Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Nutr ; 62(3): 1195-1205, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36460779

RESUMEN

BACKGROUND: Supply of choline is not guaranteed in current preterm infant nutrition. Choline serves in parenchyma formation by membrane phosphatidylcholine (PC), plasma transport of poly-unsaturated fatty acids (PUFA) via PC, and methylation processes via betaine. PUFA-PC concentrations are high in brain, liver and lung, and deficiency may result in developmental disorders. We compared different deuterated (D9-) choline components for kinetics of D9-choline, D9-betaine and D9-PC. METHODS: Prospective study (1/2021-12/2021) in 32 enterally fed preterm infants (28 0/7-32 0/7 weeks gestation). Patients were randomized to receive enterally a single dose of 2.7 mg/kg D9-choline-equivalent as D9-choline chloride, D9-phosphoryl-choline, D9-glycerophosphorylcholine (D9-GPC) or D9-1-palmitoyl-2-oleoyl-PC(D9-POPC), followed by blood sampling at 1 + 24 h or 12 + 60 h after administration. Plasma concentrations were analyzed by tandem mass spectrometry. Results are expressed as median (25th/75th percentile). RESULTS: At 1 h, plasma D9-choline was 1.8 (0.9/2.2) µmol/L, 1.3 (0.9/1.5) µmol/L and 1.2 (0.7/1.4) µmol/L for D9-choline chloride, D9-GPC and D9-phosphoryl-choline, respectively. D9-POPC did not result in plasma D9-choline. Plasma D9-betaine was maximal at 12 h, with lowest concentrations after D9-POPC. Maximum plasma D9-PC values at 12 h were the highest after D9-POPC (14.4 (9.1/18.9) µmol/L), compared to the other components (D9-choline chloride: 8.1 [5.6/9.9] µmol/L; D9-GPC: 8.4 (6.2/10.3) µmol/L; D9-phosphoryl-choline: 9.8 (8.6/14.5) µmol/L). Predominance of D9-PC comprising linoleic, rather than oleic acid, indicated fatty-acyl remodeling of administered D9-POPC prior to systemic delivery. CONCLUSION: D9-Choline chloride, D9-GPC and D9-phosphoryl-choline equally increased plasma D9-choline and D9-betaine. D9-POPC shifted metabolism from D9-betaine to D9-PC. Combined supplementation of GPC and (PO) PC may be best suited to optimize choline supply in preterm infants. Due to fatty acid remodeling of (PO) PC during its assimilation, PUFA co-supplementation with (PO) PC may increase PUFA-delivery to critical organs. This study was registered (22.01.2020) at the Deutsches Register Klinischer Studien (DRKS) (German Register for Clinical Studies), DRKS00020502. STUDY REGISTRATION: This study was registered at the Deutsches Register Klinischer Studien (DRKS) (German Register for Clinical Studies), DRKS00020502.


Asunto(s)
Betaína , Colina , Lactante , Humanos , Recién Nacido , Recien Nacido Prematuro , Deuterio , Estudios Prospectivos , Ácidos Grasos Insaturados , Fosfatidilcolinas , Suplementos Dietéticos
2.
Eur J Nutr ; 62(4): 1795-1807, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36840817

RESUMEN

BACKGROUND: Choline deficiency leads to pathologies particularly of the liver, brain and lung. Adequate supply is important for preterm infants and patients with cystic fibrosis. We analysed the assimilation of four different enterally administered deuterium-labelled (D9-) choline supplements in adults. METHODS: Prospective randomised cross-over study (11/2020-1/2022) in six healthy men, receiving four single doses of 2.7 mg/kg D9-choline equivalent each in the form of D9-choline chloride, D9-phosphorylcholine, D9-alpha-glycerophosphocholine (D9-GPC) or D9-1-palmitoyl-2-oleoyl-glycero-3-phosphoryl-choline (D9-POPC), in randomised order 6 weeks apart. Plasma was obtained at baseline (t = - 0.1 h) and at 0.5 h to 7d after intake. Concentrations of D9-choline and its D9-labelled metabolites were analysed by tandem mass spectrometry. Results are shown as median and interquartile range. RESULTS: Maximum D9-choline and D9-betaine concentrations were reached latest after D9-POPC administration versus other components. D9-POPC and D9-phosphorylcholine resulted in lower D9-trimethylamine (D9-TMAO) formation. The AUCs (0-7d) of plasma D9-PC concentration showed highest values after administration of D9-POPC. D9-POPC appeared in plasma after fatty acid remodelling, predominantly as D9-1-palmitoyl-2-linoleyl-PC (D9-PLPC), confirming cleavage to 1-palmitoyl-lyso-D9-PC and re-acylation with linoleic acid as the most prominent alimentary unsaturated fatty acid. CONCLUSION: There was a delayed increase in plasma D9-choline and D9-betaine after D9-POPC administration, with no differences in AUC over time. D9-POPC resulted in a higher AUC of D9-PC and virtually absent D9-TMAO levels. D9-POPC is remodelled according to enterocytic fatty acid availability. D9-POPC seems best suited as choline supplement to increase plasma PC concentrations, with PC as a carrier of choline and targeted fatty acid supply as required by organs. This study was registered at Deutsches Register Klinischer Studien (DRKS) (German Register for Clinical Studies), DRKS00020498, 22.01.2020. STUDY REGISTRATION: This study was registered at Deutsches Register Klinischer Studien (DRKS) (German Register for Clinical Studies), DRKS00020498.


Asunto(s)
Betaína , Fosforilcolina , Adulto , Humanos , Lactante , Recién Nacido , Masculino , Colina , Estudios Cruzados , Deuterio , Ácidos Grasos , Recien Nacido Prematuro , Fosfatidilcolinas , Estudios Prospectivos
3.
Eur J Nutr ; 61(1): 219-230, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34287673

RESUMEN

BACKGROUND: Adequate intake of choline is essential for growth and homeostasis, but its supply does often not meet requirements. Choline deficiency decreases phosphatidylcholine (PC) and betaine synthesis, resulting in organ pathology, especially of liver, lung, and brain. This is of particular clinical importance in preterm infants and cystic fibrosis patients. We compared four different choline supplements for their impact on plasma concentration and kinetics of choline, betaine as a methyl donor and trimethylamine oxide (TMAO) as a marker of bacterial degradation prior to absorption. METHODS: Prospective randomized cross-over study (1/2020-4/2020) in six healthy adult men. Participants received a single dose of 550 mg/d choline equivalent in the form of choline chloride, choline bitartrate, α-glycerophosphocholine (GPC), and egg-PC in randomized sequence at least 1 week apart. Blood was taken from t = - 0.1-6 h after supplement intake. Choline, betaine, TMAO, and total PC concentrations were analyzed by tandem mass spectrometry. Results are shown as medians and interquartile range. RESULTS: There was no difference in the AUC of choline plasma concentrations after intake of the different supplements. Individual plasma kinetics of choline and betaine differed and concentrations peaked latest for PC (at ≈3 h). All supplements similarly increased plasma betaine. All water-soluble supplements rapidly increased TMAO, whereas egg-PC did not. CONCLUSION: All supplements tested rapidly increased choline and betaine levels to a similar extent, with egg-PC showing the latest peak. Assuming that TMAO may have undesirable effects, egg-PC might be best suited for choline supplementation in adults. STUDY REGISTRATION: This study was registered at "Deutsches Register Klinischer Studien" (DRKS) (German Register for Clinical Studies), 17.01.2020, DRKS00020454.


Asunto(s)
Colina , Adulto , Betaína , Suplementos Dietéticos , Humanos , Masculino , Estudios Prospectivos , Voluntarios
4.
Am J Respir Cell Mol Biol ; 64(3): 379-390, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33351709

RESUMEN

Obesity and type 2 diabetes are nutrition-related conditions associated with lung function impairment and pulmonary diseases; however, the underlying pathomechanisms are incompletely understood. Pulmonary surfactant is essential for lung function, and surfactant synthesis by AT2 (alveolar epithelial type 2) cells relies on nutrient uptake. We hypothesized that dietary amounts of carbohydrates or fat affect surfactant homeostasis and composition. Feeding mice a starch-rich diet (StD), sucrose-rich diet (SuD), or fat-rich diet (FaD) for 30 weeks resulted in hypercholesterolemia and hyperinsulinemia compared with a fiber-rich control diet. In SuD and FaD groups, lung mechanic measurements revealed viscoelastic changes during inspiration, indicating surfactant alterations, and interfacial adsorption of isolated surfactant at the air-liquid interface was decreased under FaD. The composition of characteristic phospholipid species was modified, including a shift from dipalmitoyl-phosphatidylcholine (PC16:0/16:0) to palmitoyl-palmitoleoyl-phosphatidylcholine (PC16:0/16:1) in response to carbohydrates and decreased myristic acid-containing phosphatidylcholine species (PC14:0/14:0; PC16:0/14:0) on excess fat intake, as well as higher palmitoyl-oleoyl-phosphatidylglycerol (PG16:0/18:1) and palmitoyl-linoleoyl-phosphatidylglycerol (PG16:0/18:2) fractions in StD, SuD, and FaD groups than in the control diet. Moreover, mRNA expression levels of surfactant synthesis-related proteins within AT2 cells were altered. Under the StD regimen, AT2 cells showed prominent lipid accumulations and smaller lamellar bodies. Thus, in an established mouse model, distinct diet-related surfactant alterations were subtle, yet detectable, and may become challenging under conditions of reduced respiratory capacity. Dietary fat was the only macronutrient significantly affecting surfactant function. This warrants future studies examining alimentary effects on lung surfactant, with special regard to pulmonary complications in obesity and type 2 diabetes.


Asunto(s)
Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Surfactantes Pulmonares/metabolismo , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Fenómenos Biomecánicos , Forma de la Célula/efectos de los fármacos , Glucosa/metabolismo , Homeostasis , Espacio Intracelular/metabolismo , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/ultraestructura , Pulmón/fisiología , Masculino , Ratones Endogámicos C57BL , Fosfolípidos/sangre
5.
Eur J Nutr ; 60(4): 1737-1759, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32797252

RESUMEN

BACKGROUND: Cystic Fibrosis (CF) is an autosomal recessive disorder with life-threatening organ manifestations. 87% of CF patients develop exocrine pancreas insufficiency, frequently starting in utero and requiring lifelong pancreatic enzyme substitution. 99% develop progressive lung disease, and 20-60% CF-related liver disease, from mild steatosis to cirrhosis. Characteristically, pancreas, liver and lung are linked by choline metabolism, a critical nutrient in CF. Choline is a tightly regulated tissue component in the form of phosphatidylcholine (Ptd'Cho) and sphingomyelin (SPH) in all membranes and many secretions, particularly of liver (bile, lipoproteins) and lung (surfactant, lipoproteins). Via its downstream metabolites, betaine, dimethylglycine and sarcosine, choline is the major one-carbon donor for methionine regeneration from homocysteine. Methionine is primarily used for essential methylation processes via S-adenosyl-methionine. CLINICAL IMPACT: CF patients with exocrine pancreas insufficiency frequently develop choline deficiency, due to loss of bile Ptd'Cho via feces. ~ 50% (11-12 g) of hepatic Ptd'Cho is daily secreted into the duodenum. Its re-uptake requires cleavage to lyso-Ptd'Cho by pancreatic and small intestinal phospholipases requiring alkaline environment. Impaired CFTR-dependent bicarbonate secretion, however, results in low duodenal pH, impaired phospholipase activity, fecal Ptd'Cho loss and choline deficiency. Low plasma choline causes decreased availability for parenchymal Ptd'Cho metabolism, impacting on organ functions. Choline deficiency results in hepatic choline/Ptd'Cho accretion from lung tissue via high density lipoproteins, explaining the link between choline deficiency and lung function. Hepatic Ptd'Cho synthesis from phosphatidylethanolamine by phosphatidylethanolamine-N-methyltransferase (PEMT) partly compensates for choline deficiency, but frequent single nucleotide polymorphisms enhance choline requirement. Additionally, small intestinal bacterial overgrowth (SIBO) frequently causes intraluminal choline degradation in CF patients prior to its absorption. As adequate choline supplementation was clinically effective and adult as well as pediatric CF patients suffer from choline deficiency, choline supplementation in CF patients of all ages should be evaluated.


Asunto(s)
Fibrosis Quística , Microbioma Gastrointestinal , Adulto , Niño , Colina , Humanos , Hígado , Páncreas , Fosfatidiletanolamina N-Metiltransferasa
6.
Eur J Nutr ; 59(2): 729-739, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30859363

RESUMEN

OBJECTIVE: Choline and docosahexaenoic acid (DHA) are essential nutrients for preterm infant development. They are metabolically linked via phosphatidylcholine (PC), a constitutive plasma membrane lipid and the major transport form of DHA in plasma. Plasma choline and DHA-PC concentrations rapidly decline after preterm birth. To improve preterm infant nutrition, we evaluated combined compared to exclusive choline and DHA supplementation, and standard feeding. DESIGN: Randomized partially blinded single-center trial. SETTING: Neonatal tertiary referral center in Tübingen, Germany. PATIENTS: 24 inborn preterm infants < 32 week postmenstrual age. INTERVENTIONS: Standard nutrition (control) or, additionally, enteral choline (30 mg/kg/day), DHA (60 mg/kg/day), or both for 10 days. Single enteral administration of 3.6 mg/kg [methyl-D9-] choline chloride as a tracer at 7.5 days. MAIN OUTCOME MEASURES: Primary outcome variable was plasma choline following 7 days of supplementation. Deuterated and unlabeled choline metabolites, DHA-PC, and other PC species were secondary outcome variables. RESULTS: Choline supplementation increased plasma choline to near-fetal concentrations [35.4 (32.8-41.7) µmol/L vs. 17.8 (16.1-22.4) µmol/L, p < 0.01] and decreased D9-choline enrichment of PC. Single DHA treatment decreased DHA in PC relative to total lipid [66 (60-68)% vs. 78 (74-80)%; p < 0.01], which was prevented by choline. DHA alone increased DHA-PC only by 35 (26-45)%, but combined treatment by 63 (49-74)% (p < 0.001). D9-choline enrichment showed preferential synthesis of PC containing linoleic acid. PC synthesis via phosphatidylethanolamine methylation resulted in preferential synthesis of DHA-containing D3-PC, which was increased by choline supplementation. CONCLUSIONS: 30 mg/kg/day additional choline supplementation increases plasma choline to near-fetal concentrations, dilutes the D9-choline tracer via increased precursor concentrations and improves DHA homeostasis in preterm infants. TRIAL REGISTRATION: clinicaltrials.gov. Identifier: NCT02509728.


Asunto(s)
Colina/sangre , Colina/farmacología , Ácidos Docosahexaenoicos/sangre , Ácidos Docosahexaenoicos/farmacología , Fenómenos Fisiológicos Nutricionales del Lactante/efectos de los fármacos , Recien Nacido Prematuro , Biomarcadores/sangre , Colina/administración & dosificación , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Quimioterapia Combinada/métodos , Nutrición Enteral/métodos , Femenino , Alemania , Humanos , Recién Nacido , Masculino
7.
Eur J Nutr ; 58(3): 931-945, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30298207

RESUMEN

BACKGROUND: Choline is an essential nutrient, with increased requirements during development. It forms the headgroup of phosphatidylcholine and sphingomyelin in all membranes and many secretions. Phosphatidylcholine is linked to cell signaling as a phosphocholine donor to synthesize sphingomyelin from ceramide, a trigger of apoptosis, and is the major carrier of arachidonic and docosahexaenoic acid in plasma. Acetylcholine is important for neurodevelopment and the placental storage form for fetal choline supply. Betaine, a choline metabolite, functions as osmolyte and methyl donor. Their concentrations are all tightly regulated in tissues. CLINCAL IMPACT: During the fetal growth spurt at 24-34-week postmenstrual age, plasma choline is higher than beyond 34 weeks, and threefold higher than in pregnant women [45 (36-60) µmol/L vs. 14 (10-17) µmol/L]. The rapid decrease in plasma choline after premature birth suggests an untimely reduction in choline supply, as cellular uptake is proportional to plasma concentration. Supply via breast milk, with phosphocholine and α-glycerophosphocholine as its major choline components, does not prevent such postnatal decrease. Moreover, high amounts of liver PC are secreted via bile, causing rapid hepatic choline turnover via the enterohepatic cycle, and deficiency in case of pancreatic phospholipase A2 deficiency or intestinal resection. Choline deficiency causes hepatic damage and choline accretion at the expense of the lungs and other tissues. CONCLUSION: Choline deficiency may contribute to the impaired lean body mass growth and pulmonary and neurocognitive development of preterm infants despite adequate macronutrient supply and weight gain. In this context, a reconsideration of current recommendations for choline supply to preterm infants is required.


Asunto(s)
Desarrollo Infantil/fisiología , Deficiencia de Colina/sangre , Colina/sangre , Recien Nacido Prematuro/crecimiento & desarrollo , Betaína/sangre , Femenino , Humanos , Lactante , Recién Nacido de Bajo Peso , Recién Nacido , Leche Humana , Fosfatidilcolinas/sangre , Embarazo , Esfingomielinas/sangre
8.
Eur J Nutr ; 57(6): 2105-2112, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28638995

RESUMEN

BACKGROUND: Docosahexaenoic (C22:6) and arachidonic (C20:4) acids are long-chain polyunsaturated fatty acids (LC-PUFA) essential to neonatal development, being present in the glycerophospholipids of all organs, particularly the brain. In plasma, LC-PUFAs are mainly present in lipoprotein lipids, which are neutral lipids (triglycerides and cholesterol esters) and glycerophospholipids, like choline containing phosphatidylcholine (PC). PURPOSE: To guide future supplementation strategies of C22:6 and C20:4 in combination with choline, we determined the distribution of C20:4 and C22:6 between PC and neutral lipid. METHODS: Preterm infant plasma (N = 59, postmenstrual age [PMA] 33.9 wk (32.4-36.0)) and cord plasma (N = 34, PMA 34.0 wk (30.86-38.4)) were investigated. PC and neutral lipids were extracted and analyzed using tandem mass spectrometry and gas chromatography, respectively. Data are reported as medians and 25th/75th percentiles. RESULTS: In cord blood, C20:4-PC and C22:6-PC comprised 36.1% (34.2-38.6) and 10.2% (8.8-12.8) of total PC, respectively. In preterm infant plasma, values were only 20.8% (19.2-23.1) and 5.7% (5.2-6.0), respectively (p < 0.001 each). Nevertheless, in preterm infant plasma, 80.6% (77.6-83.0) of C20:4 and 86.0% (83.0-88.9) of C22:6 were found in PC. These values exceeded the proportions of C20:4 and C22:6 in PC of cord plasma [71.3% (67.8-72.9) and 79.2% (75.2-85.4), respectively] (p < 0.0001 each). CONCLUSION: Irrespective of the low proportions of C20:4-PC and C22:6-PC in preterm infant plasma lipids, PC is the major transporter for C20:4 and C22:6. Our data support the hypotheses that choline deficiency may impair end-organ availability of these LC-PUFA in preterm infants. Therefore, supplementation of C20:4 and C22:6 might better be accompanied by choline supplementation.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Recien Nacido Prematuro , Fosfatidilcolinas/metabolismo , Ácidos Grasos , Femenino , Cromatografía de Gases y Espectrometría de Masas , Alemania , Humanos , Recién Nacido , Masculino , Estado Nutricional
9.
BMC Pediatr ; 18(1): 154, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739389

RESUMEN

BACKGROUND: Feeding breast milk is associated with reduced morbidity and mortality, as well as improved neurodevelopmental outcome but does not meet the high nutritional requirements of preterm infants. Both plasma and urinary urea concentrations represent amino acid oxidation and low concentrations may indicate insufficient protein supply. This study assesses the effect of different levels of enteral protein on plasma and urinary urea concentrations and determines if the urinary urea-creatinine ratio provides reliable information about the protein status of preterm infants. METHODS: Sixty preterm infants (birthweight < 1500 g; gestational age < 32 weeks) were enrolled in a randomized controlled trial and assigned to either a lower-protein group (median protein intake 3.7 g/kg/d) or a higher-protein group (median protein intake 4,3 g/kg/d). Half the patients in the higher-protein group received standardized supplementation with a supplement adding 1.8 g protein/100 ml milk, the other half received individual supplementation depending on the respective mother's milk macronutrient content. Plasma urea concentration was determined in two scheduled blood samples (BS1; BS2); urinary urea and creatinine concentrations in weekly spot urine samples. RESULTS: The higher-protein group showed higher plasma urea concentrations in both BS1 and BS2 and a higher urinary urea-creatinine-ratio in week 3 and 5-7 compared to the lower-protein group. In addition, a highly positive correlation between plasma urea concentrations and the urinary urea-creatinine-ratio (p < 0.0001) and between actual protein intake and plasma urea concentrations and the urinary urea-creatinine-ratio (both p < 0.0001) was shown. CONCLUSIONS: The urinary urea-creatinine-ratio, just like plasma urea concentrations, may help to estimate actual protein supply, absorption and oxidation in preterm infants and, additionally, can be determined non-invasively. Further investigations are needed to determine reliable cut-off values of urinary urea concentrations to ensure appropriate protein intake. TRIAL REGISTRATION: Clinicaltrials.gov; NCT01773902 registered 15 January 2013, retrospectively registered.


Asunto(s)
Alimentación con Biberón/métodos , Creatinina/orina , Proteínas en la Dieta/administración & dosificación , Alimentos Fortificados , Recien Nacido Prematuro/sangre , Recien Nacido Prematuro/orina , Recién Nacido de muy Bajo Peso/sangre , Recién Nacido de muy Bajo Peso/orina , Femenino , Edad Gestacional , Humanos , Recién Nacido , Masculino , Leche Humana
10.
Eur J Nutr ; 56(4): 1733-1742, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27164830

RESUMEN

BACKGROUND: Choline, docosahexaenoic acid (DHA), and arachidonic acid (ARA) are essential to fetal development, particularly of the brain. These components are actively enriched in the fetus. Deprivation from placental supply may therefore result in impaired accretion in preterm infants. OBJECTIVE: To determine choline, choline metabolites, DHA, and ARA in human breast milk (BM) of preterm infants compared to BM of term born infants. DESIGN: We collected expressed BM samples from 34 mothers (N = 353; postnatal day 6-85), who had delivered 35 preterm infants undergoing neonatal intensive care (postmenstrual age 30 weeks, range 25.4-32.0), and from mothers after term delivery (N = 9; postnatal day 6-118). Target metabolites were analyzed using tandem mass spectrometry and gas chromatography and reported as medians and 25th/75th percentiles. RESULTS: In BM, choline was mainly present in the form of phosphocholine and glycerophosphocholine, followed by free choline, phosphatidylcholine, sphingomyelin, and lyso-phosphatidylcholine. In preterm infants' BM total choline ranged from 61 to 360 mg/L (median: 158 mg/L) and was decreased compared to term infants' BM (range 142-343 mg/L; median: 258 mg/L; p < 0.01). ARA and DHA comprised 0.81 (range: 0.46-1.60) and 0.43 (0.15-2.42) % of total preterm BM lipids, whereas term BM values were 0.68 (0.52-0.88) and 0.35 (0.18-0.75) %, respectively. Concentrations of all target parameters decreased after birth, and frequently 150 ml/kg/d BM did not meet the estimated fetal accretion rates. CONCLUSIONS: Following preterm delivery, BM choline concentrations are lower, whereas ARA and DHA levels are comparable versus term delivery. Based on these findings we suggest a combined supplementation of preterm infants' BM with choline, ARA and DHA combined to improve the nutritional status of preterm infants. STUDY REGISTRATION: This study was registered at www.clinicaltrials.gov. Identifier: NCT01773902.


Asunto(s)
Colina/administración & dosificación , Ácidos Grasos Insaturados/administración & dosificación , Recien Nacido Prematuro/sangre , Leche Humana/química , Adulto , Ácido Araquidónico/administración & dosificación , Ácido Araquidónico/sangre , Colesterol/sangre , Colina/sangre , Suplementos Dietéticos , Femenino , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro/crecimiento & desarrollo , Masculino , Estado Nutricional , Triglicéridos
11.
Am J Physiol Lung Cell Mol Physiol ; 310(10): L955-63, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944086

RESUMEN

Surfactant, synthesized by type II pneumocytes (PN-II), mainly comprises phosphatidylcholine (PC) and is essential to prevent neonatal respiratory distress. Furthermore, PC is essential to lung tissue growth and maintenance as a membrane component. Recent findings suggest that the lung contributes to systemic lipid homeostasis via PC export through ABC-A1 transporter expression. Hence it is important to consider pharmacological interventions in neonatal lung PC metabolism with respect to such export. Five-day-old rats were treated with carrier (control), intraperitoneal betamethasone, subcutaneous recombinant human keratinocyte growth factor (rhuKGF), or their combination for 48 h. Animals were intraperitoneally injected with 50 mg/kg [D9-methyl]choline chloride 1.5, 3.0, and 6.0 h before death at day 7, and lung lavage fluid (LLF) and tissue were harvested. Endogenous PC, D9-labeled PC species, and their water-soluble precursors (D9-)choline and (D9-)phosphocholine were determined by tandem mass spectrometry. Treatment increased secreted and tissue PC pools but did not change equilibrium composition of PC species in LLF. However, all treatments increased specific surfactant components in tissue. In control rats, peak D9-PC in lavaged lung was reached after 3 h and was decreased at 6 h. Only 13% of this net loss in lavaged lung was found in LLF. Such decrease was not present in lungs treated with betamethasone and/or with rhuKGF. D9-PC loss at 3-6 h and PC synthesis calculated from D9 enrichment of phosphocholine indicated that daily synthesis rate is higher than total pool size. We conclude that lung tissue contributes to systemic PC homeostasis in neonatal rats, which is altered by glucocorticoid and rhuKGF treatment.


Asunto(s)
Betametasona/farmacología , Factor 7 de Crecimiento de Fibroblastos/farmacología , Glucocorticoides/farmacología , Pulmón/metabolismo , Fosfatidilcolinas/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Cinética , Pulmón/efectos de los fármacos , Surfactantes Pulmonares/metabolismo , Ratas Sprague-Dawley
12.
Eur J Nutr ; 55(7): 2265-74, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26363610

RESUMEN

BACKGROUND: Docosahexaenoic (C22:6) and arachidonic acid (C20:4) are long-chain polyunsaturated fatty acids (LC-PUFA), essential to fetal development, and preferentially transported by plasma phospholipids. OBJECTIVE: To characterize fetal and maternal plasma phospholipid changes during gestation, and to investigate whether LC-PUFA phospholipid profiles are associated with bronchopulmonary dysplasia (BPD). DESIGN: Cord plasma and parturient serum from N = 108 pregnancies [24-42 week postmenstrual age (PMA)] were collected. Phosphatidylcholine (PC) and phosphatidylethanolamine (PE) were analyzed with tandem mass spectrometry. PMA-associated changes were quantified, and break point analyses served to describe nonlinear changes during gestation. RESULTS: PC and PE were lower in cord than in parturient samples. In parturients, PC decreased until 33 week PMA, but then re-increased, whereas in cord plasma, concentrations linearly decreased. Fetal PC and PC sub-group values correlated with maternal values. C20:4-PC was twofold higher in cord than in maternal samples throughout gestation. C22:6-PC values, however, exceeded maternal values only beyond 33 week PMA. Consequently, early preterm C20:4-PC-to-C22:6-PC ratio largely exceeded term infant values. In infants born before 28 week PMA, a low C20:4-PC-to-C22:6-PC ratio was associated with BPD severity. CONCLUSIONS: Fetal plasma LC-PUFA-PC composition correlates with maternal values. Fetal C20:4-PC exceeds maternal values throughout gestation, whereas C22:6-PC exceeds maternal values only beyond 33 week PMA, resulting in a low fetal C20:4-PC/C22:6-PC ratio only toward end gestation. A low C20:4-PC/C22:6-PC ratio before 28 week PMA is associated with BPD severity. These data point to a concept of PMA-adjusted ARA and DHA supplementation and, potentially, cord plasma phospholipid analysis for BPD prediction.


Asunto(s)
Displasia Broncopulmonar/sangre , Displasia Broncopulmonar/diagnóstico , Sangre Fetal/química , Feto/metabolismo , Fosfolípidos/sangre , Ácido Araquidónico/sangre , Cromatografía Liquida , Ácidos Docosahexaenoicos/sangre , Ácidos Grasos Insaturados/sangre , Femenino , Humanos , Lactante , Recien Nacido Prematuro/sangre , Modelos Lineales , Fenómenos Fisiologicos Nutricionales Maternos , Fosfatidilcolinas/sangre , Fosfatidiletanolaminas/sangre , Espectrometría de Masas en Tándem
13.
Cell Physiol Biochem ; 35(4): 1437-53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25791258

RESUMEN

BACKGROUND: Liver impairment, ranging from steatosis to cirrhosis, is frequent in cystic fibrosis (CF) patients and is becoming increasingly significant due to their improved life expectancy. One aspect of hepatic alterations is caused by increased fecal loss of the essential nutrient choline, following enterohepatic bile phosphatidylcholine (PC) cycle impairment. Hepatic PC synthesis, both de novo and via phosphatidylethanolamine-N-methyl-transferase (PEMT), is essential for very low-density lipoprotein (VLDL) secretion. VLDL-PC in particular contributes to the organism's supply with polyunsaturated fatty acids (LC-PUFA), namely arachidonic (C20:4) and docosahexaenoic acid (C22:6). Consequently, choline deprivation and altered hepatic PC metabolism may affect plasma PC homeostasis and extrahepatic organ function. OBJECTIVES: To investigate relationships between altered plasma choline and PC homeostasis and markers of lung function and inflammation in CF. To assess alterations in hepatic choline and PC metabolism of CF patients. DESIGN: Quantification of plasma/serum choline and PC species in adult CF patients compared to controls. Correlation of PC with forced expiratory vital capacity (FEV1) and interleukin 6 (IL-6) concentrations. Analysis of choline and PC metabolism in CF compared to controls, using deuterated choline ([D9-methyl]-choline) labeling in vivo. RESULTS: Mean choline and PC concentrations in CF patients were lower than in controls. Choline and PC concentrations as well as fractions of C22:6-PC and C20:4-PC correlated directly with FEV1, but inversely with IL-6. Plasma concentrations of deuterated PC were decreased for both pathways, whereas only in PC synthesized via PEMT precursor enrichment was decreased. CONCLUSION: In CF patients, hepatic and plasma homeostasis of choline and PC correlate with lung function and inflammation. Impaired hepatic PC metabolism, exemplarily shown in three CF patients, provides an explanation for such correlations. Larger studies are required to understand the link between hepatic PC metabolism and overall clinical performance of CF patients, and the perspective of choline substitution of these patients.


Asunto(s)
Fibrosis Quística/patología , Inflamación , Pulmón/fisiología , Fosfatidilcolinas/sangre , Adulto , Ácido Araquidónico/química , Ácido Araquidónico/metabolismo , Betaína/sangre , Colina/sangre , Colina/metabolismo , Fibrosis Quística/metabolismo , Deuterio/química , Ácidos Docosahexaenoicos/química , Ácidos Docosahexaenoicos/metabolismo , Femenino , Volumen Espiratorio Forzado , Humanos , Interleucina-6/sangre , Marcaje Isotópico , Cinética , Hígado/metabolismo , Masculino , Fosfatidilcolinas/metabolismo , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , Adulto Joven
14.
Eur J Nutr ; 54(5): 733-41, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25148882

RESUMEN

BACKGROUND: Choline is essential to human development, particularly of the brain in the form of phosphatidylcholine, sphingomyelin and acetylcholine, for bile and lipoprotein formation, and as a methyl group donator. Choline is actively transported into the fetus, and maternal supply correlates with cognitive outcome. Interruption of placental supply may therefore impair choline homeostasis in preterm infants. OBJECTIVE: Determination of postnatal plasma concentrations of choline and its derivatives betaine and dimethylglycine (DMG) in preterm infants compared to cord and maternal blood matched for postmenstrual age (PMA). DESIGN: We collected plasma of very low-birth-weight infants undergoing neonatal intensive care (n = 162), cord plasma of term and preterm infants (n = 176, 24-42-week PMA), serum of parturients (n = 36), and plasma of healthy premenopausal women (n = 40). Target metabolites were analyzed with tandem mass spectrometry and reported as median (25th/75th percentiles). RESULTS: Cord plasma choline concentration was 41.4 (31.8-51.2) µmol/L and inversely correlated with PMA. In term but not in preterm infants, cord plasma choline was lower in girls than in boys. Prenatal glucocorticoid treatment did not affect choline levels in cord plasma, whereas betaine was decreased and DMG increased. In parturients and non-pregnant women, choline concentrations were 14.1 (10.3-16.9) and 8.8 (5.7-11.2) µmol/L, respectively, whereas betaine was lowest in parturients. After delivery, preterm infant plasma choline decreased to 20.8 (16.0-27.6) µmol/L within 48 h. Betaine and DMG correlated with plasma choline in all groups. CONCLUSIONS: In preterm infants, plasma choline decreases to 50 % of cord plasma concentrations, reflecting choline undernourishment and postnatal metabolic adaptation, and potentially contributing to impaired outcome.


Asunto(s)
Colina/sangre , Sangre Fetal/química , Recien Nacido Prematuro/sangre , Adolescente , Adulto , Betaína/administración & dosificación , Cromatografía Liquida , Nutrición Enteral , Femenino , Feto/metabolismo , Glucocorticoides/administración & dosificación , Humanos , Recién Nacido de muy Bajo Peso/sangre , Unidades de Cuidado Intensivo Neonatal , Masculino , Persona de Mediana Edad , Nutrición Parenteral , Embarazo , Premenopausia/sangre , Estudios Prospectivos , Sarcosina/administración & dosificación , Sarcosina/análogos & derivados , Espectrometría de Masas en Tándem , Adulto Joven
15.
Eur J Nutr ; 53(7): 1533-47, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24464176

RESUMEN

BACKGROUND: During fetal development, docosahexaenoic (DHA) and arachidonic acid (ARA) are particularly enriched in brain phospholipids. After preterm delivery, fetal enrichment of DHA and ARA via placental transfer is replaced by enteral and parenteral nutrition, which is rich in linoleic acid (LA) instead. Specific DHA and ARA enrichment of lipoproteins is reflected by plasma phosphatidylcholine (PC) species, whereas plasma phosphatidylethanolamine (PE) composition reflects hepatic stores. OBJECTIVE: We profiled PC and PE species in preterm infant plasma, compared with cord and maternal blood, to assess whether current feeding practice meets fetal conditions in these patients. DESIGN: Preterm infant plasma (N = 171, 23-35 w postmenstrual age (PMA), postnatal day 1-103), cord plasma (N = 194) and maternal serum (N = 121) (both 24-41 w PMA) were collected. After lipid extraction, PC and PE molecular species were analyzed using tandem mass spectrometry. RESULTS: Phospholipid concentrations were higher in preterm infant than in cord plasma after correction for PMA. This was mainly due to postnatal increases in LA-containing PC and PE, resulting in decreased fractions of their DHA- and ARA-containing counterparts. These changes in preterm infant plasma phospholipids occurred during the time of transition to full enteral feeds (day 0-10 after delivery). Thereafter, the fraction of ARA-containing phospholipids further decreased, whereas that of DHA slowly reincreased but remained at a level 50% of that of PMA-matched cord blood. CONCLUSIONS: The postnatal increase in LA-PC in preterm infant plasma results in decreased fractions of DHA-PC and ARA-PC. These changes are also reflected by PE molecular composition as an indicator of altered hepatic fatty acid homeostasis. They are presumably caused by inadequately high LA, and low ARA and DHA supply, at a stage of development when ARA-PC and DHA-PC should be high, probably reducing the availability of DHA and ARA to the developing brain and contributing to impaired neurodevelopment of preterm infants.


Asunto(s)
Ácido Araquidónico/sangre , Ácidos Docosahexaenoicos/sangre , Recien Nacido Prematuro/sangre , Fosfolípidos/sangre , Femenino , Sangre Fetal/química , Homeostasis , Humanos , Lactante , Recién Nacido , Ácido Linoleico/sangre , Masculino , Fosfatidilcolinas/sangre , Fosfatidiletanolaminas/sangre , Estudios Prospectivos
16.
Nutrients ; 16(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38931230

RESUMEN

Choline is an essential nutrient, with high requirements during fetal and postnatal growth. Tissue concentrations of total choline are tightly regulated, requiring an increase in its pool size proportional to growth. Phosphatidylcholine and sphingomyelin, containing a choline headgroup, are constitutive membrane phospholipids, accounting for >85% of total choline, indicating that choline requirements are particularly high during growth. Daily phosphatidylcholine secretion via bile for lipid digestion and very low-density lipoproteins for plasma transport of arachidonic and docosahexaenoic acid to other organs exceed 50% of its hepatic pool. Moreover, phosphatidylcholine is required for converting pro-apoptotic ceramides to sphingomyelin, while choline is the source of betaine as a methyl donor for creatine synthesis, DNA methylation/repair and kidney function. Interrupted choline supply, as during current total parenteral nutrition (TPN), causes a rapid drop in plasma choline concentration and accumulating deficit. The American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) defined choline as critical to all infants requiring TPN, claiming its inclusion in parenteral feeding regimes. We performed a systematic literature search in Pubmed with the terms "choline" and "parenteral nutrition", resulting in 47 relevant publications. Their results, together with cross-references, are discussed. While studies on parenteral choline administration in neonates and older children are lacking, preclinical and observational studies, as well as small randomized controlled trials in adults, suggest choline deficiency as a major contributor to acute and chronic TPN-associated liver disease, and the safety and efficacy of parenteral choline administration for its prevention. Hence, we call for choline formulations suitable to be added to TPN solutions and clinical trials to study their efficacy, particularly in growing children including preterm infants.


Asunto(s)
Colina , Suplementos Dietéticos , Nutrición Parenteral , Colina/administración & dosificación , Humanos , Recién Nacido , Lactante , Deficiencia de Colina , Niño , Nutrición Parenteral Total , Preescolar
17.
Histochem Cell Biol ; 139(3): 461-72, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23100171

RESUMEN

Development of preterm infant lungs is frequently impaired resulting in bronchopulmoary dysplasia (BPD). BPD results from interruption of physiologic anabolic intrauterine conditions, the inflammatory basis and therapeutic consequences of premature delivery, including increased oxygen supply for air breathing. The latter requires surfactant, produced by alveolar type II (AT II) cells to lower surface tension at the pulmonary air:liquid interface. Its main components are specific phosphatidylcholine (PC) species including dipalmitoyl-PC, anionic phospholipids and surfactant proteins. Local antioxidative enzymes are essential to cope with the pro-inflammatory side effects of normal alveolar oxygen pressures. However, respiratory insufficiency frequently requires increased oxygen supply. To cope with the injurious effects of hyperoxia to epithelia, recombinant human keratinocyte growth factor (rhKGF) was proposed as a surfactant stimulating, non-catabolic and epithelial-protective therapeutic. The aim of the present study was to examine the qualification of rhKGF to improve expression parameters of lung maturity in newborn rats under hyperoxic conditions (85% O(2) for 7 days). In response to rhKGF proliferating cell nuclear antigen mRNA, as a feature of stimulated proliferation, was elevated. Similarly, the expressions of ATP-binding cassette protein A3 gene, a differentiation marker of AT II cells and of peroxiredoxin 6, thioredoxin and thioredoxin reductase, three genes involved in oxygen radical protection were increased. Furthermore, mRNA levels of acyl-coA:lysophosphatidylcholine acyltransferase 1, catalyzing dipalmitoyl-PC synthesis by acyl remodeling, and adipose triglyceride lipase, considered as responsible for fatty acid supply for surfactant PC synthesis, were elevated. These results, together with a considerable body of other confirmative evidence, suggest that rhKGF should be developed into a therapeutic option to treat preterm infants at risk for impaired lung development.


Asunto(s)
Antioxidantes/metabolismo , Factor 7 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hiperoxia/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Tensoactivos/metabolismo , Animales , Animales Recién Nacidos , Perfilación de la Expresión Génica , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Eur J Nutr ; 52(3): 1269-78, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22961562

RESUMEN

BACKGROUND: Choline forms the head group of phosphatidylcholines, comprising 40-50 % of cellular membranes and 70-95 % of phospholipids in surfactant, bile, and lipoproteins. Moreover, choline serves as the precursor of acetylcholine and is important for brain differentiation and function. While accepted as essential for fetal and neonatal development, its role in preterm infant nutrition has not yet gained much attention. METHODS: The adequate intake of choline of preterm infants was estimated from international recommendations for infants, children, and adults. Choline intake relative to other nutrients was determined retrospectively in all inborn infants below 1,000 g (extremely low birth weight) or below 28 weeks gestational age, admitted to our department in 2006 and 2007 (N = 93). RESULTS: Estimation of adequate intake showed that children with 290 g body weight need more choline than those with 1,200 g (31.4 and 25.2 mg/kg/day, respectively). Day-by-day variability was high for all nutrient intakes including choline. In contrast to the continuous intrauterine choline delivery, median supply reached a plateau at d11 (21.7 mg/kg/day; 25th/75th percentile: 19.6; 23.9). Individual choline supply at d0-d1 and d2-d3 was <10 mg/kg/day in 100 and 69 % of infants, respectively. Furthermore, intakes <10 mg/kg/day were frequently observed beyond day 11. Median adequate intakes (27.4 mg/kg/day at 735 g body weight) were achieved in <2 %. CONCLUSIONS: Nutritional intake of choline in this cohort of preterm infants was frequently less than the estimated adequate intake, with particular shortage until postnatal d10. Because choline is important for brain development, future studies are needed to investigate the effects of adequate nutritional choline intake on long-term neurodevelopment in VLBW infants.


Asunto(s)
Desarrollo Infantil , Deficiencia de Colina/etiología , Colina/administración & dosificación , Dieta/efectos adversos , Fenómenos Fisiológicos Nutricionales del Lactante , Enfermedades del Prematuro/etiología , Deficiencia de Colina/epidemiología , Deficiencia de Colina/fisiopatología , Estudios de Cohortes , Femenino , Alemania/epidemiología , Guías como Asunto , Hospitales Universitarios , Humanos , Incidencia , Recien Nacido con Peso al Nacer Extremadamente Bajo , Recien Nacido Extremadamente Prematuro , Recién Nacido , Enfermedades del Prematuro/epidemiología , Enfermedades del Prematuro/fisiopatología , Unidades de Cuidado Intensivo Neonatal , Masculino , Necesidades Nutricionales , Garantía de la Calidad de Atención de Salud , Estudios Retrospectivos
19.
BMC Pediatr ; 13: 178, 2013 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-24180239

RESUMEN

BACKGROUND: Fortified human milk may not meet all nutritional needs of very preterm infants. Early transition from complementary parenteral nutrition to full enteral feeds might further impair in-hospital growth. We aimed to investigate the impact of the cumulative intake of fortified human milk on early postnatal growth in a cohort of very low birth weight infants after early transition to full enteral feeds. METHODS: Retrospective single-centre observational study. Data are presented as median (interquartile range). RESULTS: N = 206 very preterm infants were analysed (gestational age at birth 27.6 (25.6-29.6) weeks, birth weight 915 (668-1170) g). Full enteral feeds were established at postnatal day 8 (6-10) and adequate postnatal growth was achieved (difference in standard deviation score for weight from birth to discharge -0.105(-0.603 - -0.323)). Standard deviation score for weight from birth to day 28 decreased more in infants with a cumulative human milk intake > 75% of all enteral feeds (-0.64(-1.08 - -0.34)) compared to those with < 25% human milk intake (-0.41(-0.7 - -0.17); p = 0.017). At discharge, a trend towards poorer weight gain with higher proportions of human milk intake persisted. In contrast, we observed no significant difference for head circumference growth. CONCLUSIONS: Our current standardized fortification of human milk may not adequately support early postnatal growth.


Asunto(s)
Alimentos Fortificados , Alimentos Infantiles , Recien Nacido Prematuro/crecimiento & desarrollo , Recién Nacido de muy Bajo Peso/crecimiento & desarrollo , Leche Humana , Cefalometría , Niño Hospitalizado , Estudios de Cohortes , Ingestión de Energía , Nutrición Enteral , Estudios de Seguimiento , Alemania , Edad Gestacional , Cabeza/crecimiento & desarrollo , Humanos , Fórmulas Infantiles , Recién Nacido , Necesidades Nutricionales , Resultado del Tratamiento , Aumento de Peso
20.
Nutrients ; 15(22)2023 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-38004152

RESUMEN

Choline is essential for cell membrane formation and methyl transfer reactions, impacting parenchymal and neurological development. It is therefore enriched via placental transfer, and fetal plasma concentrations are high. In spite of the greater needs of very low birth weight infants (VLBWI), choline content of breast milk after preterm delivery is lower (median (p25-75): 158 mg/L (61-360 mg/L) compared to term delivery (258 mg/L (142-343 mg/L)). Even preterm formula or fortified breast milk currently provide insufficient choline to achieve physiological plasma concentrations. This secondary analysis of a randomized controlled trial comparing growth of VLBWI with different levels of enteral protein supply aimed to investigate whether increased enteral choline intake results in increased plasma choline, betaine and phosphatidylcholine concentrations. We measured total choline content of breast milk from 33 mothers of 34 VLBWI. Enteral choline intake from administered breast milk, formula and fortifier was related to the respective plasma choline, betaine and phosphatidylcholine concentrations. Plasma choline and betaine levels in VLBWI correlated directly with enteral choline intake, but administered choline was insufficient to achieve physiological (fetus-like) concentrations. Hence, optimizing maternal choline status, and the choline content of milk and fortifiers, is suggested to increase plasma concentrations of choline, ameliorate the choline deficit and improve growth and long-term development of VLBWI.


Asunto(s)
Betaína , Enfermedades del Prematuro , Lactante , Recién Nacido , Humanos , Femenino , Embarazo , Recien Nacido Prematuro , Colina , Placenta , Recién Nacido de muy Bajo Peso , Leche Humana , Lecitinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA