Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Neurotherapeutics ; 18(2): 1151-1165, 2021 04.
Article in English | MEDLINE | ID: mdl-33782863

ABSTRACT

Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is a polyglutamine expansion disease arising from a trinucleotide CAG repeat expansion in exon 10 of the gene ATXN3. There are no effective pharmacological treatments for MJD, thus the identification of new pathogenic mechanisms, and the development of novel therapeutics is urgently needed. In this study, we performed a comprehensive, blind drug screen of 3942 compounds (many FDA approved) and identified small molecules that rescued the motor-deficient phenotype in transgenic ATXN3 Caenorhabditis elegans strain. Out of this screen, five lead compounds restoring motility, protecting against neurodegeneration, and increasing the lifespan in ATXN3-CAG89 mutant worms were identified. These compounds were alfacalcidol, chenodiol, cyclophosphamide, fenbufen, and sulfaphenazole. We then investigated how these molecules might exert their neuroprotective properties. We found that three of these compounds, chenodiol, fenbufen, and sulfaphenazole, act as modulators for TFEB/HLH-30, a key transcriptional regulator of the autophagy process, and require this gene for their neuroprotective activities. These genetic-chemical approaches, using genetic C. elegans models for MJD and the screening, are promising tools to understand the mechanisms and pathways causing neurodegeneration, leading to MJD. Positively acting compounds may be promising candidates for investigation in mammalian models of MJD and preclinical applications in the treatment of this disease.


Subject(s)
Ataxin-3/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Chenodeoxycholic Acid/administration & dosage , Phenylbutyrates/administration & dosage , Sulfaphenazole/administration & dosage , Animals , Animals, Genetically Modified , Ataxin-3/toxicity , Caenorhabditis elegans , Caenorhabditis elegans Proteins/toxicity , Drug Evaluation, Preclinical/methods , Longevity/drug effects , Longevity/physiology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics
2.
Mol Genet Metab ; 132(4): 220-226, 2021 04.
Article in English | MEDLINE | ID: mdl-33648834

ABSTRACT

Urea cycle disorders (UCDs), inborn errors of hepatocyte metabolism, result in the systemic accumulation of ammonia to toxic levels. Sodium 4-phenylbutyrate (NaPB), a standard therapy for UCDs for over 20 years, generates an alternative pathway of nitrogen deposition through glutamine consumption. Administration during or immediately after a meal is the accepted use of NaPB. However, this regimen is not based on clinical evidence. Here, an open-label, single-dose, five-period crossover study was conducted in healthy adults to investigate the effect of food on the pharmacokinetics of NaPB and determine any subsequent change in amino acid availability. Twenty subjects were randomized to one of four treatment groups. Following an overnight fast, NaPB was administered orally at 4.3 g/m2 (high dose, HD) or 1.4 g/m2 (low dose, LD) either 30 min before or just after breakfast. At both doses, compared with post-breakfast administration, pre-breakfast administration significantly increased systemic exposure of PB and decreased plasma glutamine availability. Pre-breakfast LD administration attenuated plasma glutamine availability to the same extent as post-breakfast HD administration. Regardless of the regimen, plasma levels of branched-chain amino acids (BCAA) were decreased below baseline in a dose-dependent manner. In conclusion, preprandial oral administration of NaPB maximized systemic exposure of the drug and thereby its potency to consume plasma glutamine. This finding may improve poor medication compliance because of the issues with odor, taste, and pill burden of NaPB and reduce the risk of BCAA deficiency in NaPB therapy.


Subject(s)
Eating/genetics , Pharmacokinetics , Phenylbutyrates/administration & dosage , Urea Cycle Disorders, Inborn/drug therapy , Administration, Oral , Adult , Amino Acids/genetics , Amino Acids, Branched-Chain/genetics , Biological Availability , Female , Glutamine/genetics , Healthy Volunteers , Humans , Male , Middle Aged , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/pathology , Young Adult
3.
Metab Brain Dis ; 36(5): 1015-1027, 2021 06.
Article in English | MEDLINE | ID: mdl-33620579

ABSTRACT

Maple syrup urine disease (MSUD) is a genetic disorder that leads the accumulation of branched-chain amino acids (BCAA) leucine (Leu), isoleucine, valine and metabolites. The symptomatology includes psychomotor delay and mental retardation. MSUD therapy comprises a lifelong protein strict diet with low BCAA levels and is well established that high concentrations of Leu and/or its ketoacid are associated with neurological symptoms. Recently, it was demonstrated that the phenylbutyrate (PBA) have the ability to decrease BCAA concentrations. This work aimed the development of lipid-based nanoparticles loaded with PBA, capable of targeting to the central nervous system in order to verify its action mechanisms on oxidative stress and cell death in brain of rats subjected to a MSUD chronic model. PBA-loaded nanoparticles treatment was effective in significantly decreasing BCAA concentration in plasma and Leu in the cerebral cortex of MSUD animals. Furthermore, PBA modulate the activity of catalase, superoxide dismutase, glutathione peroxidase and glutathione reductase enzymes, as well as preventing the oxidative damage to lipid membranes and proteins. PBA was also able to decrease the glial fibrillary acidic protein concentrations and partially decreased the reactive species production and caspase-3 activity in MSUD rats. Taken together, the data indicate that the PBA-loaded nanoparticles could be an efficient adjuvant in the MSUD therapy, protecting against oxidative brain damage and neuroinflammation.


Subject(s)
Amino Acids, Branched-Chain/blood , Cerebral Cortex/drug effects , Maple Syrup Urine Disease/metabolism , Nanoparticles/administration & dosage , Oxidative Stress/drug effects , Phenylbutyrates/administration & dosage , Animals , Catalase/metabolism , Cerebral Cortex/metabolism , Glutathione Peroxidase/metabolism , Maple Syrup Urine Disease/blood , Maple Syrup Urine Disease/chemically induced , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
4.
Nutrients ; 11(7)2019 Jul 21.
Article in English | MEDLINE | ID: mdl-31330899

ABSTRACT

Dysbiosis and a dysregulated gut immune barrier function contributes to chronic immune activation in HIV-1 infection. We investigated if nutritional supplementation with vitamin D and phenylbutyrate could improve gut-derived inflammation, selected microbial metabolites, and composition of the gut microbiota. Treatment-naïve HIV-1-infected individuals (n = 167) were included from a double-blind, randomized, and placebo-controlled trial of daily 5000 IU vitamin D and 500 mg phenylbutyrate for 16 weeks (Clinicaltrials.gov NCT01702974). Baseline and per-protocol plasma samples at week 16 were analysed for soluble CD14, the antimicrobial peptide LL-37, kynurenine/tryptophan-ratio, TMAO, choline, and betaine. Assessment of the gut microbiota involved 16S rRNA gene sequencing of colonic biopsies. Vitamin D + phenylbutyrate treatment significantly increased 25-hydroxyvitamin D levels (p < 0.001) but had no effects on sCD14, the kynurenine/tryptophan-ratio, TMAO, or choline levels. Subgroup-analyses of vitamin D insufficient subjects demonstrated a significant increase of LL-37 in the treatment group (p = 0.02), whereas treatment failed to significantly impact LL-37-levels in multiple regression analysis. Further, no effects on the microbiota was found in number of operational taxonomic units (p = 0.71), Shannon microbial diversity index (p = 0.82), or in principal component analyses (p = 0.83). Nutritional supplementation with vitamin D + phenylbutyrate did not modulate gut-derived inflammatory markers or microbial composition in treatment-naïve HIV-1 individuals with active viral replication.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/therapy , HIV-1 , Phenylbutyrates/pharmacology , Vitamin D/pharmacology , Adult , Anti-HIV Agents/administration & dosage , Dietary Supplements , Double-Blind Method , Female , Gastrointestinal Microbiome , Humans , Immunity, Innate , Male , Middle Aged , Phenylbutyrates/administration & dosage , Vitamin D/administration & dosage , Vitamin D/analogs & derivatives , Vitamin D/blood , Young Adult
5.
Mol Genet Metab ; 125(3): 251-257, 2018 11.
Article in English | MEDLINE | ID: mdl-30217721

ABSTRACT

INTRODUCTION: Glycerol phenylbutyrate (GPB) is approved in the US and EU for the chronic management of patients ≥2 months of age with urea cycle disorders (UCDs) who cannot be managed by dietary protein restriction and/or amino acid supplementation alone. GPB is a pre-prodrug, hydrolyzed by lipases to phenylbutyric acid (PBA) that upon absorption is beta-oxidized to the active nitrogen scavenger phenylacetic acid (PAA), which is conjugated to glutamine (PAGN) and excreted as urinary PAGN (UPAGN). Pharmacokinetics (PK) of GPB were examined to see if hydrolysis is impaired in very young patients who may lack lipase activity. METHODS: Patients 2 months to <2 years of age with UCDs from two open label studies (n = 17, median age 10 months) predominantly on stable doses of nitrogen scavengers (n = 14) were switched to GPB. Primary assessments included traditional plasma PK analyses of PBA, PAA, and PAGN, using noncompartmental methods with WinNonlin™. UPAGN was collected periodically throughout the study up to 12 months. RESULTS: PBA, PAA and PAGN rapidly appeared in plasma after GPB dosing, demonstrating evidence of GPB cleavage with subsequent PBA absorption. Median concentrations of PBA, PAA and PAGN did not increase over time and were similar to or lower than the values observed in older UCD patients. The median PAA/PAGN ratio was well below one over time, demonstrating that conjugation of PAA with glutamine to form PAGN did not reach saturation. Covariate analyses indicated that age did not influence the PK parameters, with body surface area (BSA) being the most significant covariate, reinforcing current BSA based dosing recommendations as seen in older patients. CONCLUSION: These observations demonstrate that UCD patients aged 2 months to <2 years have sufficient lipase activity to adequately convert the pre-prodrug GPB to PBA. PBA is then converted to its active moiety (PAA) providing successful nitrogen scavenging even in very young children.


Subject(s)
Glycerol/analogs & derivatives , Lipase/blood , Phenylbutyrates/administration & dosage , Prodrugs/administration & dosage , Urea Cycle Disorders, Inborn/drug therapy , Child , Child, Preschool , Female , Glutamine/blood , Glycerol/administration & dosage , Glycerol/blood , Glycerol/pharmacokinetics , Humans , Infant , Male , Nitrogen/blood , Nitrogen/metabolism , Phenylacetates/blood , Phenylbutyrates/blood , Phenylbutyrates/pharmacokinetics , Prodrugs/pharmacokinetics , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/pathology
6.
J Intern Med ; 284(3): 292-306, 2018 09.
Article in English | MEDLINE | ID: mdl-29696707

ABSTRACT

OBJECTIVE: Immunotherapy using vitamin D (vitD3 ) and phenylbutyrate (PBA) may support standard drug regimens used to treat infectious diseases. We investigated if vitD3 + PBA enhanced clinical recovery from pulmonary tuberculosis (TB). METHODS: A randomized controlled trial was conducted in Addis Ababa, Ethiopia. Patients with smear-positive or smear-negative TB received daily oral supplementation with 5000 IU vitD3 and 2 × 500 mg PBA or placebo for 16 weeks, together with 6-month chemotherapy. Primary end-point: reduction of a clinical composite TB score at week 8 compared with baseline using modified intention-to-treat (mITT, n = 348) and per-protocol (n = 296) analyses. Secondary end-points: primary and modified TB scores (week 0, 4, 8, 16, 24), sputum conversion, radiological findings and plasma 25(OH)D3 concentrations. RESULTS: Most subjects had low baseline plasma 25(OH)D3 levels that increased gradually in the vitD3 + PBA group compared with placebo (P < 0.0001) from week 0 to 16 (mean 34.7 vs. 127.4 nmol L-1 ). In the adjusted mITT analysis, the primary TB score was significantly reduced in the intervention group at week 8 (-0.52, 95% CI -0.93, -0.10; P = 0.015) while the modified TB score was reduced at week 8 (-0.58, 95% CI -1.02, -0.14; P = 0.01) and 16 (-0.34, 95% CI -0.64, -0.03; P = 0.03). VitD3 + PBA had no effect on longitudinal sputum-smear conversion (P = 0.98). Clinical adverse events were more common in the placebo group (24.3%) compared with the vitD3 + PBA group (12.6%). CONCLUSION: Daily supplementation with vitD3 + PBA may ameliorate clinical TB symptoms and disease-specific complications, while the intervention had no effect on bacterial clearance in sputum.


Subject(s)
Cholecalciferol/administration & dosage , Developing Countries , Phenylbutyrates/administration & dosage , Tuberculosis, Pulmonary/drug therapy , Administration, Oral , Adult , Antitubercular Agents/administration & dosage , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Treatment Outcome
7.
Appl Physiol Nutr Metab ; 41(10): 1108-1111, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27628198

ABSTRACT

We performed a placebo-controlled pre-clinical study to determine if sodium 4-phenylbutyrate (4PB) can reduce contraction-induced myofiber damage in the mdx mouse model of Duchenne muscular dystrophy (DMD). At 72 h post-eccentric contractions, 4PB significantly increased contractile torque and reduced myofiber damage and macrophage infiltration. We conclude that 4PB, which is approved by Health Canada (Pheburane) and the United States Food and Drug Administration (Buphenyl) for urea cycle disorders, might modify disease severity in patients with DMD.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Macrophage Activation/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Musculoskeletal Manipulations/adverse effects , Myofibrils/drug effects , Phenylbutyrates/therapeutic use , Animals , Hindlimb , Histone Deacetylase Inhibitors/administration & dosage , Injections, Intraperitoneal , Leg Injuries/prevention & control , Male , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Myofibrils/immunology , Myofibrils/pathology , Phenylbutyrates/administration & dosage , Torque
8.
J Lipid Res ; 50(12): 2486-501, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19461119

ABSTRACT

Recent studies have shown a link between obesity and endoplasmic reticulum (ER) stress. Perturbations in ER homeostasis cause ER stress and activation of the unfolded protein response (UPR). Adipocyte differentiation contributes to weight gain, and we have shown that markers of ER stress/UPR activation, including GRP78, phospho-eIF2, and spliced XBP1, are upregulated during adipogenesis. Given these findings, the objective of this study was to determine whether attenuation of UPR activation by the chemical chaperone 4-phenylbutyrate (4-PBA) inhibits adipogenesis. Exposure of 3T3-L1 preadipocytes to 4-PBA in the presence of differentiation media decreased expression of ER stress markers. Concomitant with the suppression of UPR activation, 4-PBA resulted in attenuation of adipogenesis as measured by lipid accumulation and adiponectin secretion. Consistent with these in vitro findings, female C57BL/6 mice fed a high-fat diet supplemented with 4-PBA showed a significant reduction in weight gain and had reduced fat pad mass, as compared with the high-fat diet alone group. Furthermore, 4-PBA supplementation decreased GRP78 expression in the adipose tissue and lowered plasma triglyceride, glucose, leptin, and adiponectin levels without altering food intake. Taken together, these results suggest that UPR activation contributes to adipogenesis and that blocking its activation with 4-PBA prevents adipocyte differentiation and weight gain in mice.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , DNA-Binding Proteins/chemistry , Phenylbutyrates/pharmacology , Protein Folding/drug effects , Transcription Factors/chemistry , 3T3-L1 Cells , Animals , Body Weight/drug effects , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Dietary Supplements , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Female , Mice , Mice, Inbred C57BL , Phenylbutyrates/administration & dosage , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , Transcription Factors/metabolism , X-Box Binding Protein 1
9.
Int J Artif Organs ; 18(3): 159-66, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7499019

ABSTRACT

The anaesthesiologist plays a central role in co-ordinating the combined application of the various blood saving techniques. In fact, to carry out transfusion therapy correctly, the anaesthesiologist must plan the right number of units of predeposit blood during the first examination, estimate the salvage of intra and post operative blood loss and spread the infusion of the units over the first three days in order to keep the patient in a state of haemodilution. From January 1992 to June 1994 in the department of anaesthesia and the intensive care unit, 980 patients were treated for total joint replacement: 714 total hips (7 after removal of plates and screws) 145 revisions, and 121 total knee prostheses. Basal Hb was 13.4 +/- 1.4 g/dl (range 6.7-17.9 g/dl). Homologous transfusions were carried out in 6.3% of these patients. The need to use homologous transfusions was negatively influenced by female sex, coronary heart disease (p = 0.005), length of surgery and type of antithromboembolic prophylaxis (indobufen has a significantly low incidence-p = 0.0001--compared to calcium heparin or low molecular weight heparin).


Subject(s)
Anesthesiology/methods , Blood Transfusion, Autologous/standards , Hemoglobins/metabolism , Adult , Aged , Aged, 80 and over , Anesthesiology/standards , Anticoagulants/administration & dosage , Anticoagulants/therapeutic use , Coronary Disease/physiopathology , Female , Hemodilution/standards , Hemoglobins/analysis , Heparin/administration & dosage , Heparin/therapeutic use , Hip Prosthesis/adverse effects , Humans , Intraoperative Complications/prevention & control , Isoindoles , Knee Prosthesis/adverse effects , Longitudinal Studies , Male , Middle Aged , Phenylbutyrates/administration & dosage , Phenylbutyrates/therapeutic use , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/therapeutic use , Postoperative Complications/prevention & control , Sex Factors , Thromboembolism/prevention & control
10.
J Pharm Pharmacol ; 46(12): 1017-22, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7714713

ABSTRACT

The anticonvulsant activity of 3-((+/-)-2-carboxypiperazin-4-yl)-1-propenyl-1-phosphonic acid (CPPene) and 2,3-dihydroxy-6-nitro-7-sulphamoyl-benzo(F)quinoxoline (NBQX), two excitatory amino acid antagonists, was studied against audiogenic seizures in DBA/2 mice, following intraperitoneal (i.p.) or intracerebroventricular (i.c.v.) administration. Maximal anticonvulsant protection was observed 15-30 min following NBQX and 45-180 min after CPPene. Coadministration with fenbufen (20 mg kg-1, i.p.), a non-steroidal anti-inflammatory agent, enhanced and prolonged the anticonvulsant actions of CPPene and NBQX and also potentiated and prolonged the impairment of rotarod performance. The enhancement of the anticonvulsant activity and the prolonged impairment of rotarod performance suggests that fenbufen may have some pharmacokinetic interactions with CPPene and NBQX and that fenbufen is able to increase the brain levels of these excitatory amino acid antagonists. In particular, fenbufen was able to exert a major degree of potentiation of effects of NBQX rather than those of CPPene, suggesting that the chemical structures of these excitatory amino acid antagonists are responsible for the different degree of interactions between CPPene or NBQX and fenbufen. NBQX appears to have a notable similarity with quinolones whilst CPPene does not. Additionally fenbufen may displace CPPene and NBQX from plasma binding sites or inhibit the renal excretion. The present data are also consistent with previous studies showing pharmacokinetic interactions between fenbufen and quinolones.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticonvulsants/pharmacology , Phenylbutyrates/pharmacology , Piperazines/pharmacology , Quinoxalines/pharmacology , Acoustic Stimulation , Analysis of Variance , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Drug Synergism , Injections, Intraperitoneal , Injections, Intraventricular , Mice , Motor Activity/drug effects , Phenylbutyrates/administration & dosage , Piperazines/administration & dosage , Piperazines/therapeutic use , Quinoxalines/administration & dosage , Quinoxalines/therapeutic use , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/drug therapy
11.
Res Rep Health Eff Inst ; (49): 1-35, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1315139

ABSTRACT

To investigate the relative irritant potencies of inhaled aldehydes, guinea pigs were exposed to formaldehyde or acrolein and specific total pulmonary resistance and bronchial reactivity to intravenous acetylcholine were assessed. The mechanisms associated with these responses were investigated by analyzing morphologic and biochemical changes in airway epithelial cells after in vivo and in vitro exposures. Immediately after exposure to formaldehyde or acrolein, specific resistance increased transiently and returned to control values within 30 to 60 minutes. Bronchial hyperreactivity, assessed by the acetylcholine dose necessary to double resistance, increased and became maximal two to six hours after exposure to at least 9 parts per million2 (ppm) formaldehyde or at least 1 ppm acrolein for two hours. The effect of exposure to 3 ppm formaldehyde for two hours was less than the effect of exposure to 1 ppm formaldehyde for eight hours; thus, extended exposures produced a disproportionate heightening of bronchial reactivity. Bronchial hyperreactivity often persisted for longer than 24 hours. Increases in three bronchoconstrictive eicosanoids, prostaglandin F2 alpha, thromboxane B2, and leukotriene C4, occurred immediately after exposure, whereas an influx of neutrophils into lavage fluid occurred 24 hours later. Histological examination of the tracheal epithelium and lamina propria also demonstrated a lack of inflammatory cell infiltration. Treatment with leukotriene synthesis inhibitors and receptor antagonists inhibited acrolein-induced hyperreactivity, supporting a causal role for these compounds in this response. Acrolein also stimulated eicosanoid release from bovine epithelial cells in culture. However, the profile of metabolites formed differed from that found in lavage fluid after in vivo exposure. Similarly, human airway epithelial cells did not produce cysteinyl leukotriene or thromboxane B2. However, cysteinyl leukotrienes were mitogenic for human airway epithelial cells in a concentration-dependent manner and exhibited a structure-activity relationship; leukotriene C4 was more potent than its sequential metabolites D4 and E4. The potency of leukotriene C4 was striking, stimulating colony-forming efficiency in concentrations as low as 0.01 pM. Together, these findings suggest that environmentally relevant concentrations of aldehydes can induce bronchial hyperreactivity in guinea pigs through a mechanism involving injury to cells present in the airways during exposure (rather than from subsequently recruited migratory cells) and that this response is dependent on leukotriene biosynthesis.


Subject(s)
Acrolein/toxicity , Air Pollutants/toxicity , Airway Resistance/drug effects , Bronchial Hyperreactivity/chemically induced , Formaldehyde/toxicity , Acetylcholine/administration & dosage , Acetylcholine/pharmacology , Acetylcholine/therapeutic use , Airway Resistance/physiology , Animals , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epithelium/injuries , Epithelium/metabolism , Epithelium/pathology , Epoprostenol/administration & dosage , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Guinea Pigs , Hyperplasia , Inflammation , Leukocyte Count , Leukotriene B4/biosynthesis , Leukotriene B4/chemistry , Leukotriene B4/metabolism , Lipoxygenase Inhibitors/administration & dosage , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/therapeutic use , Neutrophils/chemistry , Neutrophils/metabolism , Phenothiazines/administration & dosage , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Phenylbutyrates/administration & dosage , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Prostaglandins F/biosynthesis , Prostaglandins F/chemistry , Prostaglandins F/metabolism , SRS-A/antagonists & inhibitors , Thromboxane B2/biosynthesis , Thromboxane B2/chemistry , Thromboxane B2/metabolism , Time Factors
12.
Pediatr Res ; 29(2): 147-50, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2014149

ABSTRACT

Phenylacetylglutamine (PAG), the amino acid acetylation product of phenylacetate (or phenylbutyrate after beta-oxidation) was evaluated as a waste nitrogen product in patients with inborn errors of urea synthesis. A boy with carbamyl phosphate synthetase deficiency receiving a low nitrogen intake excreted 80-90% of administered phenylacetate or phenylbutyrate as PAG. The amount of PAG nitrogen excreted varied from 38-44% of his dietary nitrogen, similar to the relationship between urea nitrogen and dietary nitrogen found in normal subjects receiving low dietary nitrogen. With few exceptions, neither phenylacetate nor phenylbutyrate accumulated in plasma. Treatment with relatively high dose phenylacetate or phenylbutyrate (0.5-0.6 g/kg/d) resulted in normal daytime levels of glutamine. These data suggest that PAG may replace urea as a waste nitrogen product when phenylbutyrate is administered at a dose that yields PAG nitrogen excretion equal to 40-44% of a low nitrogen intake.


Subject(s)
Glutamine/analogs & derivatives , Metabolism, Inborn Errors/urine , Nitrogen/urine , Urea/urine , Adolescent , Child , Child, Preschool , Female , Glutamine/urine , Humans , Infant , Male , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/metabolism , Phenylacetates/administration & dosage , Phenylacetates/metabolism , Phenylbutyrates/administration & dosage , Phenylbutyrates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL