Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34143954

RESUMEN

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Asunto(s)
Bifidobacterium/fisiología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología , Antibacterianos/farmacología , Biomarcadores/metabolismo , Lactancia Materna , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Proliferación Celular , Citocinas/metabolismo , Heces/química , Heces/microbiología , Galectina 1/metabolismo , Microbioma Gastrointestinal , Humanos , Indoles/metabolismo , Recién Nacido , Inflamación/sangre , Inflamación/genética , Mucosa Intestinal/inmunología , Metaboloma , Leche Humana/química , Oligosacáridos/metabolismo , Células Th17/inmunología , Células Th2/inmunología , Agua
2.
Am J Perinatol ; 40(3): 260-266, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-32693415

RESUMEN

The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic has impacted all patient populations including pregnant mothers. There is an incomplete understanding of SARS-CoV-2 pathogenesis and transmission potential at this time and the resultant anxiety has led to variable breastfeeding recommendations for suspected or confirmed mothers with novel coronavirus disease 2019 (COVID-19). Due to the potential concern for transmission of infection from maternal respiratory secretions to the newborn, temporary separation of the maternal-baby dyad, allowing for expressed breast milk to be fed to the infant, was initially recommended but later revised to include breastfeeding by the American Academy of Pediatrics in contrast to international societies, which recommend direct breastfeeding. This separation can have negative health and emotional implications for both mother and baby. Only two publications have reported SARS-CoV-2 in human breast milk but the role of breast milk as a vehicle of transmission of COVID-19 to the newborns still remains unclear and may indeed be providing protective antibodies against SARS-CoV-2 infection even in infected neonates. Other modes of transmission of infection to neonates from infected mothers or any care providers cannot be overemphasized. Symptomatic mothers on hydroxychloroquine can safely breastfeed and no adverse effects were reported in a baby treated with remdesivir in another drug trial. The excretion of sarilumab in human breast milk is unknown at this time. Hence, given the overall safety of breast milk and both short-term and long-term nutritional, immunological, and developmental advantages of breast milk to newborn, breast milk should not be withheld from baby. The setting of maternal care, severity of maternal infection and availability of resources can impact the decision of breastfeeding, the role of shared decision making on breastfeeding between mother and physician needs to be emphasized. We strongly recommend direct breastfeeding with appropriate hygiene precautions unless the maternal or neonatal health condition warrants separation of this dyad. KEY POINTS: · Breastmilk does not appear to play a significant role in transmission of SARS-CoV-2.. · Mother-baby separation has negative health and emotional consequences.. · Mothers with suspected or confirmed COVID-19 can directly breastfeed with appropriate precautions..


Asunto(s)
COVID-19 , Complicaciones Infecciosas del Embarazo , Lactante , Femenino , Embarazo , Recién Nacido , Humanos , Niño , Lactancia Materna , SARS-CoV-2 , Pandemias/prevención & control , Leche Humana , Madres/psicología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/epidemiología
3.
J Nutr ; 152(5): 1239-1253, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35179194

RESUMEN

BACKGROUND: Human milk oligosaccharides (HMOs) are an abundant class of compounds found in human milk and have been linked to the development of the infant, and specifically the brain, immune system, and gut microbiome. OBJECTIVES: Advanced analytical methods were used to obtain relative quantitation of many structures in approximately 2000 samples from over 1000 mothers in urban, semirural, and rural sites across geographically diverse countries. METHODS: LC-MS-based analytical methods were used to profile the compounds with broad structural coverage and quantitative information. The profiles revealed their structural heterogeneity and their potential biological roles. Comparisons of HMO compositions were made between mothers of different age groups, lactation periods, infant sexes, and residing geographical locations. RESULTS: A common behavior found among all sites was a decrease in HMO abundances during lactation until approximately postnatal month 6, where they remained relatively constant. The greatest variations in structural abundances were associated with the presence of α(1,2)-fucosylated species. Genomic analyses of the mothers were not performed; instead, milk was phenotyped according to the abundances of α(1,2)-fucosylated structures. Mothers from the South American sites tended to have higher proportions of phenotypic secretors [mothers with relatively high concentrations of α(1,2)-fucosylated structures] in their populations compared to the rest of the globe, with Bolivia at ∼100% secretors, Peru at ∼97%, Brazil at ∼90%, and Argentina at ∼85%. Conversely, the cohort sampled in Africa manifested the lowest proportion of secretors (South Africa ∼ 63%, the Gambia ∼ 64%, and Malawi ∼ 75%). Furthermore, we compared total abundances of HMOs in secretors compared with nonsecretors and found that nonsecretors have lower abundances of HMOs compared to secretors, regardless of geographical location. We also observed compositional differences of the 50+ most abundant HMOs between milk types and geographical locations. CONCLUSIONS: This study represents the largest structural HMO study to date and reveals the general behavior of HMOs during lactation among different populations.


Asunto(s)
Leche Humana , Oligosacáridos , Lactancia Materna , Femenino , Humanos , Lactante , Lactancia , Malaui , Leche Humana/química , Oligosacáridos/química
4.
Pediatr Res ; 91(3): 627-636, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33762689

RESUMEN

BACKGROUND: Recent studies have reported a dysfunctional gut microbiome in breastfed infants. Probiotics have been used in an attempt to restore the gut microbiome; however, colonization has been transient, inconsistent among individuals, or has not positively impacted the host's gut. METHODS: This is a 2-year follow-up study to a randomized controlled trial wherein 7-day-old infants received 1.8 × 1010 colony-forming unit Bifidobacterium longum subsp. infantis (B. infantis) EVC001 (EVC) daily for 21 days or breast milk alone (unsupplemented (UNS)). In the follow-up study, mothers (n = 48) collected infant stool at 4, 6, 8, 10, and 12 months postnatal and completed the health-diet questionnaires. RESULTS: Fecal B. infantis was 2.5-3.5 log units higher at 6-12 months in the EVC group compared with the UNS group (P < 0.01) and this relationship strengthened with the exclusion of infants who consumed infant formula and antibiotics. Infants in the EVC group had significantly higher Bifidobacteriaceae and lower Bacteroidaceae and Lachnospiraceae (P < 0.05). There were no differences in any health conditions between the two groups. CONCLUSIONS: Probiotic supplementation with B. infantis within the first month postnatal, in combination with breast milk, resulted in stable colonization that persisted until at least 1 year postnatal. IMPACT: A dysfunctional gut microbiome in breastfed infants is common in resource-rich nations and associated with an increased risk of immune diseases. Probiotics only transiently exist in the gut without persistent colonization or altering the gut microbiome. This is the first study to show that early probiotic supplementation with B. infantis with breast milk results in stable colonization of B. infantis and improvements to the gut microbiome 1 year postnatal. This study addresses a key gap in the literature whereby probiotics can restore the gut microbiome if biologically selected microorganisms are matched with their specific food in an open ecological niche.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Bifidobacterium longum subspecies infantis , Lactancia Materna , Heces/microbiología , Femenino , Estudios de Seguimiento , Humanos , Lactante , Leche Humana
5.
J Pediatr Gastroenterol Nutr ; 75(4): 535-542, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35881967

RESUMEN

OBJECTIVES: To compare the impact of two probiotic supplements on fecal microbiota and metabolites, as well as on gut inflammation in human milk-fed preterm infants. METHODS: In this single-center observational cohort study, we assessed the effects of Bifidobacterium longum subsp. infantis or Lactobacillus reuteri supplementation on the infant gut microbiota by 16S rRNA gene sequencing and fecal metabolome by 1 H nuclear magnetic resonance spectroscopy. Fecal calprotectin was measured as a marker of enteric inflammation. Aliquots of human or donor milk provided to each infant were also assessed to determine human milk oligosaccharide (HMO) content. RESULTS: As expected, each probiotic treatment was associated with increased proportions of the respective bacterial taxon. Fecal HMOs were significantly higher in L. reuteri fed babies despite similar HMO content in the milk consumed. Fecal metabolites associated with bifidobacteria fermentation products were significantly increased in B. infantis supplemented infants. Fecal calprotectin was lower in infants receiving B. infantis relative to L. reuteri ( P < 0.01, Wilcoxon rank-sum test) and was negatively associated with the microbial metabolite indole-3-lactate (ILA). CONCLUSIONS: This study demonstrates that supplementing an HMO-catabolizing Bifidobacterium probiotic results in increased microbial metabolism of milk oligosaccharides and reduced intestinal inflammation relative to a noncatabolizing Lactobacillus probiotic in human milk-fed preterm infants. In this context, Bifidobacterium may provide greater benefit in human milk-fed infants via activation of the microbiota-metabolite-immune axis.


Asunto(s)
Microbiota , Probióticos , Bifidobacterium , Bifidobacterium longum subspecies infantis/metabolismo , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Inflamación , Complejo de Antígeno L1 de Leucocito/metabolismo , Oligosacáridos/metabolismo , ARN Ribosómico 16S
6.
Prostate ; 80(14): 1188-1202, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33258506

RESUMEN

BACKGROUND: As the survival of castration-resistant prostate cancer (CRPC) remains poor, and the nuclear factor-κB (NF-κB) pathways play key roles in prostate cancer (PC) progression, several studies have focused on inhibiting the NF-κB pathway through generating inhibitory κB kinase subunit α (IKKα) small molecule inhibitors. However, the identification of prognostic markers able to discriminate which patients could benefit from IKKα inhibitors is urgently required. The present study investigated the prognostic value of IKKα, IKKα phosphorylated at serine 180 (p-IKKα S180) and threonine 23 (p-IKKα T23), and their relationship with the androgen receptor (AR) and Ki67 proliferation index to predict patient outcome. METHODS: A cohort of 115 patients with hormone-naïve PC (HNPC) and CRPC specimens available were used to assess tumor cell expression of proteins within both the cytoplasm and the nucleus by immunohistochemistry. The expression levels were dichotomized (low vs high) to determine the associations between IKKα, AR, Ki67, and patients'Isurvival. In addition, an analysis was performed to assess potential IKKα associations with clinicopathological and inflammatory features, and potential IKKα correlations with other cancer pathways essential for CRPC growth. RESULTS: High levels of cytoplasmic IKKα were associated with a higher cancer-specific survival in HNPC patients with low AR expression (hazards ratio [HR], 0.33; 95% confidence interval [CI] log-rank, 0.11-0.98; P = .04). Furthermore, nuclear IKKα (HR, 2.60; 95% CI, 1.27-5.33; P = .01) and cytoplasmic p-IKKα S180 (HR, 2.10; 95% CI, 1.17-3.76; P = .01) were associated with a lower time to death from recurrence in patients with CRPC. In addition, high IKKα expression was associated with high levels of T-cells (CD3+ P = .01 and CD8+ P = .03) in HNPC; however, under castration conditions, high IKKα expression was associated with high levels of CD68+ macrophages (P = .04), higher Gleason score (P = .01) and more prostate-specific antigen concentration (P = .03). Finally, we identified crosstalk between IKKα and members of the canonical NF-κB pathway in the nucleus of HNPC. Otherwise, IKKα phosphorylated by noncanonical NF-κB and Akt pathways correlated with members of the canonical NF-κB pathway in CRPC. CONCLUSION: The present study reports that patients with CRPC expressing high levels of nuclear IKKα or cytoplasmic p-IKKα S180, which associated with a lower time to death from recurrence, may benefit from IKKα inhibitors.


Asunto(s)
Quinasa I-kappa B/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata/enzimología , Anciano , Biomarcadores de Tumor/metabolismo , Núcleo Celular/enzimología , Estudios de Cohortes , Citoplasma/enzimología , Humanos , Quinasa I-kappa B/inmunología , Inmunidad Innata , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Pronóstico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Tasa de Supervivencia
7.
Pediatr Res ; 88(Suppl 1): 48-55, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855513

RESUMEN

In spite of a large number of randomized placebo-controlled clinical trials and observational cohort studies including >50,000 preterm infants from 29 countries that have demonstrated a decrease in the risk of necrotizing enterocolitis, death, and sepsis, routine prophylactic probiotic administration to preterm infants remains uncommon in much of the world. This manuscript reflects talks given at NEC Society Symposium in 2019 and is not intended to be a state-of-the-art review or systematic review, but a summary of the probiotic-specific aspects of the symposium with limited additions including a recent strain-specific network analysis and position statement from the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN). We address ten common questions related to the intestinal microbiome and probiotic administration to the preterm infant.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/terapia , Probióticos/uso terapéutico , Ensayos Clínicos como Asunto , Disbiosis/fisiopatología , Europa (Continente) , Microbioma Gastrointestinal , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido , Recien Nacido Prematuro , Leche Humana , Estudios Observacionales como Asunto , Seguridad del Paciente , Sepsis/prevención & control , Sociedades Médicas , Resultado del Tratamiento
8.
Pediatr Res ; 88(Suppl 1): 41-47, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855512

RESUMEN

Although risk for necrotizing enterocolitis (NEC) is often presented from the perspective of a premature infant's vulnerability to nonmodifiable risk factors, in this paper we describe the evidence and present recommendations to manage modifiable risks that are amenable to clinical actions. Using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) criteria, we present recommendations in the context of their supporting evidence in a way that balances risks (e.g. potential harm, cost) and benefits. Across the prenatal, intrapartum, early and late clinical course, strategies to limit NEC risk in premature infants are presented. Our goal is to summarize modifiable NEC risk factors, grade the evidence to offer quality improvement (QI) targets for healthcare teams and offer a patient-family advocate's perspective on how to engage parents to recognize and reduce NEC risk.


Asunto(s)
Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/prevención & control , Cuidado Intensivo Neonatal/métodos , Índice de Severidad de la Enfermedad , Corticoesteroides/uso terapéutico , Anemia/complicaciones , Antibacterianos/uso terapéutico , Conducto Arterioso Permeable/complicaciones , Enterocolitis Necrotizante/etiología , Femenino , Humanos , Indometacina/uso terapéutico , Lactante , Recién Nacido , Unidades de Cuidado Intensivo Neonatal , Lactancia , Leche Humana , Probióticos , Mejoramiento de la Calidad , Riesgo , Factores de Riesgo , Cordón Umbilical , Estados Unidos
9.
Pediatr Res ; 87(3): 472-479, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31537010

RESUMEN

BACKGROUND: Postnatal growth restriction (PNGR) in premature infants increases risk of pulmonary hypertension (PH). In a rodent model, PNGR causes PH, while combining PNGR and hyperoxia increases PH severity. We hypothesized that PNGR causes intestinal dysbiosis and that treatment with a probiotic attenuates PNGR-associated PH. METHOD: Pups were randomized at birth to room air or 75% oxygen (hyperoxia), to normal milk intake (10 pups/dam) or PNGR (17 pups/dam), and to probiotic Lactobacillus reuteri DSM 17938 or phosphate-buffered saline. After 14 days, PH was assessed by echocardiography and right ventricular hypertrophy (RVH) was assessed by Fulton's index (right ventricular weight/left ventricle + septal weight). The small bowel and cecum were analyzed by high-throughput 16S ribosomal RNA gene sequencing. RESULTS: PNGR with or without hyperoxia (but not hyperoxia alone) altered the microbiota of the distal small bowel and cecum. Treatment with DSM 17938 attenuated PH and RVH in pups with PNGR, but not hyperoxia alone. DSM 17938 treatment decreased α-diversity. The intestinal microbiota differed based on oxygen exposure, litter size, and probiotic treatment. CONCLUSION: PNGR causes intestinal dysbiosis and PH. Treatment with DSM 17938 prevents PNGR-associated RVH and PH. Changes in the developing intestine and intestinal microbiota impact the developing lung vasculature and RV.


Asunto(s)
Restricción Calórica/efectos adversos , Ciego/microbiología , Microbioma Gastrointestinal , Hipertensión Pulmonar/prevención & control , Intestino Delgado/microbiología , Limosilactobacillus reuteri/fisiología , Pulmón/irrigación sanguínea , Probióticos/administración & dosificación , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Disbiosis , Femenino , Hiperoxia/complicaciones , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/microbiología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/microbiología , Hipertrofia Ventricular Derecha/fisiopatología , Hipertrofia Ventricular Derecha/prevención & control , Tamaño de la Camada , Estado Nutricional , Embarazo , Ratas Sprague-Dawley
10.
Am J Perinatol ; 37(8): 813-824, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32268381

RESUMEN

The first case of novel coronavirus disease of 2019 (COVID-19) caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) was reported in November2019. The rapid progression to a global pandemic of COVID-19 has had profound medical, social, and economic consequences. Pregnant women and newborns represent a vulnerable population. However, the precise impact of this novel virus on the fetus and neonate remains uncertain. Appropriate protection of health care workers and newly born infants during and after delivery by a COVID-19 mother is essential. There is some disagreement among expert organizations on an optimal approach based on resource availability, surge volume, and potential risk of transmission. The manuscript outlines the precautions and steps to be taken before, during, and after resuscitation of a newborn born to a COVID-19 mother, including three optional variations of current standards involving shared-decision making with parents for perinatal management, resuscitation of the newborn, disposition, nutrition, and postdischarge care. The availability of resources may also drive the application of these guidelines. More evidence and research are needed to assess the risk of vertical and horizontal transmission of SARS-CoV-2 and its impact on fetal and neonatal outcomes. KEY POINTS: · The risk of vertical transmission is unclear; transmission from family members/providers to neonates is possible.. · Optimal personal-protective-equipment (airborne vs. droplet/contact precautions) for providers is crucial to prevent transmission.. · Parents should be engaged in shared decision-making with options for rooming in, skin-to-skin contact, and breastfeeding..


Asunto(s)
Infecciones por Coronavirus , Control de Infecciones , Pandemias , Neumonía Viral , Complicaciones Infecciosas del Embarazo , Resucitación , Gestión de Riesgos/métodos , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Femenino , Humanos , Recién Nacido , Control de Infecciones/métodos , Control de Infecciones/organización & administración , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Cuidado Intensivo Neonatal/métodos , Cuidado Intensivo Neonatal/organización & administración , Pandemias/prevención & control , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neumonía Viral/prevención & control , Embarazo , Complicaciones Infecciosas del Embarazo/epidemiología , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/virología , Resucitación/métodos , Resucitación/tendencias , SARS-CoV-2
11.
J Interprof Care ; 34(5): 682-686, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32674638

RESUMEN

The COVID-19 pandemic has created multiple, complex and intense demands on hospitals, including the need for surge planning in the many locations outside epicenters such as northern Italy or New York City. We here describe such surge planning in an Academic Health Center that encompasses a children's hospital. Interprofessional teams from every aspect of inpatient care and hospital operations worked to prepare for a COVID-19 surge. In so doing, they successfully innovated ways to integrate pediatric and adult care and maximize bed capacity. The success of this intense collaborative effort offers an opportunity for ongoing teamwork to enhance efficient, effective, and high-quality patient care.


Asunto(s)
Conducta Cooperativa , Infecciones por Coronavirus , Comunicación Interdisciplinaria , Pandemias , Grupo de Atención al Paciente , Neumonía Viral , Centros Médicos Académicos , Betacoronavirus , COVID-19 , Fuerza Laboral en Salud/organización & administración , Hospitales Pediátricos , Humanos , Italia , Ciudad de Nueva York , Estudios de Casos Organizacionales , SARS-CoV-2
12.
J Proteome Res ; 18(3): 912-922, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30638015

RESUMEN

Over the course of milk digestion, native milk proteases and infant digestive proteases fragment intact proteins into peptides with potential bioactivity. This study investigated the release of peptides over 3 h of gastric digestion in 14 preterm infant sample sets. The peptide content was extracted and analyzed from milk and gastric samples via Orbitrap tandem mass spectrometry. The relative ion intensity (abundance) and count of peptides in each sample were compared over time and between infants fed milk fortified with bovine milk fortifier and infants fed unfortified milk. Bioactivity of the identified peptides was predicted by sequence homology to known bioactive milk peptides. Both total and bioactive peptide abundance and count continuously increased over 3 h of gastric digestion. After accounting for infant weight, length, and postconceptual age, fortification of milk limited the release of peptides from human milk proteins. Peptides that survived further gastric digestion after their initial release were structurally more similar to bioactive peptides than nonsurviving peptides. This work is the first to provide a comprehensive profile of milk peptides released during gastric digestion over time, which is an essential step in determining which peptides are most likely to be biologically relevant in the infant. Data are available via ProteomeXchange with identifier PXD012192.


Asunto(s)
Mucosa Gástrica/metabolismo , Recien Nacido Prematuro/metabolismo , Proteínas de la Leche/metabolismo , Péptidos/análisis , Proteómica/métodos , Animales , Bovinos , Humanos , Lactante , Recién Nacido , Leche Humana/química , Estómago , Espectrometría de Masas en Tándem
13.
J Nutr ; 149(6): 1075-1088, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31006815

RESUMEN

BACKGROUND: Infancy is a crucial period for establishing the intestinal microbiome. This process may be influenced by vitamin A (VA) status because VA affects intestinal immunity and epithelial integrity, factors that can, in turn, modulate microbiome development. OBJECTIVES: The aim of this study was to determine if neonatal VA supplementation (VAS) affected the abundance of Bifidobacterium, a beneficial commensal, or of Proteobacteria, a phylum containing enteric pathogens, in early (6-15 wk) or late (2 y) infancy. Secondary objectives were to determine if VAS affected the abundance of other bacterial taxa, and to determine if VA status assessed by measuring plasma retinol was associated with bacterial abundance. METHODS: Three hundred and six Bangladeshi infants were randomized by sex and birthweight status (above/below median) to receive 1 VA dose (50,000 IU) or placebo within 48 h of birth. Relative abundance at the genus level and above was assessed by 16S rRNA gene sequencing. A terminal restriction fragment-length polymorphism assay was used to identify Bifidobacterium species and subspecies at 6 wk. RESULTS: Linear regression showed that Bifidobacterium abundance in early infancy was lower in boys (median, 1st/3rd quartiles; 0.67, 0.52/0.78) than girls (0.73, 0.60/0.80; P = 0.003) but that boys receiving VAS (0.69, 0.55/0.78) had higher abundance than boys receiving placebo (0.65, 0.44/0.77; P = 0.039). However this difference was not seen in girls (VAS 0.71, 0.54/0.80; placebo 0.75, 0.63/0.81; P = 0.25). VAS did not affect Proteobacteria abundance. Sex-specific associations were also seen for VA status, including positive associations of plasma retinol with Actinobacteria (the phylum containing Bifidobacterium) and Akkermansia, another commensal with possible health benefits, for girls in late infancy. CONCLUSIONS: Better VA status in infancy may influence health both in infancy and later in life by promoting the establishment of a healthy microbiota. This postulated effect of VA may differ between boys and girls. This trial was registered at clinicaltrials.gov as NCT02027610.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Vitamina A/administración & dosificación , Bangladesh , Bifidobacterium/efectos de los fármacos , Bifidobacterium/aislamiento & purificación , Preescolar , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Estudios Longitudinales , Masculino , Estado Nutricional , Proteobacteria/efectos de los fármacos , Proteobacteria/aislamiento & purificación , Vitamina A/sangre
14.
Pediatr Res ; 86(6): 749-757, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31443102

RESUMEN

BACKGROUND: Infant gut dysbiosis, often associated with low abundance of bifidobacteria, is linked to impaired immune development and inflammation-a risk factor for increased incidence of several childhood diseases. We investigated the impact of B. infantis EVC001 colonization on enteric inflammation in a subset of exclusively breastfed term infants from a larger clinical study. METHODS: Stool samples (n = 120) were collected from infants randomly selected to receive either 1.8 × 1010 CFU B. infantis EVC001 daily for 21 days (EVC001) or breast milk alone (controls), starting at day 7 postnatal. The fecal microbiome was analyzed using 16S ribosomal RNA, proinflammatory cytokines using multiplexed immunoassay, and fecal calprotectin using ELISA at three time points: days 6 (Baseline), 40, and 60 postnatal. RESULTS: Fecal calprotectin concentration negatively correlated with Bifidobacterium abundance (P < 0.0001; ρ = -0.72), and proinflammatory cytokines correlated with Clostridiaceae and Enterobacteriaceae, yet negatively correlated with Bifidobacteriaceae abundance. Proinflammatory cytokines were significantly lower in EVC001-fed infants on days 40 and 60 postnatally compared to baseline and compared to control infants. CONCLUSION: Our findings indicate that gut dysbiosis (absence of B. infantis) is associated with increased intestinal inflammation. Early addition of EVC001 to diet represents a novel strategy to prevent enteric inflammation during a critical developmental phase.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Lactancia Materna , Enteritis/prevención & control , Citocinas/metabolismo , Enteritis/metabolismo , Enteritis/microbiología , Heces/química , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Recién Nacido , Mediadores de Inflamación/metabolismo , Complejo de Antígeno L1 de Leucocito/análisis , Masculino , Estudios Prospectivos
15.
Curr Opin Pediatr ; 31(2): 188-194, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30664022

RESUMEN

PURPOSE OF REVIEW: Probiotic administration to premature infants for the purpose of prevention of necrotizing enterocolitis is common in many parts of the world but uncommon in the United States. The present review will emphasize recent findings in support of routine administration of probiotics to this highly vulnerable population. RECENT FINDINGS: Additional evidence from animal models describing mechanisms of protection of probiotics in the immature gut and updated meta-analyses of randomized placebo-controlled trials and observational cohorts are presented (now including more than 40 000 premature infants from countries across the globe). SUMMARY: The preponderance of evidence suggests that probiotic administration to premature infants is well tolerated and decreases the risk of death, necrotizing enterocolitis, and sepsis. Further comparisons of probiotic administration to placebo are not likely to alter these conclusions. Rather, future work should focus on assurance of high-quality products with demonstrated purity and viability of probiotic microbes, and future clinical trials should focus on comparisons between high-quality products and doses.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Prematuro , Probióticos , Sepsis , Animales , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/prevención & control , Humanos , Recién Nacido , Recien Nacido Prematuro , Probióticos/uso terapéutico
16.
Can J Physiol Pharmacol ; 97(3): 197-205, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30512966

RESUMEN

In the premature infant, poor growth in utero (fetal growth restriction) and in the first weeks of life (postnatal growth restriction) are associated with increased risk for bronchopulmonary dysplasia and pulmonary hypertension. In this review, we summarize the epidemiologic data supporting these associations, present a novel rodent model of postnatal growth restriction, and review 5 promising mechanisms by which poor nutrition may affect the developing lung. These observations support the hypothesis that nutritional and (or) pharmacologic interventions early in life may be able to decrease risk of the pulmonary complications of extreme prematurity.


Asunto(s)
Displasia Broncopulmonar/epidemiología , Displasia Broncopulmonar/etiología , Hipertensión Pulmonar/epidemiología , Hipertensión Pulmonar/etiología , Recien Nacido Prematuro/fisiología , Animales , Humanos , Riesgo
17.
Telemed J E Health ; 25(9): 775-780, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30394853

RESUMEN

Introduction: Outcomes for premature and critically ill neonates are improved with care provided by neonatologists in a neonatal intensive care unit (NICU). For smaller hospitals, maintaining the personnel and equipment necessary for the delivery and care of unexpectedly high-risk neonates is a significant challenge. To address this disparity in access, telemedicine has been increasingly used to support providers, patients, and their families in community newborn nurseries and NICUs. The purpose of this review is to present the current state of the use of telemedicine by regional NICUs to support community newborn nurseries, NICUs, and families. Methods: A literature review was conducted by two independent reviewers. Articles were selected for inclusion if they described the use of telemedicine with neonates or in the NICU. Two reviewers assessed the quality of the articles using the National Heart, Lung, and Blood Institute Study Quality Assessment Tools. Results: Fourteen articles were identified. After consensus discussion, eight of the articles were rated good and six were rated fair by the two reviewers. Many of the articles suggested improvements in quality of care, family satisfaction, and reductions in the cost of care. Unfortunately, a majority of the studies to date have had small sample sizes or were performed in a single institution and lacked robust evaluations of patient- and family-centered outcomes and provider decision making. Conclusions: While these early studies are promising, more robust studies involving more patients and more institutions are needed to identify opportunities where telemedicine can impact health outcomes, patient-centeredness, or costs of care of neonates.


Asunto(s)
Enfermedad Crítica/terapia , Disparidades en Atención de Salud/estadística & datos numéricos , Neonatólogos/estadística & datos numéricos , Evaluación de Resultado en la Atención de Salud , Telemedicina/estadística & datos numéricos , Enfermedad Crítica/epidemiología , Femenino , Disparidades en Atención de Salud/economía , Humanos , Recién Nacido , Unidades de Cuidado Intensivo Neonatal/organización & administración , Masculino , Especialización/estadística & datos numéricos , Estados Unidos
18.
Minerva Pediatr ; 71(3): 253-262, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31017382

RESUMEN

Intestinal dysbiosis is associated with a long list of both acute and chronic inflammatory diseases and appears to be increasing in developed countries over the last century with the introduction of antibiotics, changes in sanitation, formula feeding and cesarean sections. The evidence supporting prophylactic administration of probiotic microbes to very preterm infants for the prevention of necrotizing enterocolitis, late onset sepsis and death is strong. The evidence for benefit of probiotics in infantile colic is strong but limited to the L. reuteri DSM 17938 strain and to breast-fed infants. The evidence for prevention of atopic dermatitis is mixed with the strongest benefit seen with initiation of probiotic treatment during pregnancy and continued after birth. The more provocative question of whether routine administration of probiotics to all infants can reverse trends in intestinal dysbiosis and dysbiosis-associated diseases remains unanswered. A large cohort study or randomized controlled trial of probiotics in infancy with sufficient follow-up to assess changes in dysbiosis-associated diseases is warranted and could be paradigm shifting.


Asunto(s)
Disbiosis/complicaciones , Microbioma Gastrointestinal/fisiología , Probióticos/administración & dosificación , Cólico/prevención & control , Disbiosis/microbiología , Disbiosis/terapia , Humanos , Lactante , Recien Nacido Prematuro , Intestinos/microbiología
19.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L870-L881, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30113229

RESUMEN

Pulmonary hypertension (PH) is a common consequence of bronchopulmonary dysplasia (BPD) and remains a primary contributor to increased morbidity and mortality among preterm infants. Unfortunately, at the present time, there are no reliable early predictive markers for BPD-associated PH. Considering its health consequences, understanding in utero perturbations that lead to the development of BPD and BPD-associated PH and identifying early predictive markers is of utmost importance. As part of the discovery phase, we applied a multiplatform metabolomics approach consisting of untargeted and targeted methodologies to screen for metabolic perturbations in umbilical cord blood (UCB) plasma from preterm infants that did ( n = 21; cases) or did not ( n = 21; controls) develop subsequent PH. A total of 1,656 features were detected, of which 407 were annotated by metabolite structures. PH-associated metabolic perturbations were characterized by reductions in major choline-containing phospholipids, such as phosphatidylcholines and sphingomyelins, indicating altered lipid metabolism. The reduction in UCB abundances of major choline-containing phospholipids was confirmed in an independent validation cohort consisting of UCB plasmas from 10 cases and 10 controls matched for gestational age and BPD status. Subanalyses in the discovery cohort indicated that elevations in the oxylipins PGE1, PGE2, PGF2a, 9- and 13-HOTE, 9- and 13-HODE, and 9- and 13-KODE were positively associated with BPD presence and severity. This expansive evaluation of cord blood plasma identifies compounds reflecting dyslipidemia and suggests altered metabolite provision associated with metabolic immaturity that differentiate subjects, both by BPD severity and PH development.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Dislipidemias/metabolismo , Sangre Fetal/metabolismo , Hipertensión Pulmonar/metabolismo , Biomarcadores/metabolismo , Femenino , Edad Gestacional , Humanos , Recién Nacido , Recien Nacido Prematuro , Metabolismo de los Lípidos/fisiología , Masculino , Metabolómica/métodos
20.
J Pediatr Gastroenterol Nutr ; 66(2): 318-324, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28906320

RESUMEN

OBJECTIVE: Our previous studies suggested that human milk proteases begin to hydrolyze proteins in the mammary gland and continue within the term infant' stomach. No research has measured milk protease and pepsin activity in the gastric aspirates of preterm infants after human milk feeding. This study investigated how the concentrations of human milk proteases and protease inhibitors changed in the premature infant stomach. METHODS: Human milk and infant gastric samples were collected from 18 preterm-delivering mother-infant pairs (24-32 week gestational age). Paired human milk and gastric samples were collected across postnatal age (2-47 days). Protease concentrations were determined by spectrophotometric or fluorometric assays, and the concentrations of protease inhibitors and bioactive proteins were determined by enzyme-linked immunosorbent assay. Paired t tests were applied to compare enzymes, antiproteases, and bioactive proteins between human milk and gastric samples. RESULTS: Our study reveals that although human milk proteases, including carboxypeptidase B2, kallikrein, plasmin, cathepsin D, elastase, thrombin, and cytosol aminopeptidase, are present in the preterm infant stomach, only plasmin and cathepsin D can actively hydrolyze proteins at gastric pH. Enzyme-linked immunosorbent assay and peptidomic evidence suggest that all milk antiproteases as well as lactoferrin and immunoglobulin A are partially digested in the preterm stomach. CONCLUSIONS: Most human milk proteases are active in milk but not at preterm infant gastric pH. Only cathepsin D and plasmin have potential to continue degrading milk proteins within the preterm infant stomach.


Asunto(s)
Proteínas de la Leche/metabolismo , Leche Humana/metabolismo , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/metabolismo , Estómago/fisiología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Lactante , Recién Nacido , Recien Nacido Prematuro/metabolismo , Masculino , Proteolisis , Espectrofotometría/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA