Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Pediatr ; 188: 163-172, 2017 09.
Article in English | MEDLINE | ID: mdl-28629683

ABSTRACT

OBJECTIVES: To examine relationships between dietary intake, growth and body composition patterns in patients with inborn errors of intermediary protein metabolism and to determine a safe protein:energy ratio (P:E ratio) associated with optimal growth outcomes. STUDY DESIGN: Retrospective longitudinal data of growth and dietary intake in patients (n = 75) with isovaleric acidemia (IVA; n = 7), methylmalonic acidemia/propionic acidemia (MMA/PA; n = 14), urea cycle defects (UCD; n = 44), classical maple syrup urine disease (MSUD; n = 10) were collected. Prospective longitudinal data of growth, dietary intake, and body composition from 21 patients: IVA (n = 5), MMA/PA (n = 6), UCD (n = 7), and MSUD (n = 3) were collected at clinic visits. RESULTS: Fifty-two of 75 (66%), 49 of 74 (68%), and 44 of 65 (68%) patients had a z-score of 0 (±1) for lifetime weight, height, and body mass index, respectively. Patients with MMA/PA had the lowest median height and weight z-scores, and MSUD patients had highest median body mass index z-score at all ages. In IVA, MMA/PA, and UCD, total natural protein intake met or exceeded the Food and Agriculture Organization of the United Nations (FAO)/World Health Organization (WHO)/United Nations University (UNU) recommended safe levels. Median percentage fat mass was 17.6% in IVA, 20.7% in MMA/PA, 19.4% in UCD, and 17.8% in MSUD. There was a significant negative correlation between percentage fat mass and total protein intake in IVA, MMA/PA, and UCD (r = -0.737; P = .010). The correlation between the P:E ratio and growth variables in IVA, MMA/PA, and UCD suggest a safe P:E ratio (>1.5 to < 2.9) g protein:100 kcal/day. CONCLUSION: Growth outcomes in inborn errors of intermediary protein metabolism are not always ideal. Most patients with IVA, MMA/PA, and UCD consume sufficient natural protein to meet FAO/WHO/UNU recommendations. A P:E ratio range of (>1.5 to < 2.9)g protein/100 kcal/day correlates with optimal growth outcomes.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Body Composition/physiology , Energy Intake/physiology , Adolescent , Body Fat Distribution , Body Height/physiology , Body Mass Index , Body Weight/physiology , Child , Child, Preschool , Dietary Proteins/administration & dosage , Female , Humans , Longitudinal Studies , Male , Retrospective Studies
2.
J Inherit Metab Dis ; 38(6): 1021-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26041581

ABSTRACT

3-Hydroxy-3-methylglutaryl-CoA lyase (HL) deficiency is an inherited disorder of organic acid metabolism biochemically characterized by tissue accumulation and high urinary excretion of 3-hydroxy-3-methylgutarate, 3-methylglutarate, 3-methylglutaconate and 3-hydroxyisovalerate. Affected patients predominantly present neurological symptoms that are accompanied by mild hepatopathy during episodes of catabolic crisis. The pathophysiology of this disease is poorly known, although recent animal and human in vitro and in vivo studies have suggested that oxidative stress caused by the major accumulating organic acids may represent a pathomechanism of brain and liver damage in HL deficiency. In this review we focus on the deleterious effects of these carboxylic acids on redox homeostasis in rat and human tissues that may offer new perspectives for potential novel adjuvant therapeutic strategies in this disorder.


Subject(s)
Acetyl-CoA C-Acetyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/physiopathology , Brain/pathology , Homeostasis , Liver/pathology , Oxidative Stress , Animals , Disease Models, Animal , Glutarates/urine , Humans , Mice , Oxidation-Reduction , Rats
3.
Gene ; 531(2): 191-8, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24035933

ABSTRACT

Sulfite oxidase (SO) deficiency is biochemically characterized by tissue accumulation and high urinary excretion of sulfite, thiosulfate and S-sulfocysteine. Affected patients present severe neurological symptoms and cortical atrophy, whose pathophysiology is still poorly established. Therefore, in the present work we investigated the in vitro effects of sulfite and thiosulfate on important parameters of energy metabolism in the brain of young rats. We verified that sulfite moderately inhibited the activity of complex IV, whereas thiosulfate did not alter any of the activities of the respiratory chain complexes. It was also found that sulfite and thiosulfate markedly reduced the activity of total creatine kinase (CK) and its mitochondrial and cytosolic isoforms, suggesting that these metabolites impair brain cellular energy buffering and transfer. In contrast, the activity of synaptic Na(+),K(+)-ATPase was not altered by sulfite or thiosulfate. We also observed that the inhibitory effect of sulfite and thiosulfate on CK activity was prevented by melatonin, reduced glutathione and the combination of both antioxidants, as well as by the nitric oxide synthase N(ω)-nitro-l-arginine methyl ester, indicating the involvement of reactive oxygen and nitrogen species in these effects. Sulfite and thiosulfate also increased 2',7'-dichlorofluorescin oxidation and hydrogen peroxide production and decreased the activity of the redox sensor aconitase enzyme, reinforcing a role for oxidative damage in the effects elicited by these metabolites. It may be presumed that the disturbance of cellular energy and redox homeostasis provoked by sulfite and thiosulfate contributes to the neurological symptoms and abnormalities found in patients affected by SO deficiency.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Brain Diseases, Metabolic/etiology , Brain/drug effects , Energy Metabolism/drug effects , Homeostasis/drug effects , Sulfite Oxidase/deficiency , Sulfites/pharmacology , Thiosulfates/pharmacology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain/metabolism , Brain/pathology , Brain/physiology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Electron Transport/drug effects , Electron Transport/genetics , Electron Transport/physiology , Energy Metabolism/physiology , Male , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Sulfite Oxidase/genetics , Sulfite Oxidase/metabolism , Sulfites/metabolism , Thiosulfates/metabolism
4.
Mol Cell Biochem ; 362(1-2): 225-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22081291

ABSTRACT

It is known that the accumulation of tryptophan and its metabolites is related to brain damage associated with both hypertryptophanemia and neurodegenerative diseases. In this study, we investigated the effect of tryptophan administration on various parameters of behavior in the open-field task and oxidative stress, and the effects of creatine and pyruvate, on the effect of tryptophan. Forty, 60-day-old male Wistar rats, were randomly divided into four groups: saline, tryptophan, pyruvate + creatine, tryptophan + pyruvate + creatine. Animals received three subcutaneous injections of tryptophan (2 µmol/g body weight each one at 3 h of intervals) and/or pyruvate (200 µg/g body weight 1 h before tryptophan), and/or creatine (400 µg/g body weight twice a day for 5 days before tryptophan twice a day for 5 days before training); controls received saline solution (NaCl 0.85%) at the same volumes (30 µl/g body weight) than the other substances. Results showed that tryptophan increased the activity of the animals, suggesting a reduction in the ability of habituation to the environment. Tryptophan induced increase of TBA-RS and total sulfhydryls. The effects of tryptophan in the open field, and in oxidative stress were fully prevented by the combination of creatine plus pyruvate. In case these findings also occur in humans affected by hypertryptophanemia or other neurodegenerative disease in which tryptophan accumulates, it is feasible that oxidative stress may be involved in the mechanisms leading to the brain injury, suggesting that creatine and pyruvate supplementation could benefit patients affected by these disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Behavior, Animal/drug effects , Creatine/pharmacology , Pyruvic Acid/pharmacology , Tryptophan/pharmacology , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/psychology , Animals , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar
5.
Neurochem Res ; 37(1): 205-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21935728

ABSTRACT

This study investigated the effects of acute and chronic hyperprolinemia on glutamate uptake, as well as some mechanisms underlying the proline effects on glutamatergic system in rat cerebral cortex. The protective role of guanosine on effects mediated by proline was also evaluated. Results showed that acute and chronic hyperprolinemia reduced glutamate uptake, Na(+), K(+)-ATPase activity, ATP levels and increased lipoperoxidation. GLAST and GLT-1 immunocontent were increased in acute, but not in chronic hyperprolinemic rats. Our data suggest that the effects of proline on glutamate uptake may be mediated by lipid peroxidation and disruption of Na(+), K(+)-ATPase activity, but not by decreasing in glutamate transporters. This probably induces excitotoxicity and subsequent energy deficit. Guanosine was effective to prevent most of the effects promoted by proline, reinforcing its modulator role in counteracting the glutamate toxicity. However, further studies are needed to assess the modulatory effects of guanosine on experimental hyperprolinemia.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Brain/physiopathology , Glutamic Acid/metabolism , Guanosine/pharmacology , Homeostasis , Neuroprotective Agents/pharmacology , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Proline Oxidase/deficiency , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
6.
Neurochem Res ; 36(12): 2306-15, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21792675

ABSTRACT

This study investigated whether physical exercise would reverse proline-induced performance deficits in water maze tasks, as well as its effects on brain-derived neurotrophic factor (BDNF) immunocontent and brain acetylcholinesterase (AChE) activity in Wistar rats. Proline administration followed partial time (6th-29th day of life) or full time (6th-60th day of life) protocols. Treadmill exercise was performed from 30th to 60th day of life, when behavioral testing was started. After that, animals were sacrificed for BDNF and AChE determination. Results show that proline impairs cognitive performance, decreases BDNF in cerebral cortex and hippocampus and increases AChE activity in hippocampus. All reported effects were prevented by exercise. These results suggest that cognitive, spatial learning/memory, deficits caused by hyperprolinemia may be associated, at least in part, to the decrease in BDNF levels and to the increase in AChE activity, as well as support the role of physical exercise as a potential neuroprotective strategy.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Cognition Disorders/therapy , Maze Learning/drug effects , Memory/drug effects , Physical Conditioning, Animal , 1-Pyrroline-5-Carboxylate Dehydrogenase/deficiency , Acetylcholinesterase/metabolism , Amino Acid Metabolism, Inborn Errors/psychology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Proline Oxidase/deficiency , Rats , Rats, Wistar
7.
J Bioenerg Biomembr ; 43(1): 31-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21249436

ABSTRACT

Organic acidurias or organic acidemias constitute a group of inherited disorders caused by deficient activity of specific enzymes of amino acids, carbohydrates or lipids catabolism, leading to large accumulation and excretion of one or more carboxylic (organic) acids. Affected patients usually present neurologic symptoms and abnormalities, sometimes accompanied by cardiac and skeletal muscle alterations, whose pathogenesis is poorly known. However, in recent years growing evidence has emerged indicating that mitochondrial dysfunction is directly or indirectly involved in the pathology of various organic acidemias. Mitochondrial impairment in some of these diseases are generally due to mutations in nuclear genes of the tricarboxylic acid cycle or oxidative phosphorylation, while in others it seems to result from toxic influences of the endogenous organic acids to the mitochondrion. In this minireview, we will briefly summarize the present knowledge obtained from human and animal studies showing that disruption of mitochondrial homeostasis may represent a relevant pathomechanism of tissue damage in selective organic acidemias. The discussion will focus on mitochondrial alterations found in patients affected by organic acidemias and by the deleterious effects of the accumulating organic acids on mitochondrial pathways that are crucial for ATP formation and transfer. The elucidation of the mechanisms of toxicity of these acidic compounds offers new perspectives for potential novel adjuvant therapeutic strategies in selected disorders of this group.


Subject(s)
Adenosine Triphosphate/metabolism , Carboxylic Acids/metabolism , Homeostasis/physiology , Metabolism, Inborn Errors/physiopathology , Mitochondria/physiology , Mitochondrial Diseases/physiopathology , Acetyl-CoA C-Acyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Barth Syndrome/physiopathology , Brain Diseases, Metabolic/physiopathology , Brain Diseases, Metabolic, Inborn/physiopathology , Glutaryl-CoA Dehydrogenase/deficiency , Humans , Mitochondria/metabolism , Propionic Acidemia/physiopathology , Purpura/physiopathology
8.
J Pediatr ; 154(4): 551-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19058814

ABSTRACT

OBJECTIVES: To describe 3 patients with the cblD disorder, a rare inborn error of cobalamin metabolism caused by mutations in the MMADHC gene that can result in isolated homocystinuria, isolated methylmalonic aciduria, or combined homocystinuria and methylmalonic aciduria. STUDY DESIGN: Patient clinical records were reviewed. Biochemical and somatic cell genetic studies were performed on cultured fibroblasts. Sequence analysis of the MMADHC gene was performed on patient DNA. RESULTS: Patient 1 presented with isolated methylmalonic aciduria, patient 3 with isolated homocystinuria, and patient 2 with combined methylmalonic aciduria and homocystinuria. Studies of cultured fibroblasts confirmed decreased synthesis of adenosylcobalamin in patient 1, decreased synthesis of methylcobalamin in patient 3, and decreased synthesis of both cobalamin derivatives in patient 2. The diagnosis of cblD was established in each patient by complementation analysis. Mutations in the MMADHC gene were identified in all patients. CONCLUSIONS: The results emphasize the heterogeneous clinical, cellular and molecular phenotype of the cblD disorder. The results of molecular analysis of the MMADHC gene are consistent with the hypothesis that mutations affecting the N terminus of the MMADHC protein are associated with methylmalonic aciduria, and mutations affecting the C terminus are associated with homocystinuria.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Cobamides/deficiency , Vitamin B 12 Deficiency/diagnosis , Vitamin B 12 Deficiency/genetics , Amino Acid Metabolism, Inborn Errors/physiopathology , Cells, Cultured , Family Health , Female , Fibroblasts/metabolism , Homocystinuria/genetics , Homocystinuria/physiopathology , Humans , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Membrane Transport Proteins/genetics , Methylmalonic Acid/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/genetics , Phenotype , Vitamin B 12 Deficiency/physiopathology
9.
Metab Brain Dis ; 23(2): 221-33, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18425567

ABSTRACT

Despite the significant brain abnormalities, the neurotoxic mechanisms of brain injury in hypertryptophanemia are virtually unknown. In this work, we determined the thiobarbituric acid-reactive substances, 2',7'-dihydrodichlorofluorescein oxidation, reduced glutathione and the activities of catalase, superoxide dismutase and glutathione peroxidase in cerebral cortex from rats loaded with L-tryptophan. High L-tryptophan concentrations, similar to those found in hypertryptophanemic patients were induced by three subcutaneous injections of saline-buffered tryptophan (2 micromol/g body weight) to 30-day-old Wistar rats. The parameters were assessed 1 h after the last injection. It was observed that tryptophan significantly increased thiobarbituric acid-reactive substances, 2',7'-dihydrodichlorofluorescein oxidation and reduced glutathione, whereas it reduced catalase activity. Pre-treatment with taurine (1.6 micromol/g of body weight), or alpha-tocopherol plus ascorbic acid (40 and 100 microg/g body weight, respectively) prevented those effects of tryptophan, reinforcing the hypothesis that tryptophan induces oxidative stress in brain cortex of the rats. Therefore, these findings also occur in human hypertryptophanemia or in other neurodegenerative diseases in which tryptophan accumulates, then oxidative stress may be involved in the mechanisms leading to the brain injury observed in patients affected by these disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Cerebral Cortex/metabolism , Nerve Degeneration/metabolism , Oxidative Stress/physiology , Tryptophan/metabolism , Amino Acid Metabolism, Inborn Errors/chemically induced , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Antioxidants/pharmacology , Brain Diseases, Metabolic/chemically induced , Brain Diseases, Metabolic/physiopathology , Catalase/metabolism , Cell Death/drug effects , Cell Death/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Taurine/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism , Tryptophan/toxicity , alpha-Tocopherol/pharmacology
10.
Rev Neurol ; 44(10): 610-5, 2007.
Article in Spanish | MEDLINE | ID: mdl-17523120

ABSTRACT

INTRODUCTION: Glutaric aciduria type I is an autosomal recessive inborn error of metabolism that is due to a deficiency of the enzyme glutaryl-CoA dehydrogenase, which gives rise to an accumulation of glutaric and 3-hydroxyglutaric acids in biological fluids. Clinical features present as a sudden-onset severe neurological disorder, characterised by extrapyramidal signs (dystonia-dyskinesia), hypotonia, irritability, macrocephaly and degeneration of the basal ganglia; it may also manifest with unspecific symptoms, such as hypotonia and psychomotor retardation. AIMS: To describe the clinical, biochemical, neuroimaging and molecular aspects in six Venezuelan patients and to highlight the importance of an early diagnosis of glutaric aciduria type I so as to be able to establish early treatment and thus prevent the neurological damage produced by this disease. CASE REPORTS: Two patients were referred because of macrocephaly, hypotonia and psychomotor retardation, and four more following an encephalopathic crisis. In all of them, neuroimaging studies showed delays in myelination, bilateral frontotemporal hypoplasia and symmetric widening of the Sylvian fissures with poor opercularisation. Urinary organic acid analyses showed raised levels of glutaric and 3-hydroxyglutaric acids, and a molecular analysis confirmed the diagnosis. CONCLUSIONS: Organic acid analysis should be indicated in all patients who present macrocephaly, hypotonia, psychomotor retardation or an encephalopathic crisis of unknown causation. This study allowed us to determine the behaviour of the disease in Venezuela, since no epidemiological data exist in the country.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Glutarates/urine , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Brain/pathology , Child , Child, Preschool , Chromosome Aberrations , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Venezuela
11.
Neurochem Int ; 49(1): 87-93, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16497412

ABSTRACT

Despite the significant brain abnormalities, the neurotoxic mechanisms of brain injury in hypertryptophanemia are virtually unknown. In this work, it was investigated the in vitro effect of l-tryptophan on various parameters of oxidative stress, namely spontaneous chemiluminescence, thiobarbituric acid-reactive substances (TBA-RS), total radical-trapping antioxidant potential (TRAP), total antioxidant reactivity (TAR) and glutathione (GSH) levels in cerebral cortex from 30-day-old rats. Tryptophan significantly increased chemiluminescence and TBA-RS measurements indicating that this amino acid induced lipid peroxidation in vitro. We also observed that tryptophan significantly decreased the brain antioxidant defenses by reducing the values of TRAP, TAR and GSH, reflecting that the overall content of antioxidants was reduced by tryptophan. Furthermore, the tryptophan-induced increase of TBA-RS was fully prevented by GSH and by combination of catalase plus superoxide dismutase, but not by the inhibitor of nitric oxide synthase N(omega)-nitro-L-arginine methyl ester (L-NAME). In case these findings also occur in human hypertryptophanemia or in other neurodegenerative diseases in which tryptophan accumulates, it is feasible that oxidative stress may be involved in the mechanism leading to the brain injury observed in patients affected by these disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Cerebral Cortex/metabolism , Nerve Degeneration/metabolism , Oxidative Stress/physiology , Tryptophan/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Antioxidants/metabolism , Catalase/metabolism , Catalase/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Free Radicals/metabolism , Glutathione/metabolism , Glutathione/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Luminescence , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Thiobarbituric Acid Reactive Substances/analysis , Thiobarbituric Acid Reactive Substances/metabolism , Tryptophan/toxicity
12.
Int J Dev Neurosci ; 23(6): 509-14, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16023820

ABSTRACT

Considering that pyruvate kinase activity, a crucial enzyme for glucose metabolism and energy liberation in brain, may be regulated by some amino acids, it is possible that diminution of this enzyme activity may contribute to the brain damage caused by amino acids accumulated in metabolic diseases, such as phenylalanine, tryptophan and cystine. Therefore, the present study was undertaken to investigate the effect of these amino acids on pyruvate kinase activity in the brain cortex of rats. We also investigated the effect of serine and alanine on pyruvate kinase activity in the same tissue. The results suggested that phenylalanine, tryptophan, cystine, alanine, and serine act at the same site on the enzyme, phenylalanine, tryptophan, and cystine causing inhibition, and alanine and serine preventing this effect. Cystine also inhibited the enzyme activity through a different mechanism, possibly acting on the enzyme thiol groups. Considering that this enzyme is a target for amino acids accumulated in some metabolic diseases of amino acid metabolism, it is possible that its inhibition may contribute to the brain damage found in these diseases.


Subject(s)
Amino Acids/metabolism , Cerebral Cortex/enzymology , Energy Metabolism/physiology , Pyruvate Kinase/metabolism , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acids/pharmacology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/physiopathology , Cerebral Cortex/drug effects , Cystine/metabolism , Cystine/pharmacology , Energy Metabolism/drug effects , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Phenylalanine/metabolism , Phenylalanine/pharmacology , Pyruvate Kinase/drug effects , Rats , Rats, Wistar , Serine/metabolism , Serine/pharmacology , Tryptophan/metabolism , Tryptophan/pharmacology
13.
Metab Brain Dis ; 20(1): 73-80, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15918552

ABSTRACT

In the present study we investigated the effect of chronic proline (Pro) administration on rat performance in the Morris water maze task. Rats received s.c. injections of Pro twice a day at 8 h intervals from the 6th to the 28th days of age and equivalent volume of 0.9% saline solution (control). On the 60th day of life, rats were subjected to the water maze task. Results showed that chronic Pro administration provokes impairment on spatial learning, as shown by the increase of latency in acquisition and retention and by a reduced efficiency to find the platform position in the working memory test. Present results suggest that hyperprolininemia causes cognitive dysfunction and might be relevant to explain, at least in part, the neurological dysfunction associated with hyperprolinemia.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Brain Diseases, Metabolic, Inborn/physiopathology , Maze Learning/physiology , Memory Disorders/physiopathology , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Brain Diseases, Metabolic, Inborn/metabolism , Chronic Disease , Disease Models, Animal , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Proline/administration & dosage , Proline/metabolism , Rats , Rats, Wistar , Reaction Time/drug effects , Reaction Time/physiology
14.
J Inherit Metab Dis ; 27(6): 825-8, 2004.
Article in English | MEDLINE | ID: mdl-15505388

ABSTRACT

Although the precise mechanisms underlying the CNS degeneration of patients with glutaryl-CoA dehydrogenase (GCDH) deficiency are still the subject of intense debate, many studies have highlighted that excitotoxicity plays a fundamental role in the neuropathology of this disease, particularly involving the N-methyl-D-aspartate receptor subtype of ionotropic glutamate receptors. Modulation of the glutamatergic system by these compounds involves an inhibition of glutamate uptake into synaptosomes and synaptic vesicles, and a decrease in glutamate binding. Furthermore, glutaric and 3-hydroxyglutaric acids inhibit glutamate decarboxylase, the key enzyme of GABA synthesis, and striatal GABAergic medium-spiny neurons are highly vulnerable to 3-hydroxyglutaric acid-induced neurotoxicity. In conclusion, glutaric acid and 3-hydroxyglutaric acid induce an imbalance in glutamatergic and GABAergic neurotransmission.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Glutamic Acid/physiology , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/physiology , Animals , Glutaryl-CoA Dehydrogenase , Humans , Neurotoxins/metabolism
15.
J Pediatr ; 144(4): 532-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15069406

ABSTRACT

We hypothesized that gut motility likely plays a critical role in the metabolic stability in propionic acidemia (PA). Therefore, 4 known patients with PA (aged 47 months to 185 months) were prospectively studied over 7 days in the Clinical Research Center at Children's Hospital, Boston. Determinations of ammonia, bicarbonate, and amino acids in blood; organic acids and propionylglycine in urine; and a lactulose breath test were conducted under two study conditions: on regular therapy (for 4 days) and on regular therapy plus Senekot (Purdue Frederick Company, Norwalk, Conn), an intestinal motility agent (for 3 days). The total gastrointestinal transit time was calculated using 20 nonabsorbable, inert, radio-opaque markers. The addition of an intestinal motility agent resulted in a significant decrease in blood ammonia, urinary excretion of propionylglycine, and a rise in the ratio of free to total carnitine over baseline. We concluded that enhancement of gut motility can improve metabolic stability in patients with PA.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/therapy , Gastrointestinal Motility/physiology , Glycine/analogs & derivatives , Propionates/blood , Adolescent , Amino Acid Metabolism, Inborn Errors/physiopathology , Ammonia/blood , Carnitine/analysis , Cathartics/administration & dosage , Child , Child, Preschool , Glycine/urine , Humans , Prospective Studies , Senna Extract/administration & dosage
16.
Int J Dev Neurosci ; 22(2): 67-72, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15036381

ABSTRACT

Histidinemia is an inherited metabolic disorder caused by deficiency of histidase activity, which leads to tissue accumulation of histidine and its derivatives. Affected patients usually present with speech delay and mental retardation, although asymptomatic patients have been reported. Considering that the pathophysiology of the neurological dysfunction of histidinemia is not yet understood and since histidine has been considered a pro-oxidant agent, in the present study we investigated the effect of histidine and one of its derivatives, l-beta-imidazolelactic acid, at concentrations ranging from 0.1 to 10 mM, on various parameters of oxidative stress in cerebral cortex of 30-day-old Wistar rats. Chemiluminescence, total radical-trapping antioxidant potential (TRAP), thiobarbituric acid reactive substances (TBA-RS), and the activities of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px) were measured in tissue homogenates in the presence of l-histidine or l-beta-imidazolelactic acid. We observed that l-histidine provoked an increase of chemiluminescence and a reduction of TRAP at concentrations of 2.5 mM and higher, while TBA-RS measurement, GSH-Px, CAT and SOD activities were not affected. Furthermore, l-beta-imidazolelactic acid provoked antioxidant effects at high concentrations (5-10 mM) as observed by the reduction of chemiluminescence, although this compound enhanced chemiluminescence at low concentrations (0.5-1 mM). These results suggest that in vitro oxidative stress is elicited by histidine but only at supraphysiological concentrations.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Cerebral Cortex/metabolism , Histidine/metabolism , Oxidative Stress/physiology , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain Diseases, Metabolic, Inborn/physiopathology , Catalase/drug effects , Catalase/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Free Radicals/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Histidine/pharmacology , Imidazoles/pharmacology , Lactates/pharmacology , Luminescent Measurements , Oxidants/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
17.
Int J Dev Neurosci ; 22(2): 95-101, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15036384

ABSTRACT

Hypertryptophanemia is a rare inherited metabolic disorder probably caused by a blockage in the conversion of tryptophan to kynurenine, resulting in the accumulation of tryptophan and some of its metabolites in plasma and tissues of affected patients. The patients present mild-to-moderate mental retardation with exaggerated affective responses, periodic mood swings, and apparent hypersexual behavior. Creatine kinase plays a key role in energy metabolism of tissues with intermittently high and fluctuating energy requirements, such as nervous tissue. The main objective of the present study was to investigate the effect of acute administration of tryptophan on creatine kinase activity in brain cortex of Wistar rats. We also studied the in vitro effect of this amino acid on creatine kinase activity in the brain cortex of non-treated rats. The results indicated that tryptophan inhibits creatine kinase in vitro and in vivo. We also observed that the in vitro inhibition was fully prevented but not reversed by pre-incubation with reduced glutathione, suggesting that the inhibitory effect of tryptophan on CK activity is possibly mediated by oxidation of essential thiol groups of the enzyme and/or long-lasting adduct formation. Considering the importance of creatine kinase for the maintenance of energy homeostasis in the brain, it is conceivable that an inhibition of this enzyme activity in the brain may be one of the mechanisms by which tryptophan might be neurotoxic.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Brain Diseases, Metabolic, Inborn/enzymology , Cerebral Cortex/enzymology , Creatine Kinase/metabolism , Down-Regulation/physiology , Tryptophan/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain Diseases, Metabolic, Inborn/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glutathione/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Tryptophan/blood , Tryptophan/pharmacology
18.
Rev. chil. pediatr ; 73(6): 590-594, nov.-dic. 2002. graf
Article in Spanish | LILACS | ID: lil-342294

ABSTRACT

La tirosinemia I es una enfermedad metaból ica de herencia recesiva, causada por la diferencia de la enzima terminal de la vía de degradación de la tirosina,llamada fumarilacetoacetato hidrolasa. Compromete principalmente el hígado, sistema nervioso central y riñones. Objetivo: Dada la baja frecuencia de tirosinemia tipo I en nuestro medio, consideramos importante su revisióna raíz de un caso clínico, para optimizar la sospecha diagnóstica frente a la presentaciónclínica y de laboratorio, e iniciar así su tratamiento en forma precoz, mejorando el pronóstico. Caso clínico: reportamos un lactante de 1 mes 11 días, que ingresó al Hospital Padre Hurtado, con el diagnóstico de síndrome febril sin foco, acompañado de vómitos y distensión abdominal. Al ingreso destacó hematuria macroscópica y masa palpable en fosa renal izquierda. Se realizó ecografía abdominal destacando nefrocalcinosis y nefromegalia bilateral y exámenes de laboratorio que muestran hipercalciuria, hipercalcemia, hipofosfemia, hipoalbuminemia, trnasaminasas, LDH y fosfatasas alcalinas elevadas, bilirrubian con leve aumento de predominio directo, reabsorcióntubular de fosfato disminuida, PTH normal, radiografías con signos de raquitismo, cultivos negativos. El paciente evolucionó con distensión abdominal, evidenciandose ascitis moderada en una nueva ecografía abdominal. En el perfil hepático completo destacó protombina 10 por ciento, TTPK de 112 segundos. Ante la fuerte sospecha de Tirosinemia se solicitan alfa feto proteínas que muestran valor muy elevado y aminoacidemia anormal compatible con el diagnóstico. Conclusiones: La revisión de la literatura en relación a esta patología plantea su amplia gama de presentación clínica y las nuevas opciones de tratamiento que han mejorado el pronóstico de estos pacientes, cuales disponemos en nuestro país y fueron aplicadas en este paciente


Subject(s)
Humans , Male , Infant , Amino Acid Metabolism, Inborn Errors/diagnosis , Tyrosine , Tyrosinemias , Breast-Milk Substitutes , Clinical Evolution , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/drug therapy , Hematuria , Nephrocalcinosis , Prognosis , Tyrosine , Tyrosinemias
19.
Pediatr Res ; 50(1): 61-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11420420

ABSTRACT

The treatment of multiple acyl-CoA-dehydrogenase deficiency (MADD) includes a low-fat, low-protein, high-carbohydrate diet, avoiding long fasting periods. However, there is no useful biochemical marker to determine the response to different diets or fasting periods. The aims of this study are to report a patient with MADD, diagnosed through a newborn screening program using tandem mass spectrometry, to assess her response to different feedings, and to evaluate the usefulness of acylcarnitines and FFA to monitor the response to dietary changes. The patient was diagnosed at 6 d. Family history revealed three dead siblings. Five tests were performed, one with breast milk and the subsequent four after giving the patient a bottle of a low-fat, low-protein formula (F), F with glucose polymers (GP), F+GP plus uncooked corn starch (CS), or F+GP+CS preceded by amylase. The results showed that acylcarnitines, FFA, and total nonesterified fatty acids levels were greatly improved at 2 and 4 h on F+GP compared with breast milk. At 6 mo of age, the test with F+CS was repeated to assess the response to a longer fast. The results were similar at 2 and 4 h, but showed a marked increase of acylcarnitines, FFA, and total nonesterified fatty acids at 6 h. The increase of these metabolites could not be avoided by the use of F+GP+CS, but was prevented when amylase was used simultaneously. The patient is currently 3.9 y old and has normal growth and development. We conclude that diagnosis of MADD through a newborn screening program using tandem mass spectrometry is suitable; acylcarnitines and FFA are useful to monitor the response to treatment; and exogenous amylase allows the use of CS in small children with MADD. This therapeutic approach may be an alternative to the use of continuous overnight feedings used for young children with severe fatty acid oxidation defects. Early diagnosis and treatment may change the natural history of MADD.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Amino Acid Metabolism, Inborn Errors/physiopathology , Carnitine/analogs & derivatives , Carnitine/blood , Diet , Fatty Acids/blood , Lipid Metabolism, Inborn Errors/physiopathology , Acyl-CoA Dehydrogenase , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/diet therapy , Female , Humans , Infant, Newborn , Lipid Metabolism, Inborn Errors/blood , Lipid Metabolism, Inborn Errors/diet therapy , Neonatal Screening
20.
J Pediatr ; 138(1 Suppl): S62-71, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11148551

ABSTRACT

Long-term correction of urea cycle disorders is achieved by correction of the enzymatic defect in hepatocytes. Currently, orthotopic liver transplantation is the primary means of achieving this correction. In the United States most liver transplantations for urea cycle disorders have been restricted to patients with ornithine transcarbamylase deficiency and argininosuccinic aciduria. However, patients with citrullinemia have also received transplants, but more so in Europe and Japan. Recent advances in organ procurement, surgical technique, and immunosuppression have significantly decreased morbidity and mortality. However, unique short-term complications associated with surgery and long-term complications associated with chronic immunosuppression have spurred continued efforts to develop gene replacement therapies for management of acute metabolic decompensations as intercurrent therapy until liver transplantation, and ultimately, for long-term correction. The pathophysiology of urea cycle disorders requires gene vector delivery systems that are highly efficient for liver transduction and transgene expression. To date, adenoviral vectors are unique in fulfilling these criteria, and significant data have been gained in both animal and human studies with early versions of adenoviral vectors. Ultimately, the development of helper-dependent adenoviral vectors may offer the long-term expression and increased margin of safety necessary for adjunctive therapies.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Liver Transplantation , Urea/metabolism , Adenoviridae/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/surgery , Animals , Child , Contraindications , Genetic Vectors , Humans
SELECTION OF CITATIONS
SEARCH DETAIL