Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 15(30): 35847-35859, 2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-37480336

RESUMEN

Colostrum provides bioactive components that are essential for the colonization of microbiota in the infant gut, while preventing infectious diseases such as necrotizing enterocolitis. As colostrum is not always available from the mother, particularly for premature infants, effective and safe substitutes are keenly sought after by neonatologists. The benefits of bioactive factors in colostrum are recognized; however, there have been no accounts of human colostrum being studied during digestion of the lipid components or their self-assembly in gastrointestinal environments. Due to the weaker bile pool in infants than adults, evaluating the lipid composition of human colostrum and linking it to structural self-assembly behavior is important in these settings and thus enabling the formulation of substitutes for colostrum. This study is aimed at the rational design of an appropriate lipid component for a colostrum substitute and determining the ability of this formulation to reduce inflammation in intestinal cells. Gas chromatography was utilized to map lipid composition. The self-assembly of lipid components occurring during digestion of colostrum was monitored using small-angle X-ray scattering for comparison with substitute mixtures containing pure triglyceride lipids based on their abundance in colostrum. The digestion profiles of human colostrum and the substitute mixtures were similar. Subtle differences in lipid self-assembly were evident, with the substitute mixtures exhibiting additional non-lamellar phases, which were not seen for human colostrum. The difference is attributable to the distribution of free fatty acids released during digestion. The biological markers of necrotizing enterocolitis were modulated in cells that were treated with bifidobacteria cultured on colostrum substitute mixtures, compared to those treated with infant formula. These findings provide an insight into a colostrum substitute mixture that resembles human colostrum in terms of composition and structural behavior during digestion and potentially reduces some of the characteristics associated with necrotizing enterocolitis.


Asunto(s)
Calostro , Enterocolitis Necrotizante , Animales , Embarazo , Femenino , Recién Nacido , Humanos , Animales Recién Nacidos , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/microbiología , Inflamación/prevención & control , Lípidos
2.
Anim Sci J ; 92(1): e13499, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33455052

RESUMEN

The objective of this study was to investigate the individual and combined effects of essential oils (EO; comprised of thymol and carvacrol) and lysozyme on experimental NE in broiler chickens. A total of 320 1-day-old chicks were randomly assigned to five treatment groups: no-challenge control (NC), NC + C. perfringens challenge (CC), CC + 120 mg/kg of EO, CC + 100 mg/kg of lysozyme, and CC + 120 mg/kg of EO + 100 mg/kg of lysozyme. The results showed that EO or lysozyme decreased the mortality, alleviated the gut lesions, inhibited the liver Enterobacteriaceae carriage, and increased the villus height of the ileum compared with CC (p < .05), although the proliferation of C. perfringens in the ileum was not inhibited (p > .05). Moreover, EO or lysozyme was found to decrease the ileal concentration of sialic acid and the Mucin2 mRNA expression (p < .05). However, the blend of EO and lysozyme did not display significant effect on the NE-associated mortality or gut damage in contrast with CC (p > .05). In conclusion, these findings suggest the similar protective effects of EO and lysozyme in NE-associated mortality and intestinal impairment, but their blend did not exhibit ameliorative effect.


Asunto(s)
Dieta/veterinaria , Suplementos Dietéticos , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/veterinaria , Muramidasa/administración & dosificación , Aceites Volátiles/administración & dosificación , Enfermedades de las Aves de Corral/tratamiento farmacológico , Animales , Pollos , Infecciones por Enterobacteriaceae , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Masculino , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/prevención & control
3.
Gastroenterology ; 159(6): 2130-2145.e5, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32805279

RESUMEN

BACKGROUND & AIMS: After birth, the immune system matures via interactions with microbes in the gut. The S100 calcium binding proteins S100A8 and S100A9, and their extracellular complex form, S100A8-A9, are found in high amounts in human breast milk. We studied levels of S100A8-A9 in fecal samples (also called fecal calprotectin) from newborns and during infancy, and their effects on development of the intestinal microbiota and mucosal immune system. METHODS: We collected stool samples (n = 517) from full-term (n = 72) and preterm infants (n = 49) at different timepoints over the first year of life (days 1, 3, 10, 30, 90, 180, and 360). We measured levels of S100A8-A9 by enzyme-linked immunosorbent assay and analyzed fecal microbiomes by 16S sRNA gene sequencing. We also obtained small and large intestine biopsies from 8 adults and 10 newborn infants without inflammatory bowel diseases (controls) and 8 infants with necrotizing enterocolitis and measured levels of S100A8 by immunofluorescence microscopy. Children were followed for 2.5 years and anthropometric data and medical information on infections were collected. We performed studies with newborn C57BL/6J wild-type and S100a9-/- mice (which also lack S100A8). Some mice were fed or given intraperitoneal injections of S100A8 or subcutaneous injections of Staphylococcus aureus. Blood and intestine, mesenterial and celiac lymph nodes were collected; cells and cytokines were measured by flow cytometry and studied in cell culture assays. Colon contents from mice were analyzed by culture-based microbiology assays. RESULTS: Loss of S100A8 and S100A9 in mice altered the phenotypes of colonic lamina propria macrophages, compared with wild-type mice. Intestinal tissues from neonatal S100-knockout mice had reduced levels of CX3CR1 protein, and Il10 and Tgfb1 mRNAs, compared with wild-type mice, and fewer T-regulatory cells. S100-knockout mice weighed 21% more than wild-type mice at age 8 weeks and a higher proportion developed fatal sepsis during the neonatal period. S100-knockout mice had alterations in their fecal microbiomes, with higher abundance of Enterobacteriaceae. Feeding mice S100 at birth prevented the expansion of Enterobacteriaceae, increased numbers of T-regulatory cells and levels of CX3CR1 protein and Il10 mRNA in intestine tissues, and reduced body weight and death from neonatal sepsis. Fecal samples from term infants, but not preterm infants, had significantly higher levels of S100A8-A9 during the first 3 months of life than fecal samples from adults; levels decreased to adult levels after weaning. Fecal samples from infants born by cesarean delivery had lower levels of S100A8-A9 than from infants born by vaginal delivery. S100 proteins were expressed by lamina propria macrophages in intestinal tissues from infants, at higher levels than in intestinal tissues from adults. High fecal levels of S100 proteins, from 30 days to 1 year of age, were associated with higher abundance of Actinobacteria and Bifidobacteriaceae, and lower abundance of Gammaproteobacteria-particularly opportunistic Enterobacteriaceae. A low level of S100 proteins in infants' fecal samples associated with development of sepsis and obesity by age 2 years. CONCLUSION: S100A8 and S100A9 regulate development of the intestinal microbiota and immune system in neonates. Nutritional supplementation with these proteins might aide in development of preterm infants and prevent microbiota-associated disorders in later years.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Adulto , Animales , Biopsia , Calgranulina A/administración & dosificación , Calgranulina A/análisis , Calgranulina B/análisis , Calgranulina B/genética , Preescolar , Colon/microbiología , Colon/patología , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Disbiosis/microbiología , Disbiosis/prevención & control , Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Heces/química , Heces/microbiología , Femenino , Estudios de Seguimiento , Microbioma Gastrointestinal/genética , Humanos , Inmunidad Mucosa , Lactante , Recién Nacido , Recien Nacido Prematuro/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Obesidad/epidemiología , Obesidad/inmunología , Obesidad/microbiología , Obesidad/prevención & control , ARN Ribosómico 16S/genética , Sepsis/epidemiología , Sepsis/inmunología , Sepsis/microbiología , Sepsis/prevención & control
4.
Nutrients ; 12(5)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380648

RESUMEN

Chorioamnionitis can lead to inflammation and injury of the liver and gut, thereby predisposing patients to adverse outcomes such as necrotizing enterocolitis (NEC). In addition, intestinal bile acids (BAs) accumulation is causally linked to NEC development. Plant sterols are a promising intervention to prevent NEC development, considering their anti-inflammatory properties in the liver. Therefore, we investigated whether an intra-amniotic (IA) Ureaplasma parvum (UP) infection affected the liver and enterohepatic circulation (EHC) and evaluated whether an IA administered plant sterol mixture dissolved in ß-cyclodextrin exerted prophylactic effects. An ovine chorioamnionitis model was used in which liver inflammation and the EHC were assessed following IA UP exposure in the presence or absence of IA prophylactic plant sterols (a mixture of ß-sitosterol and campesterol dissolved in ß-cyclodextrin (carrier)) or carrier alone. IA UP exposure caused an inflammatory reaction in the liver, histologically seen as clustered and conflated hepatic erythropoiesis in the parenchyma, which was partially prevented by IA administration of sterol + ß-cyclodextrin, or ß-cyclodextrin alone. In addition, IA administration of ß-cyclodextrin prior to UP caused changes in the expression of several hepatic BAs transporters, without causing alterations in other aspects of the EHC. Thereby, the addition of plant sterols to the carrier ß-cyclodextrin did not have additional effects.


Asunto(s)
Colesterol/análogos & derivados , Corioamnionitis/tratamiento farmacológico , Corioamnionitis/microbiología , Portadores de Fármacos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Circulación Enterohepática/efectos de los fármacos , Feto/irrigación sanguínea , Hígado/irrigación sanguínea , Fitosteroles/administración & dosificación , Fitoterapia , Profilaxis Posexposición/métodos , Sitoesteroles/administración & dosificación , Infecciones por Ureaplasma , Ureaplasma , beta-Ciclodextrinas , Animales , Colesterol/administración & dosificación , Colesterol/farmacología , Modelos Animales de Enfermedad , Femenino , Inflamación , Inyecciones Intralesiones , Fitosteroles/farmacología , Embarazo , Ovinos , Sitoesteroles/farmacología
5.
PLoS One ; 14(5): e0205784, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31150394

RESUMEN

Gut microbiota has been demonstrated to be involved in intestinal nutrition, defense, and immunity, as well as participating in disease progression. This study was to investigate gut microbiota changes in chickens challenged with netB-positive Clostridium perfringens strain (CP1) and/or the predisposing Eimeria species (Eimeria) and fed diets with fishmeal supplementation. In addition, the effects of lauric acid, a medium-chain fatty acid (MCFA), on necrotic enteritis (NE) reduction and modulation of microbiota were evaluated. The results demonstrated that microbial communities in the jejunum were distinct from those in the cecum, and the microbial community change was more significant in jejunum. Challenge of CP1 in conjunction with Eimeria significantly reduced species diversity in jejunal microbiota, but cecal microbiota remained stable. In the jejunum, CP1 challenge increased the abundance of the genera of Clostridium sensu stricto 1, Escherichia Shigella, and Weissella, but significantly decreased the population of Lactobacillus. Eimeria infection on its own was unable to promote NE, demonstrating decrements of Clostridium sensu stricto 1 and Lactobacillus. Co-infection with CP1 and Eimeria reproduced the majority of NE lesions with significant increment of Clostridium sensu stricto 1 and reduction in Lactobacillus. The advance of changes on these two taxa increased the severity of NE lesions. Further analyses of metagenomeSeq, STAMP, and LEfSe consistently showed significant overgrowth of Clostridium sensu stricto 1 was associated with NE. The supplementation of lauric acid did not reduce NE incidence and severity but decreased the relative abundance of Escherichia Shigella. In conclusion, significant overgrowth of C. perfringens as well as other Clostridium species in Clostridium sensu stricto 1 with the decrement of Lactobacillus in the jejunum is the featured microbiota correlated with NE. Controlling proliferation of Clostridium sensu stricto 1 and manipulation of Lactobacillus in the jejunum should be the strategy to prevent NE.


Asunto(s)
Pollos , Clostridium perfringens , Eimeria , Enterocolitis Necrotizante/veterinaria , Microbioma Gastrointestinal , Ácidos Láuricos/uso terapéutico , Enfermedades de las Aves de Corral/microbiología , Alimentación Animal , Animales , Infecciones por Clostridium/complicaciones , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/veterinaria , Coccidiosis/complicaciones , Coccidiosis/microbiología , Coccidiosis/veterinaria , Dieta/veterinaria , Suplementos Dietéticos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos Láuricos/farmacología , Masculino , Enfermedades de las Aves de Corral/parasitología , Enfermedades de las Aves de Corral/prevención & control
6.
Nutrients ; 11(6)2019 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-31195600

RESUMEN

Healthy microbiota is a critical mediator in maintaining health and it is supposed that dysbiosis could have a role in the pathogenesis of a number of diseases. Evidence supports the hypothesis that maternal dysbiosis could act as a trigger for preterm birth; aberrant colonization of preterm infant gut might have a role in feeding intolerance and pathogenesis of necrotizing enterocolitis. Despite several clinical trials and meta-analyses, it is still not clear if modulation of maternal and neonatal microbiota with probiotic supplementation decreases the risk of preterm birth and its complications.


Asunto(s)
Disbiosis/complicaciones , Enfermedades del Prematuro/microbiología , Complicaciones del Embarazo/microbiología , Nacimiento Prematuro/microbiología , Probióticos/uso terapéutico , Suplementos Dietéticos , Disbiosis/microbiología , Enterocolitis Necrotizante/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Recién Nacido , Recien Nacido Prematuro , Microbiota , Embarazo , Vagina/microbiología
7.
Curr Pediatr Rev ; 15(2): 92-105, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30868956

RESUMEN

Necrotizing enterocolitis (NEC) is an acquired severe disease of the digestive system affecting mostly premature babies, possibly fatal and frequently associated to systemic complications. Because of the severity of this condition and the possible long-term consequences on the child's development, many studies have aimed at preventing the occurrence of the primary events at the level of the bowel wall (ischemia and necrosis followed by sepsis) by modifying or manipulating the diet (breast milk versus formula) and/or the feeding pattern (time for initiation after birth, continuous versus bolus feeding, modulation of intake according clinical events). Feeding have been investigated so far in order to prevent NEC. However, currently well-established and shared clinical nutritional practices are not available in preventing NEC. Nutritional and surgical treatments of NEC are instead well defined. In selected cases surgery is a therapeutic option of NEC, requiring sometimes partial intestinal resection responsible for short bowel syndrome. In this paper we will investigate the available options for treating NEC according to the Walsh and Kliegman classification, focusing on feeding practices in managing short bowel syndrome that can complicate NEC. We will also analyze the proposed ways of preventing NEC.


Asunto(s)
Antibacterianos/uso terapéutico , Nutrición Enteral/métodos , Enterocolitis Necrotizante/terapia , Tracto Gastrointestinal/microbiología , Enfermedades del Prematuro/terapia , Suplementos Dietéticos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Humanos , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Resultado del Tratamiento
8.
Adv Exp Med Biol ; 1125: 37-45, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30656552

RESUMEN

Increasing evidence indicates that many of the health beneficial effects associated with the establishment of a symbiotic gut microbiota are driven by bacterial metabolic by-products.The term "postbiotics" indicates any soluble factor resulting from the metabolic activity of a live bacteria or any released molecule capable of providing health benefits through a direct or indirect mechanism.Alterations in preterm gut colonization associated with the intestinal barrier immaturity and the increased reactivity of the intestinal mucosa to colonizing bacteria have been implicated in the pathogenesis of necrotizing enterocolitis. Recent advances in the comprehension of the postbiotic biological effects and related mechanisms, some of them reviewed here, indicate that postbiotics may be a promising effective preventive strategy against necrotizing enterocolitis while avoiding the risk of administering live microorganisms to preterm infants that could translocate and cause infection. However, data from trials investigating the efficacy of postbiotics for the prevention of necrotizing enterocolitis in preterm infants are needed, and issues regarding their optimal regimen and start and duration of treatment need to be addressed.


Asunto(s)
Bacterias , Suplementos Dietéticos , Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal , Enfermedades del Recién Nacido/microbiología , Enterocolitis Necrotizante/prevención & control , Humanos , Recién Nacido , Enfermedades del Recién Nacido/prevención & control , Recien Nacido Prematuro , Intestinos/microbiología
9.
BMC Microbiol ; 18(1): 184, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424728

RESUMEN

BACKGROUND: The ProPrems trial, a multi-center, double-blind, placebo-controlled randomized trial, previously reported a 54% reduction in necrotizing enterocolitis (NEC) of Bell stage 2 or more from 4.4 to 2.0% in 1099 infants born before 32 completed weeks' gestation and weighing < 1500 g, receiving probiotic supplementation (with Bifidobacterium longum subsp. infantis BB-02, Streptococcus thermophilus TH-4 and Bifidobacterium animalis subsp. lactis BB-12). This sub-study investigated the effect of probiotic supplementation on the gut microbiota in a cohort of very preterm infants in ProPrems. RESULTS: Bifidobacterium was found in higher abundance in infants who received the probiotics (AOR 17.22; 95% CI, 3.49-84.99, p < 0.001) as compared to the placebo group, and Enterococcus was reduced in infants receiving the probiotic during the supplementation period (AOR 0.27; 95% CI, 0.09-0.82, p = 0.02). CONCLUSION: Probiotic supplementation with BB-02, TH-4 and BB-12 from soon after birth increased the abundance of Bifidobacterium in the gut microbiota of very preterm infants. Increased abundance of Bifidobacterium soon after birth may be associated with reducing the risk of NEC in very preterm infants.


Asunto(s)
Suplementos Dietéticos/análisis , Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal , Recien Nacido Extremadamente Prematuro/crecimiento & desarrollo , Probióticos/administración & dosificación , Bifidobacterium/genética , Bifidobacterium/aislamiento & purificación , Bifidobacterium/efectos de la radiación , Estudios de Cohortes , Método Doble Ciego , Enterocolitis Necrotizante/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Streptococcus thermophilus/genética , Streptococcus thermophilus/aislamiento & purificación , Streptococcus thermophilus/fisiología
11.
Dan Med J ; 64(6)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28566122

RESUMEN

The premature infant suffers from immaturity of all organ systems, one of them being the gastrointestinal tract. When the infant is born, the immature gastrointestinal tract is exposed to milk and simultaneously colonized by high densities of bacteria. The combination of milk, microbiota and an immature gut, leaves the infant vulnerable to developing the dreaded intestinal emergency necrotizing enterocolitis (NEC). NEC is often very aggressive and no cure exists, which means that prevention is an utmost important topic to researchers, physicians, parents - and infants.   Despite immense research during the last decades, no specific test to determine if an infant suffers from NEC exists. Most neonatal units use Bell's staging criteria, which are based on clinical and radiographic findings, as a diagnostic tool; however the diagnosis given according to Bell's stages has not been validated. In study I, we aimed to determine the validity of the NEC diagnosis given at discharge. An expert panel consisting of a neonatologist, a paediatric surgeon and a paediatric radiologist served as the golden standard. We found that the diagnosis given at discharge had a poor validity which significantly affected the reported incidence of NEC in the neonatal department at Rigshospitalet, Denmark. The validity of the NEC diagnosis was worse than the validity of most other paediatric diagnoses that had been investigated.   In studies II and III, we aimed to explore possible means of NEC prevention. The role of nutrition in NEC development is well established with mother's milk as the best option to avoid NEC in the preterm infant. Maternal milk is, however, most often not available in sufficient amounts during the first days of life, and preterm infant formula or human donor milk is used in its absence. Studies in preterm piglets showed that bovine colostrum equally to human donor milk protected against NEC compared to infant formula. Furthermore, bovine colostrum was superior to human donor milk in stimulating gut immunity and digestive functions.   Hence, in study II we aimed to design a pilot study of bovine colostrum used as a supplement to maternal milk in the first days of life and to determine if the study was feasible. In the paper, we present the protocol and the results of the first two phases of the Precolos study in which 12 infants were included and received pasteurized, spray-dried and reconstituted bovine colostrum during the first days of life as the first infants in the world. We found that the infants tolerated bovine colostrum without clinical adverse effects, but we also observed a transient hypertyrosinemia on day seven of life in five infants. The results were evaluated by a safety management board which encouraged us to continue the pilot study with the last phase, which was a randomized controlled trial of 20+20 infants comparing supplementation with bovine colostrum to supplementation with standard nutrition. The randomized trial has just finished recruitment.   At last, we wanted to shed light on a possible microbiological angle of NEC prevention. Dysbiosis and bacterial translocation are believed to play a crucial role in the development of NEC as intestinal pneumatosis, which occurs when bacteria produce gas inside the intestinal wall, is a pathognomonic radiographic finding. In a quality improvement study from the US published in 2014, NEC incidence was significantly reduced after the implementation of several quality improvement interventions. Standardized weekly exchange of nasogastric feeding tubes was suggested as one of the potential NEC-reducing interventions.   In the neonatal unit at Rigshospitalet, Denmark, preterm infants are fed 8-12 times daily through a resident nasogastric feeding tube which is exposed to body temperature, contains milk residuals from the last meal and is handled by both parents and personnel. Since bacterial pollution of milk given through the nasogastric feeding tube might be NEC-inducing, we aimed in study III to determine the bacterial load given to the infants when feeding them through a tube. We collected 92 used nasogastric feeding tubes and flushed them with one ml saline each to imitate a meal given through them. Eighty-nine percent of the tubes contaminated the meals with more than 1000 colony-forming units of bacteria and fifty-five percent contaminated the meals with the possible pathogens Enterobacteriaceae or Staphylococcus aureus. The concentration of bacteria in the saline flushed through the tubes was as high as 109 colony-forming units per ml; however, neither the risk of contamination nor the concentration of bacteria in the flush was associated with the duration of use. Implementation of standardized weekly exchange of feeding tubes would therefore not prevent the contamination of meals.   In conclusion, the studies included in this thesis serve as a base for future studies investigating the prevention of NEC. We found a poor validity of the NEC diagnosis given at discharge. This should be kept in mind when conducting epidemiological studies of NEC and especially when conducting interventional trials with NEC as an outcome. If the findings of the randomized part of the Precolos study indicate a positive effect of bovine colostrum and do not give rise to concerns regarding feasibility, safety or tolerability, a large-scale randomized controlled study with NEC as the primary outcome will be planned. Based on the high concentrations of bacteria found in the nasogastric feeding tubes, a randomized controlled trial investigating whether the frequency of feeding tube exchange affects the early colonization has been commenced in the neonatal department at Rigshospitalet. Hopefully, the results of these studies will bring us closer to preventing NEC in the future.


Asunto(s)
Calostro , Suplementos Dietéticos , Enterocolitis Necrotizante/prevención & control , Enfermedades del Prematuro/prevención & control , Animales , Bovinos , Dinamarca , Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/microbiología , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/microbiología , Intubación Gastrointestinal/efectos adversos , Masculino , Proyectos Piloto
12.
Pediatr Res ; 82(4): 584-595, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28609432

RESUMEN

Necrotizing enterocolitis (NEC) is the most common life threatening condition affecting preterm infants. NEC occurs in 1-5% of all neonatal intensive care admissions and 5-10% of very low birth weight infants. The protective role of human breast milk (BM) has been well established. It has also been shown that amniotic fluid (AF) and BM have many similarities in terms of presence of growth and other immune-modulatory factors. This finding led to the initial hypothesis that AF may exert similar protective effects against the development of NEC, as does BM. Multiple studies have elucidated the presence of growth factors in AF and the protective effect of AF against NEC. Studies have also described possible mechanisms how AF protects against NEC. At present, research in this particular area is extremely active and robust. This review summarizes the various studies looking at the protective effects of AF against the development of NEC. It also provides an insight into future directions, the vast potential of AF as a readily available biologic medium, and the ethical barriers that must be overcome before using AF.


Asunto(s)
Líquido Amniótico/metabolismo , Terapia Biológica/métodos , Enterocolitis Necrotizante/prevención & control , Mucosa Intestinal/metabolismo , Líquido Amniótico/citología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Terapia Biológica/efectos adversos , Terapia Biológica/ética , Lactancia Materna , Citocinas/metabolismo , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal , Edad Gestacional , Humanos , Recién Nacido , Recien Nacido Prematuro , Intestinos/microbiología , Leche Humana/metabolismo , Factores Protectores , Factores de Riesgo , Transducción de Señal , Trasplante de Células Madre , Células Madre/metabolismo , Receptor Toll-Like 4/metabolismo
13.
Future Microbiol ; 11(2): 273-92, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26855351

RESUMEN

Necrotizing enterocolitis (NEC) remains a devastating intestinal disease in preterm neonates. In this population, disruption of the gut microbiota development, mainly due to organ immaturity, antibiotic use and hospital microbial environment, plays a key role in the pathogenesis of NEC. This gut dysbiosis has been associated with opportunistic pathogens overgrowth, expression of virulence factors, altered metabolic functions and inflammatory dysregulated responses. In this review, we provide an updated summary of the host and gut microbiota interactions during the formative early life. We also explore the key determinants of gut dysbiosis in preterm neonates with NEC. Finally, we discuss the promising role of bacteriotherapy in the management of NEC, the aim being to shape or restore the beneficial gut bacterial communities.


Asunto(s)
Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Enfermedades del Prematuro/microbiología , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Disbiosis , Enterocolitis Necrotizante/fisiopatología , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/terapia , Microbioma Gastrointestinal/fisiología , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/fisiopatología , Enfermedades del Prematuro/prevención & control , Enfermedades del Prematuro/terapia , Naturopatía , Probióticos/uso terapéutico , Simbiosis
14.
J Pediatr Gastroenterol Nutr ; 63(2): 280-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26756878

RESUMEN

OBJECTIVE: Feeding bovine colostrum (BC) improves gut maturation and function and protects against necrotizing enterocolitis, relative to formula in newborn preterm pigs. Before BC can be used for preterm infants, it is important to test if the milk processing, required to reduce bacterial load and increase shelf life, may affect bioactivity and efficacy of a BC product. METHODS: We investigated if spray dried, pasteurised BC had protective effects on gut function in preterm pigs, relative to formula. After a 2-day total parenteral nutrition period, preterm pigs were fed formula for a few hours (to induce a proinflammatory state) followed by 2 days of formula (FORM, n = 14), BC (colostrum [COLOS], n = 14), spray-dried BC (POW, n = 8), or pasteurised, spray-dried BC (POWPAS, n = 9). RESULTS: Spray drying and pasteurisation of BC decreased the concentration of transforming growth factor-ß1, -ß2 and increased protein aggregation. All of the 3 BC groups had reduced necrotizing enterocolitis severity, small intestinal levels of IL-1ß, -8, and colonic lactic acid levels, and increased intestinal villus height, hexose absorption, and digestive enzyme activities, relative to the FORM group (all P < 0.05). All of the 3 BC diets stimulated epithelial cell migration in a wound-healing model with IEC-6 cells. CONCLUSIONS: Spray drying and pasteurisation affect BC proteins, but do not reduce the trophic and anti-inflammatory effects of BC on the immature intestine. It remains to be studied if BC products will benefit preterm infants just after birth when human milk is often not available.


Asunto(s)
Calostro , Enterocolitis Necrotizante/prevención & control , Inflamación/prevención & control , Pasteurización , Conservación de Tejido/métodos , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Bovinos , Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/microbiología , Inflamación/diagnóstico , Inflamación/metabolismo , Inflamación/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Permeabilidad , Porcinos , Resultado del Tratamiento
15.
J Perinat Neonatal Nurs ; 29(3): 240-7; quiz E2, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26218817

RESUMEN

Necrotizing enterocolitis remains a significant cause of morbidity and mortality in very low-birth-weight infants (<1500 g), with current preventive strategies unclear. Scientific evidence has recently emerged, suggesting that probiotics, prebiotics, and synbiotics may effectively and safely alter the premature intestinal microbiota, enhancing a deficient innate immune response and maturing the intestinal barrier to prevent necrotizing enterocolitis development. Currently, formal recommendations do not support routine use of these dietary supplementations for premature infants. Here, we examine how probiotic, prebiotic, and synbiotic preparations physiologically alter the underdeveloped intestinal microbial environment to potentially reduce necrotizing enterocolitis incidence and discuss current evidence that has examined safety and efficacy factors potentially supporting routine use among the premature infant population.


Asunto(s)
Terapia Biológica , Enterocolitis Necrotizante , Recien Nacido Prematuro/fisiología , Prebióticos , Probióticos , Simbióticos , Terapia Biológica/efectos adversos , Terapia Biológica/métodos , Suplementos Dietéticos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal/fisiología , Humanos , Recién Nacido , Recién Nacido de muy Bajo Peso/fisiología , Resultado del Tratamiento
16.
Neonatal Netw ; 34(2): 83-101, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26803090

RESUMEN

Common among preterm, very low birth weight (VLBW) and extremely low birth weight (ELBW) infants, necrotizing enterocolitis (NEC) is a gastrointestinal, infectious disease that remains a leading cause of morbidity and mortality among this high-risk population. To combat this devastating condition, research efforts have been redirected from treatment toward prevention strategies. Although there are several proposed risk-reduction strategies, one intervention gaining support is the administration of prophylactic enteral probiotics. Regardless of growing evidentiary support and a benign safety profile, neonatal providers have yet to embrace this therapy. This article provides an overview of the proposed benefits of probiotics, focusing on their role as a NEC prevention strategy. A review of several sentinel research studies targeting preterm, VLBW, and ELBW infants is provided. Considerations for ongoing research are reviewed. Finally, two evidence-based NEC prevention probiotics protocols are presented.


Asunto(s)
Enterocolitis Necrotizante , Probióticos/uso terapéutico , Suplementos Dietéticos , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/fisiopatología , Enterocolitis Necrotizante/prevención & control , Humanos , Recien Nacido con Peso al Nacer Extremadamente Bajo , Recién Nacido , Recien Nacido Prematuro , Resultado del Tratamiento
17.
Am J Physiol Gastrointest Liver Physiol ; 307(7): G689-99, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25147235

RESUMEN

A balance between pro- and anti-inflammatory signals from milk and microbiota controls intestinal homeostasis just after birth, and an optimal balance is particularly important for preterm neonates that are sensitive to necrotizing enterocolitis (NEC). We suggest that the intestinal cytokine IL-8 plays an important role and hypothesize that transforming growth factor-ß2 (TGF-ß2) acts in synergy with bacterial lipopolysaccharide (LPS) to control IL-8 levels, thereby supporting intestinal homeostasis. Preterm pigs were fed colostrum (containing TGF-ß2) or infant formula (IF) with or without antibiotics (COLOS, n = 27; ANTI, n = 11; IF, n = 40). Intestinal IL-8 levels and NEC incidence were much higher in IF than in COLOS and ANTI pigs (P < 0.001), but IL-8 levels did not correlate with NEC severity. Intestinal TGF-ß2 levels were high in COLOS but low in IF and ANTI pigs. Based on these observations, the interplay among IL-8, TGF-ß2, and LPS was investigated in a porcine intestinal epithelial cell line. TGF-ß2 attenuated LPS-induced IL-6, IL-1ß, and TNF-α release by reducing early ERK activation, whereas IL-8 secretion was synergistically induced by LPS and TGF-ß2 via NF-κB. The TGF-ß2/LPS-induced IL-8 levels stimulated cell proliferation and migration following epithelial injury, without continuous NF-κB activation and cyclooxygenase-2 expression. We suggest that a combined TGF-ß2-LPS induction of IL-8 stimulates epithelial repair just after birth when the intestine is first exposed to colonizing bacteria and TGF-ß2-containing milk. Moderate IL-8 levels may act to control intestinal inflammation, whereas excessive IL-8 production may enhance the damaging proinflammatory cascade leading to NEC.


Asunto(s)
Enterocolitis Necrotizante/metabolismo , Interleucina-8/metabolismo , Intestino Delgado/metabolismo , Lipopolisacáridos/farmacología , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Antibacterianos/farmacología , Línea Celular , Movimiento Celular , Proliferación Celular , Calostro , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Edad Gestacional , Homeostasis , Humanos , Fórmulas Infantiles , Recién Nacido , Interleucina-8/farmacología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Intestino Delgado/patología , FN-kappa B/metabolismo , Nacimiento Prematuro , Transducción de Señal/efectos de los fármacos , Porcinos , Factores de Tiempo , Factor de Crecimiento Transformador beta2/farmacología
18.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G864-75, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23518680

RESUMEN

Preterm neonates are susceptible to gastrointestinal disorders such as necrotizing enterocolitis (NEC). Maternal milk and colostrum protects against NEC via growth promoting, immunomodulatory, and antimicrobial factors. The fetal enteral diet amniotic fluid (AF), contains similar components, and we hypothesized that postnatal AF administration reduces inflammatory responses and NEC in preterm neonates. Preterm pigs (92% gestation) were delivered by caesarean section and fed parental nutrition (2 days) followed by enteral (2 days) porcine colostrum (COLOS, n = 7), infant formula (FORM, n = 13), or AF supplied before and after introduction of formula (AF, n = 10) in experiment 1, and supplied only during the enteral feeding period in experiment 2 (FORM, n = 16; AF, n = 14). The NEC score was reduced in both AF and COLOS pigs, relative to FORM, when AF was provided prior to full enteral feeding (9.9 and 7.7 compared with 17.3, P < 0.05). There was no effect of AF when provided only during enteral feeding. AF pigs showed decreased bacterial abundance in colon and intestinal inflammation-related genes (e.g., TNF-α, IL-1α, IL-6, NOS) were downregulated, relative to FORM pigs with NEC. Anti-inflammatory properties of AF were supported by delayed maturation and decreased TNF-α production in murine dendritic cells, as well as increased proliferation and migration, and downregulation of IL-6 expression in intestinal cells (IEC-6, IPEC-J2). Like colostrum, AF may reduce NEC development in preterm neonates by suppressing the proinflammatory responses to enteral formula feeding and gut colonization when provided before the onset of NEC.


Asunto(s)
Líquido Amniótico/fisiología , Calostro/fisiología , Enterocolitis Necrotizante/terapia , Gastroenteritis/terapia , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Células Dendríticas/metabolismo , Nutrición Enteral , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Enterocitos/metabolismo , Femenino , Gastroenteritis/microbiología , Gastroenteritis/patología , Humanos , Recién Nacido , Recien Nacido Prematuro , Absorción Intestinal , Intestinos/microbiología , Análisis por Micromatrices , Nutrición Parenteral Total , Permeabilidad , Embarazo , ARN/biosíntesis , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos
19.
JPEN J Parenter Enteral Nutr ; 35(1): 32-42, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224432

RESUMEN

BACKGROUND: A rapid advance in enteral feeding is associated with necrotizing enterocolitis (NEC) in preterm infants. Therefore, minimal enteral nutrition (MEN) combined with parenteral nutrition (PN) is common clinical practice, but the effects on NEC and intestinal function remain poorly characterized. It was hypothesized that a commonly used MEN feeding volume (16-24 mL/kg/d) prevents NEC and improves intestinal structure, function, and microbiology in preterm pigs. METHODS: After preterm birth pigs were stratified into 4 nutrition intervention groups that received the following treatments: (1) PN followed by full enteral formula feeding (OF group, n = 12); (2) PN supplemented with formula MEN and followed by full formula feeding (FF, n = 12); (3) PN plus colostrum MEN followed by formula feeding (CF, n = 12); (4) PN plus colostrum MEN followed by colostrum feeding (CC, n = 10). RESULTS: NEC was absent in the CC group but frequent in the other groups (50%-67%). Compared with other groups, CC pigs showed improved mucosal structures, brush border enzyme activities, and hexose absorption (all P < .05). Relative to formula MEN, colostrum MEN thus improved gut function but did not prevent later formula-induced gut dysfunction and NEC. However, in CF pigs, intestinal lesions were restricted to the colon, compared with all regions in OF and FF pigs, which indicated proximal protection of colostrum MEN. Bacterial composition was not affected by MEN, diet, or NEC outcomes, but bacterial load and concentrations of short-chain fatty acids were reduced in the MEN groups. CONCLUSION: Colostrum MEN improves intestinal structure, function, and NEC resistance in preterm pigs but does not protect against gut dysfunction and NEC associated with later full enteral formula feeding.


Asunto(s)
Animales Recién Nacidos , Dieta , Nutrición Enteral , Enterocolitis Necrotizante/dietoterapia , Nacimiento Prematuro , Animales , Carga Bacteriana , Calostro , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Femenino , Hexosas/metabolismo , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/fisiología , Modelos Animales , Embarazo , Porcinos
20.
J Pediatr Gastroenterol Nutr ; 51(2): 203-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20543719

RESUMEN

OBJECTIVE: To systematically evaluate and update evidence on the efficacy and safety of Bifidobacterium animalis subsp lactis CNCM I-3446 supplementation in preterm infants. MATERIALS AND METHODS: The Cochrane Library and MEDLINE databases and major pediatric conference proceedings were searched in December 2008 for randomized controlled trials (RCTs). The company that manufactures B lactis was contacted for unpublished data. The review was restricted to RCTs performed in preterm infants <37 weeks of gestation and/or with a birth weight <2500 g. RESULTS: Four RCTs involving 324 infants met the inclusion criteria. Compared with controls, B lactis supplementation has the potential to increase fecal bifidobacteria counts and to reduce Enterobacteriaceae and Clostridium spp counts. It also can reduce stool pH and fecal calprotectin concentrations, increase fecal immunoglobulin A and short-chain fatty acid concentrations, and decrease intestinal permeability. Compared with controls, B lactis supplementation had no effect on the risk of necrotizing enterocolitis stage > or = 2 (3 RCTs, n = 293, risk ratio [RR] 0.53, 95% CI 0.16-1.83), risk of sepsis (2 RCTs, 397 cultures, RR 0.6, 95% CI 0.07-5.2), and use of antibiotics (2 RCTs, n = 255, RR 0.67, 95% CI 0.28-1.62). The power of these studies, however, does not allow for a definitive statement regarding a reduced risk of necrotizing enterocolitis. B lactis supplementation did have some effects on anthropometric parameters. No adverse events associated with B lactis supplementation were reported. CONCLUSIONS: Evidence regarding the potential beneficial effects of B lactis supplementation in preterm infants is encouraging. Further studies to assess clinically relevant outcomes are needed.


Asunto(s)
Antibacterianos/farmacología , Bifidobacterium , Productos Biológicos/farmacología , Suplementos Dietéticos , Enfermedades del Prematuro/prevención & control , Probióticos/uso terapéutico , Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Productos Biológicos/uso terapéutico , Colon/efectos de los fármacos , Colon/microbiología , Colon/fisiología , Recuento de Colonia Microbiana , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Heces/microbiología , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA