Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Nutrients ; 16(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38931192

ABSTRACT

BACKGROUND: Brown seaweed is promising for the treatment of type 2 diabetes mellitus (T2DM). Its bioactive constituents can positively affect plasma glucose homeostasis in healthy humans. We investigated the effect of the brown seaweeds Sargassum (S.) fusiforme and Fucus (F.) vesiculosus in their natural form on glucose regulation in patients with T2DM. METHODS: We conducted a randomized, double-blind, placebo-controlled pilot trial. Thirty-six participants with T2DM received, on a daily basis, either 5 g of dried S. fusiforme, 5 g of dried F. vesiculosus, or 0.5 g of dried Porphyra (control) for 5 weeks, alongside regular treatment. The primary outcome was the between-group difference in the change in weekly average blood glucose levels (continuous glucose monitoring). The secondary outcomes were the changes in anthropometrics, plasma lipid levels, and dietary intake. The data were analyzed using a linear mixed-effects model. RESULTS: The change in weekly average glucose levels was 8.2 ± 2.1 to 9.0 ± 0.7 mmol/L (p = 0.2) in the S. fusiforme group (n = 12) and 10.1 ± 3.3 to 9.2 ± 0.7 mmol/L (p = 0.9) in the F. vesiculosus group (n = 10). The between-group difference was non-significant. Similarly, no between-group differences were observed for the changes in the secondary outcomes. DISCUSSION: A daily intake of 5 g of fresh, dried S. fusiforme or F. vesiculosus alongside regular treatment had no differential effect on weekly average blood glucose levels in T2DM.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 2 , Fucus , Sargassum , Humans , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Double-Blind Method , Blood Glucose/metabolism , Blood Glucose/drug effects , Male , Female , Middle Aged , Fucus/chemistry , Pilot Projects , Overweight/blood , Feasibility Studies , Aged , Adult , Seaweed , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Edible Seaweeds
2.
Nutrients ; 16(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38892548

ABSTRACT

We previously demonstrated that diet supplementation with seaweed Sargassum fusiforme (S. fusiforme) prevented AD-related pathology in a mouse model of Alzheimer's Disease (AD). Here, we tested a lipid extract of seaweed Himanthalia elongata (H. elongata) and a supercritical fluid (SCF) extract of S. fusiforme that is free of excess inorganic arsenic. Diet supplementation with H. elongata extract prevented cognitive deterioration in APPswePS1ΔE9 mice. Similar trends were observed for the S. fusiforme SCF extract. The cerebral amyloid-ß plaque load remained unaffected. However, IHC analysis revealed that both extracts lowered glial markers in the brains of APPswePS1ΔE9 mice. While cerebellar cholesterol concentrations remained unaffected, both extracts increased desmosterol, an endogenous LXR agonist with anti-inflammatory properties. Both extracts increased cholesterol efflux, and particularly, H. elongata extract decreased the production of pro-inflammatory cytokines in LPS-stimulated THP-1-derived macrophages. Additionally, our findings suggest a reduction of AD-associated phosphorylated tau and promotion of early oligodendrocyte differentiation by H. elongata. RNA sequencing on the hippocampus of one-week-treated APPswePS1ΔE9 mice revealed effects of H. elongata on, amongst others, acetylcholine and synaptogenesis signaling pathways. In conclusion, extracts of H. elongata and S. fusiforme show potential to reduce AD-related pathology in APPswePS1ΔE9 mice. Increasing desmosterol concentrations may contribute to these effects by dampening neuroinflammation.


Subject(s)
Alzheimer Disease , Dietary Supplements , Disease Models, Animal , Seaweed , Animals , Alzheimer Disease/drug therapy , Seaweed/chemistry , Mice , Hippocampus/drug effects , Hippocampus/metabolism , Plant Extracts/pharmacology , Mice, Transgenic , Sargassum/chemistry , Humans , Plaque, Amyloid , Cholesterol/metabolism , Cholesterol/blood , Male , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , tau Proteins/metabolism
3.
Nutrients ; 15(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37447330

ABSTRACT

The nuclear liver X receptors (LXRα/ß) and peroxisome proliferator-activated receptors (PPARα/γ) are involved in the regulation of multiple biological processes, including lipid metabolism and inflammation. The activation of these receptors has been found to have neuroprotective effects, making them interesting therapeutic targets for neurodegenerative disorders such as Alzheimer's Disease (AD). The Asian brown seaweed Sargassum fusiforme contains both LXR-activating (oxy)phytosterols and PPAR-activating fatty acids. We have previously shown that dietary supplementation with lipid extracts of Sargassum fusiforme prevents disease progression in a mouse model of AD, without inducing adverse effects associated with synthetic pan-LXR agonists. We now determined the LXRα/ß- and PPARα/γ-activating capacity of lipid extracts of six European brown seaweed species (Alaria esculenta, Ascophyllum nodosum, Fucus vesiculosus, Himanthalia elongata, Saccharina latissima, and Sargassum muticum) and the Asian seaweed Sargassum fusiforme using a dual luciferase reporter assay. We analyzed the sterol and fatty acid profiles of the extracts by GC-MS and UPLC MS/MS, respectively, and determined their effects on the expression of LXR and PPAR target genes in several cell lines using quantitative PCR. All extracts were found to activate LXRs, with the Himanthalia elongata extract showing the most pronounced efficacy, comparable to Sargassum fusiforme, for LXR activation and transcriptional regulation of LXR-target genes. Extracts of Alaria esculenta, Fucus vesiculosus, and Saccharina latissima showed the highest capacity to activate PPARα, while extracts of Alaria esculenta, Ascophyllum nodosum, Fucus vesiculosus, and Sargassum muticum showed the highest capacity to activate PPARγ, comparable to Sargassum fusiforme extract. In CCF-STTG1 astrocytoma cells, all extracts induced expression of cholesterol efflux genes (ABCG1, ABCA1, and APOE) and suppressed expression of cholesterol and fatty acid synthesis genes (DHCR7, DHCR24, HMGCR and SREBF2, and SREBF1, ACACA, SCD1 and FASN, respectively). Our data show that lipophilic fractions of European brown seaweeds activate LXRs and PPARs and thereby modulate lipid metabolism. These results support the potential of brown seaweeds in the prevention and/or treatment of neurodegenerative diseases and possibly cardiometabolic and inflammatory diseases via concurrent activation of LXRs and PPARs.


Subject(s)
Alzheimer Disease , Seaweed , Mice , Animals , Liver X Receptors/genetics , Liver X Receptors/metabolism , Alzheimer Disease/drug therapy , PPAR alpha/genetics , Tandem Mass Spectrometry , Receptors, Cytoplasmic and Nuclear/genetics , Cholesterol/metabolism , Fatty Acids/metabolism
4.
Int J Mol Sci ; 24(2)2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36674804

ABSTRACT

The nuclear receptors-liver X receptors (LXR α and ß) are potential therapeutic targets in cardiovascular and neurodegenerative diseases because of their key role in the regulation of lipid homeostasis and inflammatory processes. Specific oxy(phyto)sterols differentially modulate the transcriptional activity of LXRs providing opportunities to develop compounds with improved therapeutic characteristics. We isolated oxyphytosterols from Sargassum fusiforme and synthesized sidechain oxidized sterol derivatives. Five 24-oxidized sterols demonstrated a high potency for LXRα/ß activation in luciferase reporter assays and induction of LXR-target genes APOE, ABCA1 and ABCG1 involved in cellular cholesterol turnover in cultured cells: methyl 3ß-hydroxychol-5-en-24-oate (S1), methyl (3ß)-3-aldehydeoxychol-5-en-24-oate (S2), 24-ketocholesterol (S6), (3ß,22E)-3-hydroxycholesta-5,22-dien-24-one (N10) and fucosterol-24,28 epoxide (N12). These compounds induced SREBF1 but not SREBP1c-mediated lipogenic genes such as SCD1, ACACA and FASN in HepG2 cells or astrocytoma cells. Moreover, S2 and S6 enhanced cholesterol efflux from HepG2 cells. All five oxysterols induced production of the endogenous LXR agonists 24(S)-hydroxycholesterol by upregulating the CYP46A1, encoding the enzyme converting cholesterol into 24(S)-hydroxycholesterol; S1 and S6 may also act via the upregulation of desmosterol production. Thus, we identified five novel LXR-activating 24-oxidized sterols with a potential for therapeutic applications in neurodegenerative and cardiovascular diseases.


Subject(s)
Neurodegenerative Diseases , Phytosterols , Humans , Liver X Receptors , Sterols/pharmacology , Orphan Nuclear Receptors/genetics , Hydroxycholesterols , Neurodegenerative Diseases/drug therapy , Cholesterol
5.
Mar Drugs ; 19(4)2021 Mar 27.
Article in English | MEDLINE | ID: mdl-33801706

ABSTRACT

We recently found that dietary supplementation with the seaweed Sargassum fusiforme, containing the preferential LXRß-agonist 24(S)-saringosterol, prevented memory decline and reduced amyloid-ß (Aß) deposition in an Alzheimer's disease (AD) mouse model without inducing hepatic steatosis. Here, we examined the effects of 24(S)-saringosterol as a food additive on cognition and neuropathology in AD mice. Six-month-old male APPswePS1ΔE9 mice and wildtype C57BL/6J littermates received 24(S)-saringosterol (0.5 mg/25 g body weight/day) (APPswePS1ΔE9 n = 20; C57BL/6J n = 19) or vehicle (APPswePS1ΔE9 n = 17; C57BL/6J n = 19) for 10 weeks. Cognition was assessed using object recognition and object location tasks. Sterols were analyzed by gas chromatography/mass spectrometry, Aß and inflammatory markers by immunohistochemistry, and gene expression by quantitative real-time PCR. Hepatic lipids were quantified after Oil-Red-O staining. Administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice without affecting the Aß plaque load. Moreover, 24(S)-saringosterol prevented the increase in the inflammatory marker Iba1 in the cortex of APPswePS1ΔE9 mice (p < 0.001). Furthermore, 24(S)-saringosterol did not affect the expression of lipid metabolism-related LXR-response genes in the hippocampus nor the hepatic neutral lipid content. Thus, administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice independent of effects on Aß load and without adverse effects on liver fat content. The anti-inflammatory effects of 24(S)-saringosterol may contribute to the prevention of cognitive decline.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents/pharmacology , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cognition/drug effects , Nootropic Agents/pharmacology , Stigmasterol/analogs & derivatives , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Animals , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Recognition, Psychology/drug effects , Stigmasterol/pharmacology
6.
Diabetologia ; 61(11): 2386-2397, 2018 11.
Article in English | MEDLINE | ID: mdl-30145664

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to evaluate the effect of sitagliptin on glucose tolerance, plasma lipids, energy expenditure and metabolism of brown adipose tissue (BAT), white adipose tissue (WAT) and skeletal muscle in overweight individuals with prediabetes (impaired glucose tolerance and/or impaired fasting glucose). METHODS: We performed a randomised, double-blinded, placebo-controlled trial in 30 overweight, Europid men (age 45.9 ± 6.2 years; BMI 28.8 ± 2.3 kg/m2) with prediabetes in the Leiden University Medical Center and the Alrijne Hospital between March 2015 and September 2016. Participants were initially randomly allocated to receive sitagliptin (100 mg/day) (n = 15) or placebo (n = 15) for 12 weeks, using a randomisation list that was set up by an unblinded pharmacist. All people involved in the study as well as participants were blinded to group assignment. Two participants withdrew from the study prior to completion (both in the sitagliptin group) and were subsequently replaced with two new participants that were allocated to the same treatment. Before and after treatment, fasting venous blood samples and skeletal muscle biopsies were obtained, OGTT was performed and body composition, resting energy expenditure and [18F] fluorodeoxyglucose ([18F]FDG) uptake by metabolic tissues were assessed. The primary study endpoint was the effect of sitagliptin on BAT volume and activity. RESULTS: One participant from the sitagliptin group was excluded from analysis, due to a distribution error, leaving 29 participants for further analysis. Sitagliptin, but not placebo, lowered glucose excursion (-40%; p < 0.003) during OGTT, accompanied by an improved insulinogenic index (+38%; p < 0.003) and oral disposition index (+44%; p < 0.003). In addition, sitagliptin lowered serum concentrations of triacylglycerol (-29%) and very large (-46%), large (-35%) and medium-sized (-24%) VLDL particles (all p < 0.05). Body weight, body composition and energy expenditure did not change. In skeletal muscle, sitagliptin increased mRNA expression of PGC1ß (also known as PPARGC1B) (+117%; p < 0.05), a main controller of mitochondrial oxidative energy metabolism. Although the primary endpoint of change in BAT volume and activity was not met, sitagliptin increased [18F] FDG uptake in subcutaneous WAT (sWAT; +53%; p < 0.05). Reported side effects were mild and transient and not necessarily related to the treatment. CONCLUSIONS/INTERPRETATION: Twelve weeks of sitagliptin in overweight, Europid men with prediabetes improves glucose tolerance and lipid metabolism, as related to increased [18F] FDG uptake by sWAT, rather than BAT, and upregulation of the mitochondrial gene PGC1ß in skeletal muscle. Studies on the effect of sitagliptin on preventing or delaying the progression of prediabetes into type 2 diabetes are warranted. TRIAL REGISTRATION: ClinicalTrials.gov NCT02294084. FUNDING: This study was funded by Merck Sharp & Dohme Corp, Dutch Heart Foundation, Dutch Diabetes Research Foundation, Ministry of Economic Affairs and the University of Granada.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Overweight/drug therapy , Overweight/metabolism , Prediabetic State/drug therapy , Sitagliptin Phosphate/therapeutic use , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adult , Blood Glucose/drug effects , Body Weight/drug effects , Carrier Proteins/genetics , Double-Blind Method , Energy Metabolism/drug effects , Humans , Male , Middle Aged , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Prediabetic State/metabolism , RNA-Binding Proteins
7.
J Endocrinol ; 238(2): 91-106, 2018 08.
Article in English | MEDLINE | ID: mdl-29743343

ABSTRACT

Cold exposure of mice is a common method to stimulate brown adipose tissue (BAT) activity and induce browning of white adipose tissue (WAT) that has beneficial effects on whole-body lipid metabolism, including reduced plasma triglyceride (TG) concentrations. The liver is a key regulatory organ in lipid metabolism as it can take up as well as oxidize fatty acids. The liver can also synthesize, store and secrete TGs in VLDL particles. The effects of cold exposure on murine hepatic lipid metabolism have not been addressed. Here, we report the effects of 24-h exposure to 4°C on parameters of hepatic lipid metabolism of male C57BL/6J mice. Cold exposure increased hepatic TG concentrations by 2-fold (P < 0.05) but reduced hepatic lipogenic gene expression. Hepatic expression of genes encoding proteins involved in cholesterol synthesis and uptake such as the LDL receptor (LDLR) was significantly increased upon cold exposure. Hepatic expression of Cyp7a1 encoding the rate-limiting enzyme in the classical bile acid (BA) synthesis pathway was increased by 4.3-fold (P < 0.05). Hepatic BA concentrations and fecal BA excretion were increased by 2.8- and 1.3-fold, respectively (P < 0.05 for both). VLDL-TG secretion was reduced by approximately 50% after 24 h of cold exposure (P < 0.05). In conclusion, cold exposure has various, likely intertwined effects on the liver that should be taken into account when studying the effects of cold exposure on whole-body metabolism.


Subject(s)
Cold Temperature , Liver/metabolism , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Animals , Cell Transdifferentiation/genetics , Down-Regulation/genetics , Gene Expression Regulation , Glycogen/metabolism , Lipid Metabolism/physiology , Lipogenesis/genetics , Lipoproteins, VLDL/blood , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Thermogenesis/physiology , Triglycerides/blood
8.
Am J Clin Nutr ; 101(6): 1155-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25926506

ABSTRACT

BACKGROUND: Phenylalanine, which is an essential aromatic amino acid, is either used for protein synthesis or irreversibly hydroxylated to tyrosine. The provision of optimal amounts of dietary phenylalanine is not only important for growth and development but might also influence catecholamine synthesis and release rates. The current recommended aromatic amino acid requirement for infants aged 0-6 mo is based on the amino acid content of human milk. OBJECTIVE: We quantified the requirements for phenylalanine in the presence of excess tyrosine (166 or 177 mg/kg per day for term and preterm infants, respectively) for term and preterm neonates by using the indicator amino acid oxidation method with l-[1-(13)C]lysine 2HCl as an indicator. Hence, we determined the minimum obligatory phenylalanine requirement. DESIGN: Fully enterally fed term and preterm infants received randomly graded amounts of phenylalanine (5-177 mg/kg per day) as part of an elemental formula. Data are expressed as means ± SDs. RESULTS: Twenty term (birth weight: 3.19 ± 0.34 kg; gestational age: 38.9 ± 1 wk) and 16 preterm (birth weight: 1.75 ± 0.17 kg; gestational age: 32.5 ± 0.6 wk) Asian infants participated at a postnatal age of 17 ± 8 d. In total, 44 studies were performed. The minimum obligatory phenylalanine requirement was 58 mg/kg per day (95% CI: 38-78 mg/kg per day) and 80 mg/kg per day (95% CI: 40-119 mg/kg per day) for term and preterm infants, respectively. CONCLUSION: The determined mean phenylalanine-requirement estimates are lower than the contents of term and preterm formulas currently on the market. This trial was registered at www.trialregister.nl as NTR1610.


Subject(s)
Enteral Nutrition/methods , Infant Nutritional Physiological Phenomena , Nutritional Requirements , Phenylalanine/administration & dosage , Cross-Over Studies , Female , Gestational Age , Humans , Infant , Infant, Newborn , Infant, Premature/growth & development , Linear Models , Male , Milk, Human/chemistry , Term Birth , Tyrosine/metabolism
9.
J Pediatr Gastroenterol Nutr ; 61(3): 373-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25844708

ABSTRACT

OBJECTIVE: Threonine is one of the essential amino acids. Its major fate is incorporation into intestinal mucosal proteins and synthesis of secretory glycoproteins. Therefore, it has an important function in the neonatal gut barrier integrity. The objective was to quantify the threonine requirement in fully enterally fed term neonates by means of the indicator amino acid oxidation (IAAO) method, using L-[1-C]phenylalanine as indicator. METHODS: After a 24-hour test diet adaptation, containing randomly assigned amounts of threonine (range 5-182 mg · kg · day), the participating neonates received a primed continuous infusion of [C]bicarbonate and L-[1-C]phenylalanine. At baseline and during the plateau phase of both infusions, breath samples were obtained for CO2. The fractional L-[1-C]phenylalanine oxidation (FCO2) was estimated and plotted against the threonine intakes. Biphasic linear regression crossover analysis was used to calculate the breakpoint of the FCO2, representing the mean threonine requirement. Data are presented as mean ±â€ŠSD. RESULTS: Thirty-two term neonates (gestational age 39 ±â€Š1 weeks, birth weight 3.3 ±â€Š0.3 kg, mean postnatal age 10 ±â€Š4 days) were studied. The mean threonine requirement was estimated to be 68 mg · kg · day with an upper and lower 95% confidence interval of 104 and 32 mg · kg · day, respectively (r = 0.37). CONCLUSIONS: The determined threonine requirement is extremely close to the existing requirement recommendations (∼90% of the present World Health Organization requirement guidelines). Infant formula preparations presently on the market, however, contain up to twice as much threonine as recommended. The threonine intake in formula-fed infants may therefore be reduced considerably.


Subject(s)
Enteral Nutrition , Infant Nutritional Physiological Phenomena/standards , Nutritional Requirements , Threonine/analysis , Bicarbonates/metabolism , Breath Tests , Female , Humans , Infant, Newborn , Linear Models , Male , Oxidation-Reduction , Phenylalanine/metabolism , Threonine/administration & dosage
10.
J Pediatr Gastroenterol Nutr ; 59(3): 374-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24824360

ABSTRACT

OBJECTIVES: Tryptophan not only is an amino acid essential to protein synthesis but also serves as a precursor in 2 important metabolic pathways: the serotonin and the kynurenine pathways. Tryptophan is related to sleeping patterns. The objective of the present study was to determine the tryptophan requirement of term infants using the indicator amino acid oxidation (IAAO) method with L-[1-C]phenylalanine as the indicator. METHODS: Enterally fed infants were randomly assigned to tryptophan intakes ranging from 0.5 to 73 mg ·â€Škg ·â€Šday as part of an elemental diet. After 1-day adaptation to the test diet, [C]bicarbonate and L-[1-C]phenylalanine tracers were given enterally. Breath samples were collected at baseline and during isotopic plateaus. The mean tryptophan requirement was determined by using the biphasic linear regression crossover analysis on the fraction of CO2 recovery from L-[1-C]phenylalanine oxidation (FCO2). Data are presented as mean ±â€Šstandard deviation. RESULTS: A total of 30 term neonates (gestational age 39 ±â€Š1 weeks) were studied at 9 ±â€Š4 days. FCO2 decreased until a tryptophan intake of 15 mg ·â€Škg ·â€Šday; additional increases in tryptophan intake did not affect FCO2. Mean requirement was determined to be 15 mg ·â€Škg ·â€Šday. CONCLUSIONS: The mean tryptophan requirement for elemental formula-fed term infants is 15 mg ·â€Škg ·â€Šday. This requirement is lower than the present recommended intake of 29 mg ·â€Škg ·â€Šday, which is based on the average intake of a breastfed infant.


Subject(s)
Enteral Nutrition , Nutritional Requirements , Tryptophan/administration & dosage , Bicarbonates/administration & dosage , Breath Tests , Carbon Radioisotopes , Female , Humans , Infant Formula/chemistry , Infant, Newborn , Male , Oxidation-Reduction , Phenylalanine/administration & dosage , Phenylalanine/metabolism , Term Birth
11.
Am J Clin Nutr ; 99(1): 62-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24284437

ABSTRACT

BACKGROUND: Knowledge of essential amino acid requirements in infants is important because excessive intake of protein can lead to increased long-term morbidity such as obesity. A deficient intake may lead to suboptimal growth and impaired neurodevelopment. The current recommended branched-chain amino acid requirements in infants aged 0-1 mo are based on the amino acid content of human milk. OBJECTIVE: We quantified the requirements for isoleucine, leucine, and valine for term neonates by using the indicator amino acid oxidation method with [1-(13)C]phenylalanine as the indicator. DESIGN: Fully enterally fed term infants received randomly graded amounts of isoleucine (5-216 mg · kg(-1) · d(-1)), leucine (5-370 mg · kg(-1) · d(-1)), or valine (5-236 mg · kg(-1) · d(-1)) as part of an elemental formula. Data are expressed as means ± SDs. RESULTS: Eighty-three Asian, term neonates (mean ± SD birth weight: 3.3 ± 0.4 kg; gestational age: 39.4 ± 1.3 wk) were studied at a postnatal age of 13 ± 5 d. Mean requirements for isoleucine, leucine, and valine (measured in boys only) were 105 mg · kg(-1) · d(-1) (r(2) = 0.61, P < 0.001), 140 mg · kg(-1) · d(-1) (r(2) = 0.26, P < 0.01), and 110 mg · kg(-1) · d(-1) (r(2) = 0.35, P = 0.001), respectively. CONCLUSIONS: Current human milk-based recommendations for isoleucine and valine in term infants aged 0-1 mo are correct. However, the current recommendation for leucine (166 mg · kg(-1) · d(-1)) is higher than the mean requirement of 140 mg · kg(-1) · d(-1) that we determined in this study. This trial was registered at www.trialregister.nl as NTR1610.


Subject(s)
Amino Acids, Branched-Chain/administration & dosage , Infant Nutritional Physiological Phenomena , Nutritional Requirements , Cross-Over Studies , Energy Intake , Female , Gestational Age , Humans , Infant, Newborn , Isoleucine/metabolism , Leucine/metabolism , Linear Models , Male , Oxidation-Reduction , Recommended Dietary Allowances , Valine/metabolism
12.
Pediatr Res ; 73(5): 679-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23407117

ABSTRACT

BACKGROUND: We determined the effect of adaptation to the study diet on oxidation of the indicator amino acid and the required tracer washout time in preterms. METHODS: Subjects received a study diet for 6 d that entailed a 50% reduction in leucine. Tracer studies using enterally infused [(13)C]bicarbonate and [1-(13)C]phenylalanine were performed on days 1, 2, 4, and 6. Breath samples containing (13)CO2 were collected during steady state and measured by infrared spectrometric analysis, and the fraction of (13)CO2 recovery from (13)C oxidation (F(13)CO2) was calculated. RESULTS: Preterm infants (n = 11, birth weight 1.9 ± 0.1 kg, gestational age 32.6 ± 1.5 wk) received 166 mg/kg/d of leucine. Baseline enrichment changed significantly at day 1 of the study diet. F(13)CO2 did not change significantly between days 2 and 4 but was significantly lower at day 6. The tracer washout time was determined to be 7.5 h using a biphasic regression analysis. CONCLUSION: One day of adaptation to a new diet is necessary to adapt to the (13)C enrichment of the study formula before starting infant requirement studies. Adaptation for a period of 5 d results in a protein-sparing response. The minimal time between two studies within the same subject is 7.5 h.


Subject(s)
Amino Acids/metabolism , Infant, Premature , Humans , Infant, Newborn , Oxidation-Reduction
13.
J Nutr ; 142(11): 1983-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23054309

ABSTRACT

It is unknown what feeding strategy to use during chemotherapy-induced gastrointestinal mucositis, which causes weight loss and possibly malabsorption. To study the absorptive capacity of amino acids during mucositis, we determined the plasma availability of enterally administered amino acids (AA), their utilization for protein synthesis, and the preferential side of the intestine for AA uptake in rats with and without methotrexate (MTX)-induced mucositis. Four days after injection with MTX (60 mg/kg) or saline (controls), rats received a primed, continuous dual-isotope infusion (intraduodenal and intravenous) of labeled L-leucine, L-lysine, L-phenylalanine, L-threonine, and L-methionine. We collected blood samples, assessed jejunal histology, and determined labeled AA incorporation in proximal and distal small intestinal mucosa, plasma albumin, liver, and thigh muscle. MTX-induced mucositis was confirmed by histology. The median systemic availability of all AA except for leucine was similar in MTX-treated rats and in controls. However, the individual availability of all AA differed substantially within the group of MTX-treated rats, ranging from severely reduced (<10% of intake) to not different from controls (>40% of intake in 5 of 9 rats). More AA originating from basolateral uptake than those originating from apical uptake were used for intestinal protein synthesis in MTX-treated rats (≥420% more, P < 0.05). We conclude that continuous enteral administration can enable normal AA absorption in rats with MTX-induced mucositis. The intestine prefers basolateral AA uptake to meet its need for AA for protein synthesis during mucositis.


Subject(s)
Amino Acids/metabolism , Methotrexate/toxicity , Mucositis/chemically induced , Nucleic Acid Synthesis Inhibitors/toxicity , Absorption , Albumins/metabolism , Animals , Enteral Nutrition , Gene Expression Regulation , Injections, Intravenous , Intestinal Mucosa/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Specific Pathogen-Free Organisms
14.
Am J Clin Nutr ; 95(5): 1048-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22492372

ABSTRACT

BACKGROUND: The essential amino acid methionine can be used for protein synthesis but also serves as a precursor for homocysteine and cysteine. OBJECTIVE: The objective of this study was to determine the minimal obligatory methionine requirement of infants in the presence of excess cysteine (91 mg ⋅ kg(-1) ⋅ d(-1)) by using the indicator amino acid oxidation (IAAO) method with l-[1-(13)C]phenylalanine as the indicator. DESIGN: Fully enterally fed term infants <1 mo of age were randomly assigned to methionine intakes that ranged from 3 to 59 mg ⋅ kg(-1) ⋅ d(-1) as part of an elemental formula. After 1 d of adaptation to the test diet, [(13)C]bicarbonate and l-[1-(13)C]phenylalanine tracers were given enterally. Breath samples were collected at baseline and during isotopic plateaus. The mean methionine requirement was determined by using biphasic linear regression crossover analysis on the fraction of (13)CO(2) recovery from l-[1-(13)C]phenylalanine oxidation (F(13)CO(2)). Data are presented as means ± SDs. RESULTS: Thirty-three neonates (gestational age: 39 ± 1 wk) were studied at 13 ± 6 d. With increasing methionine intakes, F(13)CO(2) decreased until a methionine intake of 38 mg ⋅ kg(-1) ⋅ d(-1); additional increases in methionine intake did not affect F(13)CO(2). The mean methionine requirement was determined at 38 mg ⋅ kg(-1) ⋅ d(-1), and the upper and lower CIs were 48 and 27 mg ⋅ kg(-1) ⋅ d(-11), respectively (P < 0.0001, r(2) = 0.59). CONCLUSIONS: Although the current recommended methionine intake of 28 mg ⋅ kg(-1) ⋅ d(-1) is within the CIs of our study, the estimated mean requirement is substantially higher. However, most of the infant formulas provide a methionine intake of 49-80 mg ⋅ kg(-1) ⋅ d(-1), which is above the upper CI of our study. This trial was registered at www.trialregister.nl as NTR1610.


Subject(s)
Cysteine/administration & dosage , Cysteine/metabolism , Enteral Nutrition/methods , Methionine/administration & dosage , Methionine/metabolism , Carbon Isotopes/chemistry , Cross-Over Studies , Dietary Supplements , Dose-Response Relationship, Drug , Female , Humans , Infant Nutritional Physiological Phenomena , Infant, Newborn , Linear Models , Male , Nutritional Requirements , Oxidation-Reduction
15.
Am J Clin Nutr ; 94(6): 1496-503, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22049162

ABSTRACT

BACKGROUND: Infant nutrition has a major impact on child growth and functional development. Low and high intakes of protein or amino acids could have a detrimental effect. OBJECTIVE: The objective of the study was to determine the lysine requirement of enterally fed term neonates by using the indicator amino acid oxidation (IAAO) method. L-[1-(13)C]phenylalanine was used as an indicator amino acid. DESIGN: Twenty-one neonates were randomly assigned to lysine intakes that ranged from 15 to 240 mg · kg(-1) · d(-1). Breath, urine, and blood samples were collected at baseline and during the plateau. The mean lysine requirement was determined by using biphasic linear regression crossover analysis on the fraction of (13)CO(2) recovery from L-[1-(13)C]phenylalanine oxidation (F(13)CO(2)) and phenylalanine oxidation rates calculated from the L-[1-(13)C]phenylalanine enrichment of urine and plasma. RESULTS: The mean (±SD) phenylalanine flux calculated from urine and plasma L-[1-(13)C]phenylalanine enrichment data were 88.3 ± 6.9 and 84.5 ± 7.4 µmol · kg(-1) · h(-1), respectively. Graded intakes of lysine had no effect on phenylalanine fluxes. The mean lysine requirement determined by F(13)CO(2) was 130 mg · kg(-1) · d(-1) (upper and lower CIs: 183.7 and 76.3 mg · kg(-1) · d(-1), respectively). The mean requirement was identical to the requirement determined by using phenylalanine oxidation rates in urine and plasma. CONCLUSIONS: The mean lysine requirement of enterally fed term neonates was determined by using F(13)CO(2) and phenylalanine oxidation rates calculated from the L-[1-(13)C]phenylalanine enrichment of urine and plasma. These methods yielded a similar result of 130 mg lysine · kg(-1) · d(-1). This study demonstrates that sampling of (13)CO(2) in expired air is sufficient to estimate the lysine requirement by using the IAAO method in infants. This trial was registered at www.trialregister.nl as NTR1610.


Subject(s)
Enteral Nutrition , Infant Nutritional Physiological Phenomena , Infant, Newborn , Lysine/administration & dosage , Nutritional Requirements , Carbon Dioxide/metabolism , Carbon Isotopes/metabolism , Cross-Over Studies , Female , Humans , Isotope Labeling , Lysine/metabolism , Male , Oxidation-Reduction , Phenylalanine/metabolism
16.
Rapid Commun Mass Spectrom ; 23(18): 2897-902, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19670340

ABSTRACT

Determination of glutathione kinetics using stable isotopes requires accurate measurement of the tracers and tracees. Previously, the precursor and synthesized product were measured with two separate techniques, liquid chromatography/isotope ratio mass spectrometry (LC/IRMS) and gas chromatography/combustion/isotope ratio mass spectrometry (GC/C/IRMS). In order to reduce sample volume and minimize analytical effort we developed a method to simultaneously determine (13)C-glutathione as its dimeric form (GSSG) and its precursor [1-(13)C]glycine in a small volume of erythrocytes in one single analysis. After having transformed (13)C-glutathione into its dimeric form GSSG, we determined both the intra-erythrocytic concentrations and the (13)C-isotopic enrichment of GSSG and glycine in 150 microL of whole blood using liquid chromatography coupled to LC/IRMS. The results show that the concentration (range of micromol/mL) was reliably measured using cycloleucine as internal standard, i.e. with a precision better than 0.1 micromol/mL. The (13)C-isotopic enrichment of GSSG and glycine measured in the same run gave reliable values with excellent precision (standard deviation (sd) <0.3 per thousand) and accuracy (measured between 0 and 5 APE). This novel method opens up a variety of kinetic studies with relatively low dose administration of tracers, reducing the total cost of the study design. In addition, only a minimal sample volume is required, enabling studies even in very small subjects, such as preterm infants.


Subject(s)
Chromatography, Liquid/methods , Glutathione Disulfide/chemistry , Glutathione/chemistry , Glycine/chemistry , Mass Spectrometry/methods , Carbon Isotopes/chemistry , Dimerization , Erythrocytes/chemistry , Erythrocytes/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Infant, Newborn , Isotope Labeling
17.
Pediatrics ; 121(3): e561-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18283068

ABSTRACT

OBJECTIVE: Optimal nutrition is of utmost importance for the preterm infant's later health and developmental outcome. Amino acid requirements for preterm infants differ from those for term and older infants, because growth rates differ. Some nonessential amino acids, however, cannot be sufficiently synthesized endogenously. Cyst(e)ine is supposed to be such a conditionally essential amino acid in preterm infants. The objective of this study was to determine, at 32 and 35 weeks' postmenstrual age, cyst(e)ine requirements in fully enterally fed very low birth weight preterm infants with gestational ages of <29 weeks. METHODS: Infants were randomly assigned to 1 of the 5 graded cystine test diets that contained generous amounts of methionine. Cyst(e)ine requirement was determined with the indicator amino acid oxidation technique ([1-(13)C]phenylalanine) after 24-hour adaptation. RESULTS: Fractional [1-(13)C]phenylalanine oxidation was established in 47 very low birth weight preterm infants (mean gestational age: 28 weeks +/- 1 week SD; birth weight: 1.07 kg +/- 0.21 kg SD). Increase in dietary cyst(e)ine intake did not result in a decrease in fractional [1-(13)C]phenylalanine oxidation. CONCLUSIONS: These data do not support the hypothesis that endogenous cyst(e)ine synthesis is limited in very low birth weight preterm infants with gestational ages of <29 weeks, both at 32 and 35 weeks postmenstrual age. It is safe to conclude that cyst(e)ine requirement is <18 mg/kg per day in enterally fed very low birth weight preterm infants who are older than 32 weeks' postmenstrual age and whose methionine intake is adequate. Therefore, cyst(e)ine is probably not a conditionally essential amino acid in these infants.


Subject(s)
Cysteine/administration & dosage , Enteral Nutrition/methods , Infant, Very Low Birth Weight , Nutritional Requirements , Weight Gain , Analysis of Variance , Body Weight , Female , Follow-Up Studies , Humans , Infant, Newborn , Male , Nutritional Support , Probability , Regression Analysis , Treatment Outcome
18.
Am J Clin Nutr ; 86(4): 1120-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17921391

ABSTRACT

BACKGROUND: Cyst(e)ine can be synthesized de novo from methionine and serine and is, therefore, a nonessential amino acid in human adults. Several studies have suggested that cyst(e)ine might be a conditionally essential amino acid in preterm infants because of biochemical immaturity. No data are available on cyst(e)ine requirements in low-birth-weight (LBW) preterm infants. OBJECTIVE: The aim was to determine cyst(e)ine requirements in LBW infants with gestational ages from 32 to 34 wk, measured 1 mo after birth with the use of the indicator amino acid oxidation technique. DESIGN: LBW infants were randomly assigned to 1 or 2 of the 5 formulas containing graded cystine concentrations (11, 22, 32, 43, or 65 mg cyst(e)ine/100 mL) and generous amounts of methionine. After 24-h adaptation, cyst(e)ine requirement was determined by (13)CO(2) release from [1-(13)C]phenylalanine in expired breath. (13)CO(2) enrichment was measured by isotopic ratio mass spectrometry. RESULTS: Cyst(e)ine requirement was determined in 25 LBW infants with a mean (+/-SD) gestational age of 33 +/- 1 wk and birth weight of 1.78 +/- 0.32 kg. Fractional oxidation of [1-(13)C]phenylalanine did not differ between the 5 groups. CONCLUSIONS: There is no evidence for limited endogenous cyst(e)ine synthesis in 4-wk-old LBW preterm infants born at gestational ages from 32 to 34 wk. It is safe to conclude that the cyst(e)ine requirement is <18 mg kg(-1) d(-1) providing generous amounts of methionine and that cyst(e)ine is probably not a conditionally essential amino acid in fully enterally fed LBW preterm infants born at 32-34 wk.


Subject(s)
Cysteine/administration & dosage , Cysteine/metabolism , Infant Nutritional Physiological Phenomena/physiology , Infant, Low Birth Weight/metabolism , Infant, Newborn/growth & development , Nutritional Requirements , Amino Acids, Essential/administration & dosage , Amino Acids, Essential/metabolism , Breath Tests , Carbon Isotopes , Dose-Response Relationship, Drug , Female , Gestational Age , Humans , Infant, Low Birth Weight/growth & development , Infant, Premature/growth & development , Infant, Premature/metabolism , Male , Methionine/administration & dosage , Methionine/metabolism , Oxidation-Reduction
19.
Pediatr Res ; 59(5): 732-5, 2006 May.
Article in English | MEDLINE | ID: mdl-16627891

ABSTRACT

We previously showed that, in prematurely born infants, an anabolic state without metabolic acidosis can be achieved upon intravenous amino acid (AA) administration in the immediate postnatal phase, despite a low energy intake. We hypothesized that the anabolic state resulted from an increased protein synthesis and not a decreased proteolysis. Furthermore, we hypothesized that the energy needed for the higher protein synthesis rate would be derived from an increased glucose oxidation. To test our hypotheses, 32 ventilated premature infants (<1500 g) received intravenously either solely glucose or glucose and 2.4 g AA/kg/d immediately postnatally. On postnatal d 2, each group received primed continuous infusions of either [1-13C]leucine or [U-13C6]glucose. 13CO2 enrichments in expiratory air and plasma [1-13C]alpha-KICA (as an intracellular leucine precursor) and [U-13C6]glucose enrichments were measured by mass spectrometry techniques. The AA administration resulted in an increased incorporation of leucine into body protein and a higher leucine oxidation rate, whereas leucine release from proteolysis was not affected. Glucose oxidation rate did not increase upon AA administration. In conclusion, the anabolic state resulting from AA administration in the immediate postnatal period resulted from increased protein synthesis and not decreased proteolysis. The energy needed for the additional protein synthesis was not derived from an increased glucose oxidation.


Subject(s)
Amino Acids/administration & dosage , Glucose/metabolism , Infant, Premature/metabolism , Leucine/metabolism , Carbon Isotopes , Female , Gluconeogenesis , Glycogenolysis , Humans , Infant, Newborn , Infant, Small for Gestational Age , Infusions, Intravenous , Kinetics , Male , Nitrogen/metabolism , Nutritional Status , Oxidation-Reduction , Protein Biosynthesis
20.
Pediatr Res ; 58(5): 861-4, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16183827

ABSTRACT

The metabolic fate of substrates in humans can be examined by the use of stable isotopes, one of which, [13C]bicarbonate, may serve to estimate CO2 production rate. In view of minimizing the burden of metabolic studies for preterm infants, the authors determined whether intragastric and intravenous infusions of [13C]bicarbonate would achieve the same 13CO2 enrichment in expired air during steady state. A second aim of this study was to determine the minimum time required to reach steady state during intragastric infusion. Ten preterm infants received a primed continuous [13C]bicarbonate infusion intragastrically, followed by an intravenous infusion the next day. Breath samples were obtained every 30 min by the direct sampling method. 13CO2 isotopic enrichment, expressed as atom percent excess, was measured by isotopic ratio mass spectrometry. Two-tailed t tests were used to detect statistically significant differences between the infusion routes. The isotopic enrichment at plateau did not differ between intragastric and intravenous infusion. A steady state of 13CO2 enrichment was achieved after 60 min of intravenous infusion and after 120 min of intragastric infusion. In conclusion, intragastric infusion of [13C]bicarbonate may serve to estimate the whole-body CO2 production rate in preterm infants. To reach 13CO2 steady state, a minimum of 120 min of bicarbonate administration is required.


Subject(s)
Bicarbonates/administration & dosage , Infant, Premature , Bicarbonates/metabolism , Birth Weight , Carbon Isotopes , Drug Administration Routes , Female , Humans , Infant, Newborn , Infusions, Intravenous , Male , Stomach
SELECTION OF CITATIONS
SEARCH DETAIL
...