MCT-S = 22.80 1.41; p = 0.018; DGLA: ARA:DHA-S = 1.68 0.38, MCT-S = 2.32 0.58; p = 0.018). Furthermore, α-linolenic acid (ALA) was increased in ARA:DHA-S (ARA:DHA-S = 0.52 0.33, MCT-S = 0.22 0.10; p = 0.018). Additionally, LA:DHA ratio was decreased in the ARA:DHA-S compared to control group (ARA:DHA-S = 6.26 2.35, MCT-S = 8.21 2.65; p = 0.045). By the end of supplementation (36 WPA), catalase, thiol groups, and Antiox-S were significantly higher in neonates receiving ARA:DHA-S compared to those receiving MCT-S, with no differences in oxidative stress biomarkers. In conclusion, ARA:DHA supplementation in preterm neonates resulted in an overall improvement in antioxidant to oxidant balance and a decrease in early fatty acid precursors of the n-6 relative to the n-3 pathway. These effects may reduce oxidative stress and inflammation.


Assuntos
Antioxidantes/metabolismo , Suplementos Nutricionais , Ácidos Graxos Insaturados/administração & dosagem , Recém-Nascido Prematuro/sangue , Estresse Oxidativo/fisiologia , Ácido Araquidônico/administração & dosagem , Ácidos Docosa-Hexaenoicos/administração & dosagem , Feminino , Humanos , Recém-Nascido , Masculino , Projetos Piloto , Triglicerídeos/administração & dosagem , Ácido alfa-Linolênico/administração & dosagem
9.
JPEN J Parenter Enteral Nutr ; 44(1): 69-79, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31441521

RESUMO

BACKGROUND: Preterm delivery and current nutrition strategies result in deficiencies of critical long-chain fatty acids (FAs) and lipophilic nutrients, increasing the risk of preterm morbidities. We sought to determine the efficacy of preventing postnatal deficits in FAs and lipophilic nutrients using an enteral concentrated lipid supplement in preterm piglets. METHODS: Preterm piglets were fed a baseline diet devoid of arachidonic acid (AA) and docosahexaenoic acid (DHA) and randomized to enteral supplementation as follows: (1) Intralipid (IL), (2) complex lipid supplement 1 (CLS1) with an AA:DHA ratio of 0.25, or (3) CLS2 with an AA:DHA ratio of 1.2. On day 8, plasma and tissue levels of FAs and lipophilic nutrients were measured and ileum histology performed. RESULTS: Plasma DHA levels decreased in the IL group by day 2. In contrast, DHA increased by day 2 compared with birth levels in both CLS1 and CLS2 groups. The IL and CLS1 groups demonstrated a continued decline in AA levels during the 8-day protocol, whereas AA levels in the CLS2 group on day 8 were comparable to birth levels. Preserving AA levels in the CLS2 group was associated with greater ileal villus height and muscular layer thickness. Lipophilic nutrients were effectively absorbed in plasma and tissues. CONCLUSIONS: Enteral administration of CLS1 and CLS2 demonstrated similar increases in DHA levels compared with birth levels. Only CLS2 maintained AA birth levels. Providing a concentrated complex lipid emulsion with an AA:DHA ratio > 1 is important in preventing postnatal AA deficits.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Ácidos Araquidônicos/metabolismo , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/metabolismo , Nutrição Enteral/veterinária , Ração Animal , Animais , Animais Recém-Nascidos , Ácidos Araquidônicos/deficiência , Ácidos Docosa-Hexaenoicos/deficiência , Emulsões/administração & dosagem , Nutrientes , Distribuição Aleatória , Suínos
10.
J Nutr ; 149(10): 1724-1731, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31179494

RESUMO

BACKGROUND: Developmental expression of fatty acid transporters and their role in polyunsaturated fatty acid concentrations in the postnatal period have not been evaluated. OBJECTIVE: We hypothesized that transporter expression is developmentally regulated, tissue-specific, and that expression can modulate fatty acid accretion independently of diet. METHODS: Brain and lung transporter expression were quantified in C57BL/6 wild-type (WT) and Fat1 mice. Pups were dam-fed until day 21. Dams were fed AIN-76A 10% corn oil to represent a typical North American/European diet. After weaning, mice were fed the same diet as dams. Gene expression of Fatp1, Fatp4, Fabp5, and Fat/cd36 was quantified by quantitative reverse transcriptase-polymerase chain reaction. Fatty acid concentrations were measured by GC-MS. RESULTS: Brain docosahexaenoic acid (DHA) concentrations increased from day 3 to day 28 in both genotypes, with higher concentrations at days 3 and 14 in Fat1 than in WT mice [median (IQR)]: 10.7 (10.6-11.2) mol% compared with 6.6 (6.4-7.2) mol% and 12.5 (12.4-12.9) mol% compared with 8.9 (8.7-9.1) mol%, respectively; P < 0.05). During DHA accrual, transporter expression decreased. Fold changes in brain Fatp4, Fabp5, and Fat/cd36 were inversely correlated with fold changes in brain DHA concentrations in Fat1 relative to WT mice (ρ = -0.85, -0.75, and -0.78, respectively; P ≤ 0.001). Lung DHA concentrations were unchanged across the 3 time points for both genotypes. Despite unchanging DHA concentrations, there was increased expression of Fatp1 at days 14 and 28 (5-fold), Fatp4 at day 14 (2.3-fold), and Fabp5 at day 14 (3.8-fold) relative to day 3 in Fat1 mice. In WT mice, Fatp1 increased almost 5-fold at day 28 relative to day 3. There was no correlation between lung transporters and DHA concentrations in Fat1 relative to WT mice. CONCLUSIONS: Development of fatty acid transporter expression in C57BL/6 WT and Fat1 mice is genotype and tissue specific. Further, postnatal accretion of brain DHA appears independent of transporter status, with tissue concentrations representing dietary contributions.


Assuntos
Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/farmacologia , Proteínas de Transporte de Ácido Graxo/metabolismo , Pulmão/metabolismo , Animais , Óleo de Milho/administração & dosagem , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Ácidos Docosa-Hexaenoicos/metabolismo , Proteínas de Transporte de Ácido Graxo/genética , Ácidos Graxos/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/isolamento & purificação
11.
Semin Fetal Neonatal Med ; 22(1): 8-14, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27599697

RESUMO

Fatty acids are critical nutrient regulators of intracellular signaling and influence key pathways including inflammatory responses, hemostasis as well as central nervous system development and function. Preterm birth interrupts the maternal-fetal transfer of essential fatty acids including docosahexaenoic and arachidonic acids, which occurs during the third trimester. Postnatal deficits of these nutrients accrue in preterm infants during the first week and they remain throughout the first months. Due to the regulatory roles of these fatty acids, such deficits contribute an increased risk of developing prematurity-related morbidities including impaired growth and neurodevelopment. The fatty acid contents of parenteral and enteral nutrition are insufficient to meet current recommendations. This chapter summarizes the regulatory roles of fatty acids, current recommendations and limitations of parenteral and enteral nutrition in meeting these recommendations in preterm infants. Suggested areas for research on the roles of fatty acids in preterm infant health are also provided.


Assuntos
Desenvolvimento Infantil/fisiologia , Ácidos Graxos/fisiologia , Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Nutrição Enteral , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Nutrição Parenteral
12.
Clin Perinatol ; 42(4): 797-806, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26593079

RESUMO

Bronchopulmonary dysplasia (BPD) remains a common morbidity of prematurity. Although the pathogenesis of BPD is recognized to be both multifactorial and complex, the role of nutrition in the pathophysiology of BPD is typically limited to management after a diagnosis has been made. Infants born small for gestational age and those who experience postnatal growth failure are more likely to have BPD. Therapies for lung disease, such as fluid restriction, diuretics, and corticosteroids, can negatively impact postnatal growth. Future research is needed to optimize nutritional strategies in the neonatal intensive care unit and following hospital discharge.


Assuntos
Displasia Broncopulmonar , Recém-Nascido Prematuro/crescimento & desenvolvimento , Estado Nutricional , Nutrição Parenteral/métodos , Displasia Broncopulmonar/fisiopatologia , Displasia Broncopulmonar/terapia , Desenvolvimento Infantil , Suplementos Nutricionais , Idade Gestacional , Humanos , Recém-Nascido
13.
Clin Perinatol ; 41(2): 363-82, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24873838

RESUMO

Challenges remain in optimizing the delivery of fatty acids to attain their nutritional and therapeutic benefits in neonatal health. In this review, knowledge about placental transfer of fatty acids to the developing fetus is summarized, the potential role and mechanisms of fatty acids in enhancing neonatal health and minimizing morbidities is outlined, the unique considerations for fatty acid delivery in the preterm population are defined, and the research questions are proposed that need to be addressed before new standards of care are adopted at the bedside for the provision of critical fatty acids to preterm infants.


Assuntos
Ácidos Graxos/farmacologia , Fenômenos Fisiológicos da Nutrição do Lactente/fisiologia , Recém-Nascido Prematuro/crescimento & desenvolvimento , Apoio Nutricional/métodos , Humanos , Recém-Nascido
14.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L599-606, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20656894

RESUMO

Cystic fibrosis (CF) patients display a fatty acid imbalance characterized by low linoleic acid levels and variable changes in arachidonic acid. This led to the recommendation that CF patients consume a high-fat diet containing >6% linoleic acid. We hypothesized that increased conversion of linoleic acid to arachidonic acid in CF leads to increased levels of arachidonate-derived proinflammatory metabolites and that this process is exacerbated by increasing linoleic acid levels in the diet. To test this hypothesis, we determined the effect of linoleic acid supplementation on downstream proinflammatory biomarkers in two CF models: 1) in vitro cell culture model using 16HBE14o(-) sense [wild-type (WT)] and antisense (CF) human airway epithelial cells; and 2) in an in vivo model using cftr(-/-) transgenic mice. Fatty acids were analyzed by gas chromatography-mass spectrometry (GC/MS), and IL-8 and eicosanoids were measured by ELISA. Neutrophils were quantified in bronchoalveolar lavage fluid from knockout mice following linoleic acid supplementation and exposure to aerosolized Pseudomonas LPS. Linoleic acid supplementation increased arachidonic acid levels in CF but not WT cells. IL-8, PGE(2), and PGF(2α) secretion were increased in CF compared with WT cells, with a further increase following linoleic acid supplementation. cftr(-/-) Mice supplemented with 100 mg of linoleic acid had increased arachidonic acid levels in lung tissue associated with increased neutrophil infiltration into the airway compared with control mice. These findings support the hypothesis that increasing linoleic acid levels in the setting of loss of cystic fibrosis transmembrane conductance regulator (CFTR) function leads to increased arachidonic acid levels and proinflammatory mediators.


Assuntos
Ácido Araquidônico/biossíntese , Fibrose Cística/dietoterapia , Suplementos Nutricionais , Eicosanoides/biossíntese , Ácido Linoleico/administração & dosagem , Mucosa Respiratória/citologia , Animais , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Fibrose Cística/fisiopatologia , Modelos Animais de Doenças , Eicosanoides/metabolismo , Ácidos Graxos/metabolismo , Humanos , Inflamação/fisiopatologia , Interleucina-8/imunologia , Interleucina-8/metabolismo , Ácido Linoleico/uso terapêutico , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos CFTR , Camundongos Knockout , Camundongos Transgênicos , Pseudomonas aeruginosa/imunologia
15.
Semin Perinatol ; 32(2): 127-37, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18346537

RESUMO

Necrotizing enterocolitis (NEC) is an inflammatory bowel disease largely affecting low birth weight, premature infants. Once acquired, NEC is accompanied by significant mortality and morbid sequelae. Our understanding of the pathophysiology of NEC continues to evolve, and the development of NEC is likely multifactorial with resultant bowel injury mediated through a final, common inflammatory pathway. The predisposition for NEC appears to involve the interplay between intestinal integrity and function, enteral feeding and bacterial colonization, and regulation of the gastrointestinal and systemic inflammatory response. Commensal organisms or probiotics have been shown to be crucial in the development and modulation of each of these factors within the intestinal epithelium. As a result, probiotic supplementation has been proposed as a promising new intervention for the prevention of NEC. To understand the potential utility of probiotics in NEC, we will discuss: the components of gut defense; the role of the intestinal ecosystem in modulating immunity and inflammation; bacterial colonization patterns in the preterm infant compared with patterns seen in the healthy, full-term infant; the evidence for probiotic use in other populations and diseases; and finally, the evidence of probiotic use specific to the preterm infant and NEC.


Assuntos
Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/prevenção & controle , Intestinos/microbiologia , Probióticos/uso terapêutico , Suplementos Nutricionais , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Ensaios Clínicos Controlados Aleatórios como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA