Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Sci Signal ; 8(384): ra68, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26152695

ABSTRACT

Nitric oxide (NO) is a signaling intermediate during glutamatergic neurotransmission in the central nervous system (CNS). NO signaling is in part accomplished through cysteine S-nitrosylation, a posttranslational modification by which NO regulates protein function and signaling. In our investigation of the protein targets and functional impact of S-nitrosylation in the CNS under physiological conditions, we identified 269 S-nitrosocysteine residues in 136 proteins in the wild-type mouse brain. The number of sites was significantly reduced in the brains of mice lacking endothelial nitric oxide synthase (eNOS(-/-)) or neuronal nitric oxide synthase (nNOS(-/-)). In particular, nNOS(-/-) animals showed decreased S-nitrosylation of proteins that participate in the glutamate/glutamine cycle, a metabolic process by which synaptic glutamate is recycled or oxidized to provide energy. (15)N-glutamine-based metabolomic profiling and enzymatic activity assays indicated that brain extracts from nNOS(-/-) mice converted less glutamate to glutamine and oxidized more glutamate than those from mice of the other genotypes. GLT1 [also known as EAAT2 (excitatory amino acid transporter 2)], a glutamate transporter in astrocytes, was S-nitrosylated at Cys(373) and Cys(561) in wild-type and eNOS(-/-) mice, but not in nNOS(-/-) mice. A form of rat GLT1 that could not be S-nitrosylated at the equivalent sites had increased glutamate uptake compared to wild-type GLT1 in cells exposed to an S-nitrosylating agent. Thus, NO modulates glutamatergic neurotransmission through the selective, nNOS-dependent S-nitrosylation of proteins that govern glutamate transport and metabolism.


Subject(s)
Brain/metabolism , Cysteine/metabolism , Glutamic Acid/metabolism , Nitric Oxide/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Liquid , Cysteine/analogs & derivatives , Cysteine/genetics , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Glutamine/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Proteome/metabolism , Proteomics/methods , Rats , S-Nitrosothiols/metabolism , Tandem Mass Spectrometry
2.
J Clin Invest ; 125(7): 2841-50, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26098218

ABSTRACT

Increasing evidence indicates that the gut microbiota can be altered to ameliorate or prevent disease states, and engineering the gut microbiota to therapeutically modulate host metabolism is an emerging goal of microbiome research. In the intestine, bacterial urease converts host-derived urea to ammonia and carbon dioxide, contributing to hyperammonemia-associated neurotoxicity and encephalopathy in patients with liver disease. Here, we engineered murine gut microbiota to reduce urease activity. Animals were depleted of their preexisting gut microbiota and then inoculated with altered Schaedler flora (ASF), a defined consortium of 8 bacteria with minimal urease gene content. This protocol resulted in establishment of a persistent new community that promoted a long-term reduction in fecal urease activity and ammonia production. Moreover, in a murine model of hepatic injury, ASF transplantation was associated with decreased morbidity and mortality. These results provide proof of concept that inoculation of a prepared host with a defined gut microbiota can lead to durable metabolic changes with therapeutic utility.


Subject(s)
Biological Therapy/methods , Digestive System/microbiology , Hyperammonemia/microbiology , Hyperammonemia/therapy , Microbiota , Ammonia/metabolism , Animals , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bioengineering , Chemical and Drug Induced Liver Injury/therapy , Digestive System/metabolism , Disease Models, Animal , Feces/microbiology , Female , Genes, Bacterial , Hyperammonemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Microbiota/physiology , Time Factors , Urease/genetics , Urease/metabolism
3.
J Pediatr ; 165(2): 401-403.e3, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24880889

ABSTRACT

Identical studies using stable isotopes were performed before and after a 3-day trial of oral N-carbamyl-l-glutamate (NCG) in 5 subjects with late-onset carbamyl phosphate synthetase deficiency. NCG augmented ureagenesis and decreased plasma ammonia in 4 of 5 subjects. There was marked improvement in nitrogen metabolism with long-term NCG administration in 1 subject.


Subject(s)
Carbamoyl-Phosphate Synthase I Deficiency Disease/drug therapy , Glutamates/therapeutic use , Glutamine/blood , Urea/metabolism , Adolescent , Adult , Ammonia/blood , Carbamoyl-Phosphate Synthase I Deficiency Disease/blood , Child , Child, Preschool , Female , Humans , Linear Models , Male , Mass Spectrometry , Treatment Outcome , Young Adult
4.
J Biol Chem ; 289(14): 9710-29, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24523404

ABSTRACT

Agmatine (AGM), a product of arginine decarboxylation, influences multiple physiologic and metabolic functions. However, the mechanism(s) of action, the impact on whole body gene expression and metabolic pathways, and the potential benefits and risks of long term AGM consumption are still a mystery. Here, we scrutinized the impact of AGM on whole body metabolic profiling and gene expression and assessed a plausible mechanism(s) of AGM action. Studies were performed in rats fed a high fat diet or standard chow. AGM was added to drinking water for 4 or 8 weeks. We used (13)C or (15)N tracers to assess metabolic reactions and fluxes and real time quantitative PCR to determine gene expression. The results demonstrate that AGM elevated the synthesis and tissue level of cAMP. Subsequently, AGM had a widespread impact on gene expression and metabolic profiling including (a) activation of peroxisomal proliferator-activated receptor-α and its coactivator, PGC1α, and (b) increased expression of peroxisomal proliferator-activated receptor-γ and genes regulating thermogenesis, gluconeogenesis, and carnitine biosynthesis and transport. The changes in gene expression were coupled with improved tissue and systemic levels of carnitine and short chain acylcarnitine, increased ß-oxidation but diminished incomplete fatty acid oxidation, decreased fat but increased protein mass, and increased hepatic ureagenesis and gluconeogenesis but decreased glycolysis. These metabolic changes were coupled with reduced weight gain and a curtailment of the hormonal and metabolic derangements associated with high fat diet-induced obesity. The findings suggest that AGM elevated the synthesis and levels of cAMP, thereby mimicking the effects of caloric restriction with respect to metabolic reprogramming.


Subject(s)
Agmatine/pharmacology , Cyclic AMP/metabolism , Fatty Acids/metabolism , Gluconeogenesis/drug effects , Liver/metabolism , Obesity/drug therapy , Agmatine/pharmacokinetics , Animals , Biological Transport, Active/drug effects , Carnitine/analogs & derivatives , Carnitine/metabolism , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Gene Expression Regulation/drug effects , Humans , Male , Metabolome , Obesity/chemically induced , Obesity/metabolism , Oxidation-Reduction/drug effects , PPAR gamma/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Sprague-Dawley , Time Factors , Transcription Factors/biosynthesis
5.
J Biol Chem ; 288(6): 3938-51, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23266825

ABSTRACT

Paracrine signaling between pancreatic islet ß-cells and α-cells has been proposed to play a role in regulating glucagon responses to elevated glucose and hypoglycemia. To examine this possibility in human islets, we used a metabolomic approach to trace the responses of amino acids and other potential neurotransmitters to stimulation with [U-(13)C]glucose in both normal individuals and type 2 diabetics. Islets from type 2 diabetics uniformly showed decreased glucose stimulation of insulin secretion and respiratory rate but demonstrated two different patterns of glucagon responses to glucose: one group responded normally to suppression of glucagon by glucose, but the second group was non-responsive. The non-responsive group showed evidence of suppressed islet GABA levels and of GABA shunt activity. In further studies with normal human islets, we found that γ-hydroxybutyrate (GHB), a potent inhibitory neurotransmitter, is generated in ß-cells by an extension of the GABA shunt during glucose stimulation and interacts with α-cell GHB receptors, thus mediating the suppressive effect of glucose on glucagon release. We also identified glycine, acting via α-cell glycine receptors, as the predominant amino acid stimulator of glucagon release. The results suggest that glycine and GHB provide a counterbalancing receptor-based mechanism for controlling α-cell secretory responses to metabolic fuels.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucose/metabolism , Glycine/metabolism , Insulin-Secreting Cells/metabolism , Sodium Oxybate/metabolism , Adult , Diabetes Mellitus, Type 2/pathology , Female , Glucagon-Secreting Cells/pathology , Humans , Insulin-Secreting Cells/pathology , Male , Middle Aged , Receptors, GABA/metabolism , Receptors, Glycine/metabolism , gamma-Aminobutyric Acid/metabolism
6.
Biochem J ; 444(3): 537-51, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22448977

ABSTRACT

GKAs (glucokinase activators) are promising agents for the therapy of Type 2 diabetes, but little is known about their effects on hepatic intermediary metabolism. We monitored the fate of (13)C-labelled glucose in both a liver perfusion system and isolated hepatocytes. MS and NMR spectroscopy were deployed to measure isotopic enrichment. The results demonstrate that the stimulation of glycolysis by GKA led to numerous changes in hepatic metabolism: (i) augmented flux through the TCA (tricarboxylic acid) cycle, as evidenced by greater incorporation of (13)C into the cycle (anaplerosis) and increased generation of (13)C isotopomers of citrate, glutamate and aspartate (cataplerosis); (ii) lowering of hepatic [Pi] and elevated [ATP], denoting greater phosphorylation potential and energy state; (iii) stimulation of glycogen synthesis from glucose, but inhibition of glycogen synthesis from 3-carbon precursors; (iv) increased synthesis of N-acetylglutamate and consequently augmented ureagenesis; (v) increased synthesis of glutamine, alanine, serine and glycine; and (vi) increased production and outflow of lactate. The present study provides a deeper insight into the hepatic actions of GKAs and uncovers the potential benefits and risks of GKA for treatment of diabetes. GKA improved hepatic bioenergetics, ureagenesis and glycogenesis, but decreased gluconeogenesis with a potential risk of lactic acidosis and fatty liver.


Subject(s)
Benzeneacetamides/pharmacology , Glucokinase/metabolism , Hepatocytes/enzymology , Metabolomics/methods , Animals , Enzyme Activation/drug effects , Enzyme Activation/physiology , Hepatocytes/drug effects , Liver/drug effects , Liver/enzymology , Male , Rats , Rats, Sprague-Dawley
7.
Hum Mutat ; 32(10): 1153-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21681857

ABSTRACT

N-acetylglutamate synthase (NAGS) catalyzes the conversion of glutamate and acetyl-CoA to NAG, the essential allosteric activator of carbamyl phosphate synthetase I, the first urea cycle enzyme in mammals. A 17-year-old female with recurrent hyperammonemia attacks, the cause of which remained undiagnosed for 8 years in spite of multiple molecular and biochemical investigations, showed markedly enhanced ureagenesis (measured by isotope incorporation) in response to N-carbamylglutamate (NCG). This led to sequencing of the regulatory regions of the NAGS gene and identification of a deleterious single-base substitution in the upstream enhancer. The homozygous mutation (c.-3064C>A), affecting a highly conserved nucleotide within the hepatic nuclear factor 1 (HNF-1) binding site, was not found in single nucleotide polymorphism databases and in a screen of 1,086 alleles from a diverse population. Functional assays demonstrated that this mutation decreases transcription and binding of HNF-1 to the NAGS gene, while a consensus HNF-1 binding sequence enhances binding to HNF-1 and increases transcription. Oral daily NCG therapy restored ureagenesis in this patient, normalizing her biochemical markers, and allowing discontinuation of alternate pathway therapy and normalization of her diet with no recurrence of hyperammonemia. Inc.


Subject(s)
Amino-Acid N-Acetyltransferase/genetics , Enhancer Elements, Genetic , Glutamates/therapeutic use , Sequence Deletion , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/genetics , Adolescent , Alleles , Base Sequence , Binding Sites , Cell Line, Tumor , Child , Female , Gene Frequency , Glutamates/metabolism , Hep G2 Cells , Hepatocyte Nuclear Factor 1/metabolism , Humans , Nucleotide Motifs , Polymorphism, Single Nucleotide , Sequence Alignment , Treatment Outcome , Urea Cycle Disorders, Inborn/metabolism
8.
J Biol Chem ; 286(25): 22055-68, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21540182

ABSTRACT

We previously reported that isobutylmethylxanthine (IBMX), a derivative of oxypurine, inhibits citrulline synthesis by an as yet unknown mechanism. Here, we demonstrate that IBMX and other oxypurines containing a 2,6-dione group interfere with the binding of glutamate to the active site of N-acetylglutamate synthetase (NAGS), thereby decreasing synthesis of N-acetylglutamate, the obligatory activator of carbamoyl phosphate synthase-1 (CPS1). The result is reduction of citrulline and urea synthesis. Experiments were performed with (15)N-labeled substrates, purified hepatic CPS1, and recombinant mouse NAGS as well as isolated mitochondria. We also used isolated hepatocytes to examine the action of various oxypurines on ureagenesis and to assess the ameliorating affect of N-carbamylglutamate and/or l-arginine on NAGS inhibition. Among various oxypurines tested, only IBMX, xanthine, or uric acid significantly increased the apparent K(m) for glutamate and decreased velocity of NAGS, with little effect on CPS1. The inhibition of NAGS is time- and dose-dependent and leads to decreased formation of the CPS1-N-acetylglutamate complex and consequent inhibition of citrulline and urea synthesis. However, such inhibition was reversed by supplementation with N-carbamylglutamate. The data demonstrate that xanthine and uric acid, both physiologically occurring oxypurines, inhibit the hepatic synthesis of N-acetylglutamate. An important and novel concept emerging from this study is that xanthine and/or uric acid may have a role in the regulation of ureagenesis and, thus, nitrogen homeostasis in normal and disease states.


Subject(s)
Amino-Acid N-Acetyltransferase/antagonists & inhibitors , Down-Regulation/drug effects , Liver/metabolism , Urea/metabolism , Uric Acid/pharmacology , Xanthine/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Amino-Acid N-Acetyltransferase/isolation & purification , Amino-Acid N-Acetyltransferase/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/isolation & purification , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Citrulline/biosynthesis , Dose-Response Relationship, Drug , Glutamates/biosynthesis , Hepatocytes/drug effects , Hepatocytes/metabolism , Kinetics , Liver/cytology , Liver/enzymology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley
9.
Pediatrics ; 126(1): e208-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20566609

ABSTRACT

OBJECTIVES: The objective of this study was to determine whether N-carbamylglutamate (NCG) reduces plasma levels of ammonia and glutamine and increases the rate of ureagenesis in patients with propionic acidemia (PA). METHODS: Identical 4-hour studies were performed before and immediately after a 3-day trial of oral NCG in 7 patients with PA. An oral bolus of [(13)C]sodium acetate was administered at the start of each study, and sequential blood samples were obtained to measure [(13)C]urea, ammonia, urea, and amino acids. RESULTS: With longitudinal mixed-effects linear regression, peak [(13)C]urea increased after treatment with NCG (from 2.2 to 3.8 microM; P < .0005). There were concomitant decreases in mean plasma ammonia (59-43 microM; P < .018) and glutamine (552-331 microM; P < .0005). CONCLUSIONS: NCG augments ureagenesis and decreases plasma ammonia and glutamine in patients with PA. The drug may serve as an important therapeutic adjunct in the treatment of acute hyperammonemia in this disorder.


Subject(s)
Ammonia/blood , Glutamates/administration & dosage , Glutamine/blood , Propionic Acidemia/diagnosis , Propionic Acidemia/drug therapy , Urea/blood , Administration, Oral , Adolescent , Ammonia/metabolism , Blood Chemical Analysis , Child , Child, Preschool , Confidence Intervals , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Glutamine/metabolism , Humans , Infant , Male , Probability , Prospective Studies , Risk Assessment , Treatment Outcome , Urea/metabolism
10.
Mol Genet Metab ; 100 Suppl 1: S37-41, 2010.
Article in English | MEDLINE | ID: mdl-20338795

ABSTRACT

Stable isotopes have been an invaluable adjunct to biomedical research for more than 70years. Indeed, the isotopic approach has revolutionized our understanding of metabolism, revealing it to be an intensely dynamic process characterized by an unending cycle of synthesis and degradation. Isotopic studies have taught us that the urea cycle is intrinsic to such dynamism, since it affords a capacious mechanism by which to eliminate waste nitrogen when rates of protein degradation (or dietary protein intake) are especially high. Isotopes have enabled an appreciation of the degree to which ureagenesis is compromised in patients with urea cycle defects. Indeed, isotopic studies of urea cycle flux correlate well with the severity of cognitive impairment in these patients. Finally, the use of isotopes affords an ideal tool with which to gauge the efficacy of therapeutic interventions to augment residual flux through the cycle.


Subject(s)
Isotope Labeling/methods , Urea/metabolism , Amino-Acid N-Acetyltransferase/deficiency , Amino-Acid N-Acetyltransferase/metabolism , Ammonium Chloride/administration & dosage , Ammonium Chloride/pharmacology , Carbon Dioxide/metabolism , Carbon Isotopes/metabolism , Humans , Urea/blood
11.
Mol Genet Metab ; 98(4): 325-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19660971

ABSTRACT

We studied the effect on ureagenesis of a single dose of N-carbamylglutamate (NCG) in healthy young adults who received a constant infusion (300 min) of NaH(13)CO(3). Isotope ratio-mass spectrometry was used to measure the appearance of label in [(13)C]urea. At 90 min after initiating the H(13)CO3-infusion each subject took a single dose of NCG (50 mg/kg). In 5/6 studies the administration of NCG increased the formation of [(13)C]urea. Treatment with NCG significantly diminished the concentration of blood alanine, but not that of glutamine or arginine. The blood glucose concentration was unaffected by NCG administration. No untoward side effects were observed. The data indicate that treatment with NCG stimulates ureagenesis and could be useful in clinical settings of acute hyperammonemia of various etiologies.


Subject(s)
Glutamates/administration & dosage , Glutamates/pharmacology , Urea/metabolism , Adult , Amino Acids/blood , Blood Glucose/drug effects , Carbon Dioxide/metabolism , Carbon Isotopes , Demography , Dose-Response Relationship, Drug , Exhalation/drug effects , Female , Health , Humans , Infusions, Intravenous , Male , Sodium Bicarbonate/administration & dosage , Sodium Bicarbonate/pharmacology , Urea/blood , Young Adult
12.
Epilepsia ; 49 Suppl 8: 73-5, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19049594

ABSTRACT

We hypothesize that one mechanism of the anti-epileptic effect of the ketogenic diet is to alter brain handling of glutamate. According to this formulation, in ketotic brain astrocyte metabolism is more active, resulting in enhanced conversion of glutamate to glutamine. This allows for: (a) more efficient removal of glutamate, the most important excitatory neurotransmitter; and (b) more efficient conversion of glutamine to GABA, the major inhibitory neurotransmitter.


Subject(s)
Brain/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Ketosis/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Diet, Ketogenic , Humans
13.
J Biol Chem ; 283(25): 17238-49, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18445600

ABSTRACT

Pancreatic beta cells are hyper-responsive to amino acids but have decreased glucose sensitivity after deletion of the sulfonylurea receptor 1 (SUR1) both in man and mouse. It was hypothesized that these defects are the consequence of impaired integration of amino acid, glucose, and energy metabolism in beta cells. We used gas chromatography-mass spectrometry methodology to study intermediary metabolism of SUR1 knock-out (SUR1(-/-)) and control mouse islets with d-[U-(13)C]glucose as substrate and related the results to insulin secretion. The levels and isotope labeling of alanine, aspartate, glutamate, glutamine, and gamma-aminobutyric acid (GABA) served as indicators of intermediary metabolism. We found that the GABA shunt of SUR1(-/-) islets is blocked by about 75% and showed that this defect is due to decreased glutamate decarboxylase synthesis, probably caused by elevated free intracellular calcium. Glutaminolysis stimulated by the leucine analogue d,l-beta-2-amino-2-norbornane-carboxylic acid was, however, enhanced in SUR1(-/-) and glyburide-treated SUR1(+/+) islets. Glucose oxidation and pyruvate cycling was increased in SUR1(-/-) islets at low glucose but was the same as in controls at high glucose. Malic enzyme isoforms 1, 2, and 3, involved in pyruvate cycling, were all expressed in islets. High glucose lowered aspartate and stimulated glutamine synthesis similarly in controls and SUR1(-/-) islets. The data suggest that the interruption of the GABA shunt and the lack of glucose regulation of pyruvate cycling may cause the glucose insensitivity of the SUR1(-/-) islets but that enhanced basal pyruvate cycling, lowered GABA shunt flux, and enhanced glutaminolytic capacity may sensitize the beta cells to amino acid stimulation.


Subject(s)
Adenosine Triphosphate/chemistry , Glucose/metabolism , Glutamine/chemistry , Potassium/chemistry , Pyruvates/chemistry , gamma-Aminobutyric Acid/metabolism , Amino Acids/chemistry , Animals , Gas Chromatography-Mass Spectrometry/methods , Genotype , Glutamate Decarboxylase/metabolism , Mice , Mice, Transgenic , Models, Biological , Oxygen/metabolism
14.
J Biol Chem ; 283(22): 15063-71, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18375388

ABSTRACT

We previously showed that agmatine stimulated hepatic ureagenesis. In this study, we sought to determine whether the action of agmatine is mediated via cAMP signaling. A pilot experiment demonstrated that the phosphodiesterase inhibitor, 3-isobutylmethylxanthine (IBMX), inhibited urea synthesis albeit increased [cAMP]. Thus, we hypothesized that IBMX inhibits hepatic urea synthesis independent of [cAMP]. We further theorized that agmatine would negate the IBMX action and improve ureagenesis. Experiments were carried out with isolated mitochondria and (15)NH(4)Cl to trace [(15)N]citrulline production or [5-(15)N]glutamine and a rat liver perfusion system to trace ureagenesis. The results demonstrate that IBMX induced the following: (i) inhibition of the mitochondrial respiratory chain and diminished O(2) consumption during liver perfusion; (ii) depletion of the phosphorylation potential and overall hepatic energetic capacity; (iii) inhibition of [(15)N]citrulline synthesis; and (iv) inhibition of urea output in liver perfusion with little effect on [N-acetylglutamate]. The results indicate that IBMX directly and specifically inhibited complex I of the respiratory chain and carbamoyl-phosphate synthase-I (CPS-I), with an EC(50) about 0.6 mm despite a significant elevation of hepatic [cAMP]. Perfusion of agmatine with IBMX stimulated O(2) consumption, restored hepatic phosphorylation potential, and significantly stimulated ureagenesis. The action of agmatine may signify a cascade effect initiated by increased oxidative phosphorylation and greater ATP synthesis. In addition, agmatine may prevent IBMX from binding to one or more active site(s) of CPS-I and thus protect against inhibition of CPS-I. Together, the data may suggest a new experimental application of IBMX in studies of CPS-I malfunction and the use of agmatine as intervention therapy.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Agmatine/pharmacology , Liver/metabolism , Mitochondria, Liver/metabolism , Phosphodiesterase Inhibitors/pharmacology , Urea/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Cyclic AMP/metabolism , Electron Transport/drug effects , Male , Oxygen Consumption/drug effects , Perfusion , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley
15.
Pediatr Res ; 64(2): 213-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18414145

ABSTRACT

N-acetylglutamate (NAG) is an endogenous essential cofactor for conversion of ammonia to urea in the liver. Deficiency of NAG causes hyperammonemia and occurs because of inherited deficiency of its producing enzyme, NAG synthase (NAGS), or interference with its function by short fatty acid derivatives. N-carbamylglutamate (NCG) can ameliorate hyperammonemia from NAGS deficiency and propionic and methylmalonic acidemia. We developed a stable isotope (13)C tracer method to measure ureagenesis and to evaluate the effect of NCG in humans. Seventeen healthy adults were investigated for the incorporation of (13)C label into urea. [(13)C]urea appeared in the blood within minutes, reaching maximum by 100 min, whereas breath (13)CO(2) reached a maximum by 60 min. A patient with NAGS deficiency showed very little urea labeling before treatment with NCG and normal labeling thereafter. Correspondingly, plasma levels of ammonia and glutamine decreased markedly and urea tripled after NCG treatment. Similarly, in a patient with propionic acidemia, NCG treatment resulted in a marked increase in urea labeling and decrease in glutamine, alanine, and glycine. These results provide a reliable method for measuring the effect of NCG on nitrogen metabolism and strongly suggest that NCG could be an effective treatment for inherited and secondary NAGS deficiency.


Subject(s)
Glutamates/deficiency , Glutamates/pharmacology , Metabolic Diseases/blood , Propionates/blood , Urea/blood , Acetyl Coenzyme A/metabolism , Adult , Amino Acids/blood , Ammonia/blood , Biomarkers/blood , Carbon Dioxide/metabolism , Carbon Isotopes , Child , Feasibility Studies , Female , Glutamates/metabolism , Glutamates/therapeutic use , Humans , Male , Metabolic Diseases/drug therapy , Middle Aged
16.
Annu Rev Nutr ; 27: 415-30, 2007.
Article in English | MEDLINE | ID: mdl-17444813

ABSTRACT

In many epileptic patients, anticonvulsant drugs either fail adequately to control seizures or they cause serious side effects. An important adjunct to pharmacologic therapy is the ketogenic diet, which often improves seizure control, even in patients who respond poorly to medications. The mechanisms that explain the therapeutic effect are incompletely understood. Evidence points to an effect on brain handling of amino acids, especially glutamic acid, the major excitatory neurotransmitter of the central nervous system. The diet may limit the availability of oxaloacetate to the aspartate aminotransferase reaction, an important route of brain glutamate handling. As a result, more glutamate becomes accessible to the glutamate decarboxylase reaction to yield gamma-aminobutyric acid (GABA), the major inhibitory neurotransmitter and an important antiseizure agent. In addition, the ketogenic diet appears to favor the synthesis of glutamine, an essential precursor to GABA. This occurs both because ketone body carbon is metabolized to glutamine and because in ketosis there is increased consumption of acetate, which astrocytes in the brain quickly convert to glutamine. The ketogenic diet also may facilitate mechanisms by which the brain exports to blood compounds such as glutamine and alanine, in the process favoring the removal of glutamate carbon and nitrogen.


Subject(s)
Amino Acids/metabolism , Brain/metabolism , Ketone Bodies/metabolism , Ketosis/metabolism , Seizures/metabolism , Anticonvulsants/therapeutic use , Combined Modality Therapy , Diet , Glutamic Acid/metabolism , Humans , Seizures/diet therapy , Seizures/drug therapy , Treatment Outcome
17.
Am J Clin Nutr ; 85(1): 144-51, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17209190

ABSTRACT

BACKGROUND: To support age-appropriate growth and to prevent and treat malnutrition in children with cystic fibrosis (CF), energy requirements for those children are often set above the requirements for healthy children. Care providers use one of several empirically derived formulas to calculate energy requirements, yet the validity of these formulas has seldom been tested. OBJECTIVE: We evaluated 6 proposed formulas for calculating energy requirements in children with CF against a total energy requirement for children with CF (TER-CF) derived from measured total energy expenditure, fecal fat energy loss, and the theoretic energy required for age-appropriate tissue accretion. DESIGN: Subjects were children aged 6-8 y who had CF and pancreatic insufficiency. Calculated TERs from each formula were evaluated against TER-CF by using summary statistics, regression analysis, and residual plots. RESULTS: Subjects (n = 53) had suboptimal nutrition and growth status and mild-to-moderate lung disease. The formula that most closely (within 2%) approximated TER-CF was the estimated energy requirement (EER) formula at the active level (EERact). Regression analysis of TER-CF onto calculated TER from each formula yielded the best indexes of model fit for the EERact formula; residual plots of the EERact formula were tightly and normally distributed around zero. CONCLUSIONS: The EERact formula should be used to establish TER-CF in children in this age group who have mild-to-moderate CF. Changes in weight, height, and other indicators of nutritional status must be monitored to modify TER-CF as needed to support individual patient care goals.


Subject(s)
Cystic Fibrosis/therapy , Exocrine Pancreatic Insufficiency/therapy , Food, Formulated , Growth/drug effects , Nutritional Requirements , Weight Gain/drug effects , Anthropometry , Child , Child Development , Cystic Fibrosis/complications , Cystic Fibrosis/metabolism , Energy Intake/physiology , Energy Metabolism/physiology , Exocrine Pancreatic Insufficiency/complications , Exocrine Pancreatic Insufficiency/metabolism , Feces/chemistry , Female , Growth/physiology , Humans , Intestinal Absorption , Longitudinal Studies , Male , Nutrition Assessment , Nutritional Status , Weight Gain/physiology
18.
Am J Clin Nutr ; 84(3): 523-30, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16960165

ABSTRACT

BACKGROUND: Suboptimal growth and nutritional status are common among children with cystic fibrosis (CF) and pancreatic insufficiency (PI). A better understanding of energy balance is required to improve prevention and treatment of malnutrition. OBJECTIVE: Our objective was to characterize energy balance and the reporting accuracy of dietary intake in children with CF by evaluating the relations between energy intake (EI), energy expenditure (EE), fecal energy loss, nutritional status, and growth. DESIGN: The subjects were participants of a 24-mo prospective study of children 6-10 y of age with CF and PI. EE, EI, fecal energy loss, and anthropometric measures were obtained at baseline and at 24 mo. RESULTS: The children (n = 69) had suboptimal growth at baseline (x +/- SD: weight-for-age z score, -0.53 +/- 1.19; adjusted height-for-age z score, -0.67 +/- 1.06; body mass index z score, -0.29 +/- 1.12), and these variables remained suboptimal at 24 mo. The median ratios of EI to EE at baseline and 24 mo were 1.15 and 1.18, respectively, which decreased to 1.09 and 1.10, respectively, when adjusted for fecal energy loss (EI(-FL):EE). At baseline, 7% of subjects were underreporters, 64% were accurate reporters, and 23% were overreporters of energy intake; the percentages were similar at 24 mo. CONCLUSIONS: Although EI(-FL):EE ratios were higher than expected at both baseline and 24 mo, this cohort showed only age-appropriate weight gain. Self-reported dietary intake data at the individual level should be interpreted with caution, and weight gain velocity may serve as an objective measure of long-term energy balance.


Subject(s)
Child Nutrition Disorders/prevention & control , Cystic Fibrosis/metabolism , Energy Intake/physiology , Energy Metabolism/physiology , Growth/physiology , Anthropometry , Child , Child Development , Child Nutrition Disorders/etiology , Cohort Studies , Cystic Fibrosis/complications , Feces/chemistry , Female , Humans , Intestinal Absorption , Longitudinal Studies , Male , Nutritional Requirements , Nutritional Status , Prospective Studies , Weight Gain/physiology
19.
Cancer Res ; 66(15): 7824-31, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885387

ABSTRACT

The efficacy of ifosfamide (IFO), an antineoplastic drug, is severely limited by a high incidence of nephrotoxicity of unknown etiology. We hypothesized that inhibition of complex I (C-I) by chloroacetaldehyde (CAA), a metabolite of IFO, is the chief cause of nephrotoxicity, and that agmatine (AGM), which we found to augment mitochondrial oxidative phosphorylation and beta-oxidation, would prevent nephrotoxicity. Our model system was isolated mitochondria obtained from the kidney cortex of rats treated with IFO or IFO + AGM. Oxidative phosphorylation was determined with electron donors specific to complexes I, II, III, or IV (C-I, C-II, C-III, or C-IV, respectively). A parallel study was done with (13)C-labeled pyruvate to assess metabolic dysfunction. Ifosfamide treatment significantly inhibited oxidative phosphorylation with only C-I substrates. Inhibition of C-I was associated with a significant elevation of [NADH], depletion of [NAD], and decreased flux through pyruvate dehydrogenase and the TCA cycle. However, administration of AGM with IFO increased [cyclic AMP (cAMP)] and prevented IFO-induced inhibition of C-I. In vitro studies with various metabolites of IFO showed that only CAA inhibited C-I, even with supplementation with 2-mercaptoethane sulfonic acid. Following IFO treatment daily for 5 days with 50 mg/kg, the level of CAA in the renal cortex was approximately 15 micromol/L. Taken together, these observations support the hypothesis that CAA is accumulated in renal cortex and is responsible for nephrotoxicity. AGM may be protective by increasing tissue [cAMP], which phosphorylates NADH:oxidoreductase. The current findings may have an important implication for the prevention of IFO-induced nephrotoxicity and/or mitochondrial diseases secondary to defective C-I.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Ifosfamide/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Acetaldehyde/analogs & derivatives , Acetaldehyde/pharmacokinetics , Agmatine/pharmacology , Animals , Drug Interactions , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Ifosfamide/pharmacokinetics , Kidney Cortex/enzymology , Kidney Cortex/metabolism , Kidney Diseases/enzymology , Male , Oxidative Phosphorylation/drug effects , Rats
20.
J Biol Chem ; 281(22): 15064-72, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16574664

ABSTRACT

Glutamate dehydrogenase (GDH) plays an important role in insulin secretion as evidenced in children by gain of function mutations of this enzyme that cause a hyperinsulinism-hyperammonemia syndrome (GDH-HI) and sensitize beta-cells to leucine stimulation. GDH transgenic mice were generated to express the human GDH-HI H454Y mutation and human wild-type GDH in islets driven by the rat insulin promoter. H454Y transgene expression was confirmed by increased GDH enzyme activity in islets and decreased sensitivity to GTP inhibition. The H454Y GDH transgenic mice had hypoglycemia with normal growth rates. H454Y GDH transgenic islets were more sensitive to leucine- and glutamine-stimulated insulin secretion but had decreased response to glucose stimulation. The fluxes via GDH and glutaminase were measured by tracing 15N flux from [2-15N]glutamine. The H454Y transgene in islets had higher insulin secretion in response to glutamine alone and had 2-fold greater GDH flux. High glucose inhibited both glutaminase and GDH flux, and leucine could not override this inhibition. 15NH4Cl tracing studies showed 15N was not incorporated into glutamate in either H454Y transgenic or normal islets. In conclusion, we generated a GDH-HI disease mouse model that has a hypoglycemia phenotype and confirmed that the mutation of H454Y is disease causing. Stimulation of insulin release by the H454Y GDH mutation or by leucine activation is associated with increased oxidative deamination of glutamate via GDH. This study suggests that GDH functions predominantly in the direction of glutamate oxidation rather than glutamate synthesis in mouse islets and that this flux is tightly controlled by glucose.


Subject(s)
Glutamate Dehydrogenase/genetics , Insulin/metabolism , Mutation , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/drug effects , Glucose/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/metabolism , Glutamine/pharmacology , Guanosine Triphosphate/pharmacology , Humans , Hyperinsulinism/enzymology , Hyperinsulinism/genetics , Hyperinsulinism/physiopathology , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Kinetics , Leucine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...