Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Amino Acids ; 52(9): 1319-1335, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32974749

ABSTRACT

When neonatal pigs continuously fed formula are supplemented with leucine pulses, muscle protein synthesis and body weight gain are enhanced. To identify the responsible mechanisms, we combined plasma metabolomic analysis with transcriptome expression of the transcriptome and protein catabolic pathways in skeletal muscle. Piglets (n = 23, 7-day-old) were fed continuously a milk replacement formula via orogastric tube for 21 days with an additional parenteral infusion (800 µmol kg-1 h-1) of either leucine (LEU) or alanine (CON) for 1 h every 4 h. Plasma metabolites were measured by liquid chromatography-mass spectrometry. Gene and protein expression analyses of longissimus dorsi muscle were performed by RNA-seq and Western blot, respectively. Compared with CON, LEU pigs had increased plasma levels of leucine-derived metabolites, including 4-methyl-2-oxopentanoate, beta-hydroxyisovalerate, ß-hydroxyisovalerylcarnitine, and 3-methylglutaconate (P ≤ 0.05). Leucine pulses downregulated transcripts enriched in the Kyoto Encyclopedia of Genes and Genomes terms "spliceosome," "GAP junction," "endocytosis," "ECM-receptor interaction," and "DNA replication". Significant correlations were identified between metabolites derived from leucine catabolism and muscle genes involved in protein degradation, transcription and translation, and muscle maintenance and development (P ≤ 0.05). Further, leucine pulses decreased protein expression of autophagic markers and serine/threonine kinase 4, involved in muscle atrophy (P ≤ 0.01). In conclusion, results from our studies support the notion that leucine pulses during continuous enteral feeding enhance muscle mass gain in neonatal pigs by increasing protein synthetic activity and downregulating protein catabolic pathways through concerted responses in the transcriptome and metabolome.


Subject(s)
Dietary Supplements , Leucine/pharmacology , Metabolome/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscular Atrophy/pathology , Transcriptome/drug effects , Animals , Animals, Newborn , Female , Leucine/administration & dosage , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Phosphorylation , Swine
2.
J Nutr ; 150(1): 22-30, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31518419

ABSTRACT

BACKGROUND: Rapid growth of skeletal muscle in the neonate requires the coordination of protein deposition and myonuclear accretion. During this developmental stage, muscle protein synthesis is highly sensitive to amino acid supply, especially Leu, but we do not know if this is true for satellite cells, the source of muscle fiber myonuclei. OBJECTIVE: We examined whether dietary protein restriction reduces myonuclear accretion in the neonatal pig, and if any reduction in myonuclear accretion is mitigated by restoring Leu intake. METHODS: Neonatal pigs (1.53 ± 0.2 kg) were fitted with jugular vein and gastric catheters and fed 1 of 3 isoenergetic milk replacers every 4 h for 21 d: high protein [HP; 22.5 g protein/(kg/d); n= 8]; restricted protein [RP; 11.2 g protein/(kg/d); n= 10]; or restricted protein with Leu [RPL; 12.0 g protein/(kg/d); n= 10]. Pigs were administered 5-bromo-2'-deoxyuridine (BrdU; 15 mg/kg) intravenously every 12 h from days 6 to 8. Blood was sampled on days 6 and 21 to measure plasma Leu concentrations. On day 21, pigs were killed and the longissimus dorsi (LD) muscle was collected to measure cell morphometry, satellite cell abundance, myonuclear accretion, and insulin-like growth factor (IGF) system expression. RESULTS: Compared with HP pigs, postprandial plasma Leu concentration in RP pigs was 37% and 47% lower on days 6 and 21, respectively (P < 0.05); Leu supplementation in RPL pigs restored postprandial Leu to HP concentrations. Dietary protein restriction reduced LD myofiber cross-sectional area by 21%, satellite cell abundance by 35%, and BrdU+ myonuclear abundance by 25% (P < 0.05); Leu did not reverse these outcomes. Dietary protein restriction reduced LD muscle IGF2 expression by 60%, but not IGF1 or IGF1R expression (P < 0.05); Leu did not rescue IGF2 expression. CONCLUSIONS: Satellite cell abundance and myonuclear accretion in neonatal pigs are compromised when dietary protein intake is restricted and are not restored with Leu supplementation.


Subject(s)
Dietary Proteins/administration & dosage , Dietary Supplements , Leucine/administration & dosage , Satellite Cells, Skeletal Muscle/drug effects , Swine/physiology , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Diet/veterinary , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/physiology
3.
Nat Commun ; 10(1): 5364, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792207

ABSTRACT

DNA methylation regulates cell type-specific gene expression. Here, in a transgenic mouse model, we show that deletion of the gene encoding DNA methyltransferase Dnmt3a in hypothalamic AgRP neurons causes a sedentary phenotype characterized by reduced voluntary exercise and increased adiposity. Whole-genome bisulfite sequencing (WGBS) and transcriptional profiling in neuronal nuclei from the arcuate nucleus of the hypothalamus (ARH) reveal differentially methylated genomic regions and reduced expression of AgRP neuron-associated genes in knockout mice. We use read-level analysis of WGBS data to infer putative ARH neural cell types affected by the knockout, and to localize promoter hypomethylation and increased expression of the growth factor Bmp7 to AgRP neurons, suggesting a role for aberrant TGF-ß signaling in the development of this phenotype. Together, these data demonstrate that DNA methylation in AgRP neurons is required for their normal epigenetic development and neuron-specific gene expression profiles, and regulates voluntary exercise behavior.


Subject(s)
DNA Methylation , Neurons/metabolism , Physical Conditioning, Animal , Adiposity , Animals , Behavior, Animal , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Signal Transduction
4.
Article in English | MEDLINE | ID: mdl-29991029

ABSTRACT

Skeletal myogenesis begins in the embryo with proliferation and differentiation of muscle progenitor cells that ultimately fuse to form multinucleated myofibers. After midgestation, muscle growth occurs through hypertrophy of these myofibers. The most rapid growth phase occurs in the perinatal period, resulting in the expansion of muscle mass from 25% of lean mass at birth to 40-45% at maturity. These 2 phases of muscle growth are regulated by distinct molecular mechanisms engaged by extracellular cues and intracellular signaling pathways and regulatory networks they activate. Nutrients influence muscle growth by both providing the necessary substrates and eliciting extracellular cues which regulate the signal transduction pathways that control the anabolic processes of the fibers. The uniquely large capacity of immature myofibers for hypertrophy is enabled by a heightened capacity and sensitivity of protein synthesis to feeding-induced changes in plasma insulin and amino acids, and the ability to expand their myonuclear population through proliferation of muscle precursor cells (satellite cells). With maturation, satellite cells become quiescent, limiting myonuclear accretion, and the capacity of the muscles for protein anabolism progressively diminishes. Therefore, the early developmental phases represent critical windows for muscle growth which, if disrupted, result in muscle mass deficits that are unlikely to be entirely recoverable.


Subject(s)
Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Nutritional Physiological Phenomena/physiology , Age Factors , Amino Acids/blood , Animals , Cell Differentiation , Female , Fetal Development/physiology , Fetal Nutrition Disorders/physiopathology , Humans , Hypertrophy , Infant , Infant Nutrition Disorders/physiopathology , Infant Nutritional Physiological Phenomena/physiology , Infant, Newborn , Insulin/blood , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/biosynthesis , Perinatal Care , Pregnancy
5.
Nutrients ; 10(5)2018 May 22.
Article in English | MEDLINE | ID: mdl-29786674

ABSTRACT

Hepatic steatosis is a hallmark feature of kwashiorkor malnutrition. However, the pathogenesis of hepatic steatosis in kwashiorkor is uncertain. Our objective was to develop a mouse model of childhood undernutrition in order to test the hypothesis that feeding a maize vegetable diet (MVD), like that consumed by children at risk for kwashiorkor, will cause hepatic steatosis which is prevented by supplementation with choline. A MVD was developed with locally sourced organic ingredients, and fed to weanling mice (n = 9) for 6 or 13 days. An additional group of mice (n = 4) were fed a choline supplemented MVD. Weight, body composition, and liver changes were compared to control mice (n = 10) at the beginning and end of the study. The MVD resulted in reduced weight gain and hepatic steatosis. Choline supplementation prevented hepatic steatosis and was associated with increased hepatic concentrations of the methyl donor betaine. Our findings show that (1) feeding a MVD to weanling mice rapidly induces hepatic steatosis, which is a hallmark disturbance of kwashiorkor; and that (2) hepatic steatosis associated with feeding a MVD is prevented by choline supplementation. These findings support the concept that insufficient choline intake may contribute to the pathogenesis of hepatic steatosis in kwashiorkor.


Subject(s)
Choline/administration & dosage , Dietary Supplements , Fatty Liver/prevention & control , Kwashiorkor/diet therapy , Liver/pathology , Zea mays , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Choline/metabolism , Disease Models, Animal , Eating , Energy Metabolism/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Gene Expression Regulation , Kwashiorkor/genetics , Kwashiorkor/metabolism , Kwashiorkor/pathology , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Nutritional Status , Time Factors , Transcription, Genetic , Weight Gain
6.
PLoS One ; 12(1): e0170127, 2017.
Article in English | MEDLINE | ID: mdl-28099477

ABSTRACT

Offspring of murine dams chronically fed a protein-restricted diet have an increased risk for metabolic and neurobehavioral disorders. Previously we showed that adult offspring, developmentally exposed to a chronic maternal low-protein (MLP) diet, had lower body and hind-leg muscle weights and decreased liver enzyme serum levels. We conducted energy expenditure, neurobehavioral and circadian rhythm assays in male offspring to examine mechanisms for the body-weight phenotype and assess neurodevelopmental implications of MLP exposure. C57BL/6J dams were fed a protein restricted (8%protein, MLP) or a control protein (20% protein, C) diet from four weeks before mating until weaning of offspring. Male offspring were weaned to standard rodent diet (20% protein) and single-housed until 8-12 weeks of age. We examined body composition, food intake, energy expenditure, spontaneous rearing activity and sleep patterns and performed behavioral assays for anxiety (open field activity, elevated plus maze [EPM], light/dark exploration), depression (tail suspension and forced swim test), sociability (three-chamber), repetitive (marble burying), learning and memory (fear conditioning), and circadian behavior (wheel-running activity during light-dark and constant dark cycles). We also measured circadian gene expression in hypothalamus and liver at different Zeitgeber times (ZT). Male offspring from separate MLP exposed dams had significantly greater body fat (P = 0.03), less energy expenditure (P = 0.004), less rearing activity (P = 0.04) and a greater number of night-time rest/sleep bouts (P = 0.03) compared to control. MLP offspring displayed greater anxiety-like behavior in the EPM (P<0.01) but had no learning and memory deficit in fear-conditioning assay (P = 0.02). There was an effect of time on Per1, Per 2 and Clock circadian gene expression in the hypothalamus but not on circadian behavior. Thus, transplacental and early developmental exposure of dams to chronic MLP reduces food intake and energy expenditure, increases anxiety like behavior and disturbs sleep patterns but not circadian rhythm in adult male offspring.


Subject(s)
Anxiety/etiology , Circadian Rhythm/physiology , Diet, Protein-Restricted/adverse effects , Energy Metabolism , Sleep/physiology , Adipose Tissue , Animals , Behavior, Animal , Circadian Rhythm/genetics , Female , Gene Expression , Hypothalamus/physiology , Liver/physiology , Male , Maternal Nutritional Physiological Phenomena , Mice, Inbred C57BL
7.
Am J Physiol Endocrinol Metab ; 310(11): E1072-84, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27143558

ABSTRACT

Many low-birth weight infants are at risk for poor growth due to an inability to achieve adequate protein intake. Administration of the amino acid leucine stimulates protein synthesis in skeletal muscle of neonates. To determine the effects of enteral supplementation of the leucine metabolite ß-hydroxy-ß-methylbutyrate (HMB) on protein synthesis and the regulation of translation initiation and degradation pathways, overnight-fasted neonatal pigs were studied immediately (F) or fed one of five diets for 24 h: low-protein (LP), high-protein (HP), or LP diet supplemented with 4 (HMB4), 40 (HMB40), or 80 (HMB80) µmol HMB·kg body wt(-1)·day(-1) Cell replication was assessed from nuclear incorporation of BrdU in the longissimus dorsi (LD) muscle and jejunum crypt cells. Protein synthesis rates in LD, gastrocnemius, rhomboideus, and diaphragm muscles, lung, and brain were greater in HMB80 and HP and in brain were greater in HMB40 compared with LP and F groups. Formation of the eIF4E·eIF4G complex and S6K1 and 4E-BP1 phosphorylation in LD, gastrocnemius, and rhomboideus muscles were greater in HMB80 and HP than in LP and F groups. Phosphorylation of eIF2α and eEF2 and expression of SNAT2, LAT1, MuRF1, atrogin-1, and LC3-II were unchanged. Numbers of BrdU-positive myonuclei in the LD were greater in HMB80 and HP than in the LP and F groups; there were no differences in jejunum. The results suggest that enteral supplementation with HMB increases skeletal muscle protein anabolism in neonates by stimulation of protein synthesis and satellite cell proliferation.


Subject(s)
Dietary Supplements , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Valerates/administration & dosage , Administration, Oral , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Enteral Nutrition , Female , Male , Muscle, Skeletal/cytology , Protein Biosynthesis/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Swine , Up-Regulation/drug effects , Up-Regulation/physiology
8.
Am J Physiol Endocrinol Metab ; 310(8): E699-E713, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26884386

ABSTRACT

Neonatal pigs are used as a model to study and optimize the clinical treatment of infants who are unable to maintain oral feeding. Using this model, we have shown previously that pulsatile administration of leucine during continuous feeding over 24 h via orogastric tube enhanced protein synthesis in skeletal muscle compared with continuous feeding alone. To determine the long-term effects of leucine pulses, neonatal piglets (n = 11-12/group) were continuously fed formula via orogastric tube for 21 days, with an additional parenteral infusion of either leucine (CON + LEU; 800 µmol·kg-1·h-1) or alanine (CON + ALA) for 1 h every 4 h. The results show that body and muscle weights and lean gain were ∼25% greater, and fat gain was 48% lower in CON + LEU than CON + ALA; weights of other tissues were unaffected by treatment. Fractional protein synthesis rates in longissimus dorsi, gastrocnemius, and soleus muscles were ∼30% higher in CON + LEU compared with CON + ALA and were associated with decreased Deptor abundance and increased mTORC1, mTORC2, 4E-BP1, and S6K1 phosphorylation, SNAT2 abundance, and association of eIF4E with eIF4G and RagC with mTOR. There were no treatment effects on PKB, eIF2α, eEF2, or PRAS40 phosphorylation, Rheb, SLC38A9, v-ATPase, LAMTOR1, LAMTOR2, RagA, RagC, and LAT1 abundance, the proportion of polysomes to nonpolysomes, or the proportion of mRNAs encoding rpS4 or rpS8 associated with polysomes. Our results demonstrate that pulsatile delivery of a leucine supplement during 21 days of continuous enteral feeding enhances lean growth by stimulating the mTORC1-dependent translation initiation pathway, leading to protein synthesis in skeletal muscle of neonates.


Subject(s)
Leucine/pharmacology , Muscle Proteins/drug effects , Muscle, Skeletal/drug effects , Protein Biosynthesis/drug effects , Alanine/pharmacology , Amino Acid Transport System A/drug effects , Amino Acid Transport System A/metabolism , Animals , Animals, Newborn , Back Muscles , Dietary Supplements , Enteral Nutrition , Infusions, Parenteral , Leucine/administration & dosage , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/drug effects , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomal Proteins/drug effects , Ribosomal Proteins/genetics , Sus scrofa , Swine , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
9.
Amino Acids ; 48(1): 257-267, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26334346

ABSTRACT

Suboptimal nutrient intake represents a limiting factor for growth and long-term survival of low-birth weight infants. The objective of this study was to determine if in neonates who can consume only 70 % of their protein and energy requirements for 8 days, enteral leucine supplementation will upregulate the mammalian target of rapamycin (mTOR) pathway in skeletal muscle, leading to an increase in protein synthesis and muscle anabolism. Nineteen 4-day-old piglets were fed by gastric tube 1 of 3 diets, containing (kg body weight(-1) · day(-1)) 16 g protein and 190 kcal (CON), 10.9 g protein and 132 kcal (R), or 10.8 g protein + 0.2 % leucine and 136 kcal (RL) at 4-h intervals for 8 days. On day 8, plasma AA and insulin levels were measured during 6 post-feeding intervals, and muscle protein synthesis rate and mTOR signaling proteins were determined at 120 min post-feeding. At 120 min, leucine was highest in RL (P < 0.001), whereas insulin, isoleucine and valine were lower in RL and R compared to CON (P < 0.001). Compared to RL and R, the CON diet increased (P < 0.01) body weight, protein synthesis, phosphorylation of S6 kinase (p-S6K1) and 4E-binding protein (p-4EBP1), and activation of eukaryotic initiation factor 4 complex (eIF4E · eIF4G). RL increased (P ≤ 0.01) p-S6K1, p-4EBP1 and eIF4E · eIF4G compared to R. In conclusion, when protein and energy intakes are restricted for 8 days, leucine supplementation increases muscle mTOR activation, but does not improve body weight gain or enhance skeletal muscle protein synthesis in neonatal pigs.


Subject(s)
Animal Feed/analysis , Dietary Supplements/analysis , Leucine/metabolism , Muscle Proteins/metabolism , Protein Biosynthesis , Swine/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Energy Metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Female , Insulin/metabolism , Male , Muscle Proteins/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Phosphorylation , Swine/genetics , Swine/growth & development
10.
Am J Physiol Endocrinol Metab ; 309(6): E601-10, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26374843

ABSTRACT

Most low-birth weight infants experience extrauterine growth failure due to reduced nutrient intake as a result of feeding intolerance. The objective of this study was to determine whether prolonged enteral leucine supplementation improves lean growth in neonatal pigs fed a restricted protein diet. Neonatal pigs (n = 14-16/diet, 5 days old, 1.8 ± 0.3 kg) were fed by gastric catheter a whey-based milk replacement diet with either a high protein (HP) or restricted protein (RP) content or RP supplemented with leucine to the same level as in the HP diet (RPL). Pigs were fed 40 ml·kg body wt(-1)·meal(-1) every 4 h for 21 days. Feeding the HP diet resulted in greater total body weight and lean body mass compared with RP-fed pigs (P < 0.05). Masses of the longissimus dorsi muscle, heart, and kidneys were greater in the HP- than RP-fed pigs (P < 0.05). Body weight, lean body mass, and masses of the longissimus dorsi, heart, and kidneys in pigs fed the RPL diet were intermediate to RP- and HP-fed pigs. Protein synthesis and mTOR signaling were increased in all muscles with feeding (P < 0.05); leucine supplementation increased mTOR signaling and protein synthesis rate in the longissimus dorsi (P < 0.05). There was no effect of diet on indices of protein degradation signaling in any tissue (P > 0.05). Thus, when protein intake is chronically restricted, the capacity for leucine supplementation to enhance muscle protein accretion in neonatal pigs that are meal-fed milk protein-based diets is limited.


Subject(s)
Body Weight/drug effects , Diet, Protein-Restricted , Heart/drug effects , Kidney/drug effects , Leucine/pharmacology , Muscle, Skeletal/drug effects , Protein Biosynthesis/drug effects , Animals , Animals, Newborn , Dietary Supplements , Energy Intake , Heart/growth & development , Kidney/growth & development , Muscle, Skeletal/growth & development , Organ Size/drug effects , Random Allocation , Signal Transduction/drug effects , Sus scrofa , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
11.
Curr Opin Clin Nutr Metab Care ; 18(1): 102-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25474017

ABSTRACT

PURPOSE OF REVIEW: Neonates with feeding difficulties can be fed by orogastric tube, using either continuous or bolus delivery. This review reports on recent findings that bolus is advantageous compared to continuous feeding in supporting optimal protein anabolism. RECENT FINDINGS: Whether bolus or continuous feeding is more beneficial has been controversial, largely due to limitations inherent in clinical studies, such as the presence of confounding variables and the inability to use invasive approaches. Recent studies using the piglet as a model of the human neonate showed that, compared to continuous feeding, bolus feeding enhances protein synthesis and promotes greater protein deposition. The increase in protein synthesis occurs in muscles of varying fiber type and in visceral tissues whereas muscle protein degradation is largely insensitive to feeding pattern. This higher protein synthesis rate is enabled by the rapid and profound increases in circulating amino acids and insulin that occur following a bolus feed, which activate the intracellular signaling pathways leading to mRNA translation. SUMMARY: Recent findings indicate that bolus feeding enhances protein synthesis more than continuous feeding and promotes greater protein anabolism. The difference in response is attributable to the pulsatile pattern of amino acid-induced and insulin-induced translation initiation induced only by bolus feeding.


Subject(s)
Feeding Methods , Growth , Infant Nutritional Physiological Phenomena , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Protein Biosynthesis , Amino Acids/blood , Animals , Humans , Infant, Newborn , Insulin/blood
12.
J Nutr ; 144(9): 1349-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991043

ABSTRACT

Aging is often associated with overweight and obesity. There exists a long-standing debate about whether meal pattern also contributes to the development of obesity. The orexigenic hormone ghrelin regulates appetite and satiety by activating its receptor, growth hormone secretagogue receptor (GHS-R). In mice, circulating ghrelin concentrations and brain GHS-R expression were shown to increase with aging. To assess whether GHS-R regulates feeding pattern during aging, we studied meal patterns for the following cohorts of male mice fed a normal unpurified diet: 1) 3-4 mo, young wild-type (WT) mice; 2) 3-4 mo, young Ghsr-null (Ghsr(-/-)) mice; 3) 12-14 mo, middle-aged WT (WT-M) mice; 4) 12-14 mo, middle-aged Ghsr(-/-) (Ghsr(-/-)-M) mice; 5) 24-26 mo, old WT (WT-O) mice; and 6) 24-26 mo, old Ghsr(-/-) (Ghsr(-/-)-O) mice. Although the total daily food intake of Ghsr(-/-) mice was similar to that of WT controls, Ghsr(-/-)-M and Ghsr(-/-)-O mice had 9% (P = 0.07) and 16% (P < 0.05) less body weight compared with WT-M and WT-O mice, respectively, primarily due to reduced fat mass (P < 0.05, WT-M vs. Ghsr(-/-)-M and WT-O vs. Ghsr(-/-)-O). Intriguingly, Ghsr(-/-)-M mice ate larger meals (on average, Ghsr(-/-)-M mice ate 0.117 g/meal and WT-M mice ate 0.080 g/meal; P < 0.01) and took a longer time to eat (Ghsr(-/-)-M, 196.0 s and WT-M, 128.9 s; P < 0.01), but ate less frequently (Ghsr(-/-)-M, 31.0 times/d and WT-M, 42.3 times/d; P < 0.05) than WT-M controls. In addition, we found that expression of hypothalamic orexigenic peptides, neuropeptide Y (NPY) and agouti-related peptide (AgRP), was relatively lower in aged WT mice (P = 0.09 for NPY and P = 0.06 for AgRP), but anorexic peptide pro-opiomelanocortin (POMC) expression remained unchanged between the WT age groups. Interestingly, old Ghsr(-/-) mice had greater hypothalamic NPY expression (102% higher; P < 0.05) and AgRP expression (P = 0.07) but significantly lower POMC expression (P < 0.05) when compared with age-matched WT-O controls. Thus, our results indicate that GHS-R plays an important role in the regulation of meal pattern and that GHS-R ablation may modulate feeding behavior through the regulation of hypothalamic neuropeptides. Our results collectively suggest that ghrelin receptor antagonism may have a beneficial effect on metabolism during aging.


Subject(s)
Appetite Regulation , Energy Intake , Feeding Behavior , Ghrelin/metabolism , Portion Size , Receptors, Ghrelin/metabolism , Satiety Response , Adipose Tissue , Age Factors , Aging , Animals , Body Weight , Eating , Hypothalamus , Male , Meals , Mice , Mice, Inbred Strains , Neuropeptides/metabolism , Satiation
13.
Diabetes ; 62(8): 2773-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23545705

ABSTRACT

Decades of research in rodent models has shown that early postnatal overnutrition induces excess adiposity and other components of metabolic syndrome that persist into adulthood. The specific biologic mechanisms explaining the persistence of these effects, however, remain unknown. On postnatal day 1 (P1), mice were fostered in control (C) or small litters (SL). SL mice had increased body weight and adiposity at weaning (P21), which persisted to adulthood (P180). Detailed metabolic studies indicated that female adult SL mice have decreased physical activity and energy expenditure but not increased food intake. Genome-scale DNA methylation profiling identified extensive changes in hypothalamic DNA methylation during the suckling period, suggesting that it is a critical period for developmental epigenetics in the mouse hypothalamus. Indeed, SL mice exhibited subtle and sex-specific changes in hypothalamic DNA methylation that persisted from early life to adulthood, providing a potential mechanistic basis for the sustained physiological effects. Expression profiling in adult hypothalamus likewise provided evidence of widespread sex-specific alterations in gene expression. Together, our data indicate that early postnatal overnutrition leads to a reduction in spontaneous physical activity and energy expenditure in females and suggest that early postnatal life is a critical period during which nutrition can affect hypothalamic developmental epigenetics.


Subject(s)
Eating/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Motor Activity/physiology , Adiposity/genetics , Animals , Animals, Newborn/metabolism , Body Weight/physiology , DNA Methylation , Female , Gene Expression , Male , Mice , Nutritional Status , Obesity/genetics , Obesity/metabolism , Overnutrition/genetics , Overnutrition/metabolism
14.
Pediatr Res ; 71(4 Pt 1): 324-31, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22391631

ABSTRACT

INTRODUCTION: Leucine (Leu) activates mammalian target of rapamycin (mTOR) to upregulate protein synthesis (PS). RESULTS: PS in skeletal muscles, heart, liver, pancreas, and jejunum, but not kidney, were greater in low protein supplemented with Leu (LP+L) than LP, but lower than high protein (HP). In longissimus dorsi muscle, protein kinase B phosphorylation was similar in LP and LP+L, but lower than HP. Although less than HP, p70 ribosomal S6 kinase 1 (S6K1) and eukaryotic initiation factor (eIF) 4E binding protein 1 (4EBP1) association with regulatory associated protein of mammalian target of rapamycin was greater in LP+L than LP, resulting in higher S6K1 and 4EBP1 phosphorylation. Feeding LP+L vs. LP decreased 4EBP1·eIF4E and increased eIF4E·eIF4G formation, but not to HP. Similar results were obtained for S6K1 and 4EBP1 phosphorylation in gastrocnemius, masseter, heart, liver, pancreas, and jejunum, but not kidney. eIF2α and elongation factor 2 phosphorylation was unaffected by treatment. DICUSSION: Our results suggest that enteral Leu supplementation of a low protein diet enhances PS in most tissues through mTOR complex 1 pathways. METHODS: To examine enteral Leu effects on PS and signaling activation, 5-d-old piglets were fed for 24 h diets containing: (i) LP, (ii) LP+L, or (iii) HP.


Subject(s)
Leucine/therapeutic use , Muscle, Skeletal/metabolism , Myocardium/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids/metabolism , Animals , Animals, Newborn , Blood Glucose/metabolism , Dietary Supplements , Enteral Nutrition/methods , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factors/chemistry , Glycolysis , Insulin/blood , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Swine , Time Factors , Tissue Distribution
15.
J Nutr ; 140(12): 2145-52, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20962152

ABSTRACT

Protein synthesis and eukaryotic initiation factor (eIF) activation are increased in skeletal muscle of neonatal pigs parenterally infused with amino acids. Leucine appears to be the most effective single amino acid to trigger these effects. To examine the response to enteral leucine supplementation, overnight food-deprived 5-d-old pigs were gavage fed at 0 and 60 min a: 1) low-protein diet (LP); 2) LP supplemented with leucine (LP+L) to equal leucine in the high-protein diet (HP); or 3) HP diet. Diets were isocaloric and equal in lactose. Fractional protein synthesis rates and translation initiation control mechanisms were examined in skeletal muscles and visceral tissues 90 min after feeding. Protein synthesis rates in longissimus dorsi, gastrocnemius, and masseter muscles, heart, jejunum, kidney, and pancreas, but not liver, were greater in the LP+L group compared with the LP group and did not differ from the HP group. Feeding LP+L and HP diets compared with the LP diet increased phosphorylation of mammalian target of rapamycin (mTOR), 4E-binding protein 1, ribosomal protein S6 kinase-1, and eIF4G and formation of the active eIF4E·eIF4G complex in longissimus dorsi muscle. In all tissues except liver, activation of mTOR effectors increased in pigs fed LP+L and HP vs. LP diets. Our results suggest that leucine supplementation of a low-protein meal stimulates protein synthesis in muscle and most visceral tissues to a rate similar to that achieved by feeding a high-protein meal and this stimulation involves activation of mTOR downstream effectors.


Subject(s)
Dietary Proteins/administration & dosage , Leucine/administration & dosage , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Protein Biosynthesis/physiology , TOR Serine-Threonine Kinases/physiology , Amino Acids/blood , Animals , Animals, Newborn , Blood Glucose/analysis , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Insulin/blood , Pregnancy , Swine
SELECTION OF CITATIONS
SEARCH DETAIL