Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Genet Metab ; 142(3): 108495, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38772223

ABSTRACT

PURPOSE: To identify therapies for combined D, L-2-hydroxyglutaric aciduria (C-2HGA), a rare genetic disorder caused by recessive variants in the SLC25A1 gene. METHODS: Patients C-2HGA were identified and diagnosed by whole exome sequencing and biochemical genetic testing. Patient derived fibroblasts were then treated with phenylbutyrate and the functional effects assessed by metabolomics and RNA-sequencing. RESULTS: In this study, we demonstrated that C-2HGA patient derived fibroblasts exhibited impaired cellular bioenergetics. Moreover, Fibroblasts form one patient exhibited worsened cellular bioenergetics when supplemented with citrate. We hypothesized that treating patient cells with phenylbutyrate (PB), an FDA approved pharmaceutical drug that conjugates glutamine for renal excretion, would reduce mitochondrial 2-ketoglutarate, thereby leading to improved cellular bioenergetics. Metabolomic and RNA-seq analyses of PB-treated fibroblasts demonstrated a significant decrease in intracellular 2-ketoglutarate, 2-hydroxyglutarate, and in levels of mRNA coding for citrate synthase and isocitrate dehydrogenase. Consistent with the known action of PB, an increased level of phenylacetylglutamine in patient cells was consistent with the drug acting as 2-ketoglutarate sink. CONCLUSION: Our pre-clinical studies suggest that citrate supplementation has the possibility exacerbating energy metabolism in this condition. However, improvement in cellular bioenergetics suggests phenylbutyrate might have interventional utility for this rare disease.


Subject(s)
Fibroblasts , Glutarates , Phenylbutyrates , Humans , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Fibroblasts/metabolism , Fibroblasts/drug effects , Glutarates/metabolism , Ketoglutaric Acids/metabolism , Energy Metabolism/drug effects , Energy Metabolism/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/genetics , Metabolomics , Exome Sequencing , Citrate (si)-Synthase/metabolism , Citrate (si)-Synthase/genetics , Brain Diseases, Metabolic, Inborn/drug therapy , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Multiomics , Mitochondrial Proteins , Organic Anion Transporters
2.
Int J Neuropsychopharmacol ; 24(9): 734-748, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34165516

ABSTRACT

BACKGROUND: Minocycline (MIN) is a tetracycline with antioxidant, anti-inflammatory, and neuroprotective properties. Given the likely involvement of inflammation and oxidative stress (IOS) in schizophrenia, MIN has been proposed as a potential adjuvant treatment in this pathology. We tested an early therapeutic window, during adolescence, as prevention of the schizophrenia-related deficits in the maternal immune stimulation (MIS) animal model. METHODS: On gestational day 15, Poly I:C or vehicle was injected in pregnant Wistar rats. A total 93 male offspring received MIN (30 mg/kg) or saline from postnatal day (PND) 35-49. At PND70, rats were submitted to the prepulse inhibition test. FDG-PET and T2-weighted MRI brain studies were performed at adulthood. IOS markers were evaluated in frozen brain tissue. RESULTS: MIN treatment did not prevent prepulse inhibition test behavioral deficits in MIS offspring. However, MIN prevented morphometric abnormalities in the third ventricle but not in the hippocampus. Additionally, MIN reduced brain metabolism in cerebellum and increased it in nucleus accumbens. Finally, MIN reduced the expression of iNOS (prefrontal cortex, caudate-putamen) and increased the levels of KEAP1 (prefrontal cortex), HO1 and NQO1 (amygdala, hippocampus), and HO1 (caudate-putamen). CONCLUSIONS: MIN treatment during adolescence partially counteracts volumetric abnormalities and IOS deficits in the MIS model, likely via iNOS and Nrf2-ARE pathways, also increasing the expression of cytoprotective enzymes. However, MIN treatment during this peripubertal stage does not prevent sensorimotor gating deficits. Therefore, even though it does not prevent all the MIS-derived abnormalities evaluated, our results suggest the potential utility of early treatment with MIN in other schizophrenia domains.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Brain Diseases, Metabolic/drug therapy , Minocycline/pharmacology , Nervous System Malformations/pathology , Neurodevelopmental Disorders/drug therapy , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects/drug therapy , Prepulse Inhibition/drug effects , Schizophrenia/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Behavior, Animal/drug effects , Brain Diseases, Metabolic/etiology , Disease Models, Animal , Female , Magnetic Resonance Imaging , Male , Minocycline/administration & dosage , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/etiology , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/immunology , Positron-Emission Tomography , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/immunology , Rats , Rats, Wistar , Schizophrenia/chemically induced , Schizophrenia/immunology
3.
Arch Biochem Biophys ; 709: 108970, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34181873

ABSTRACT

Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1ß, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1ß and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1ß levels in striatum of Gcdh-/- mice. Finally, IL-1ß was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/drug therapy , Carnitine/therapeutic use , Glutaryl-CoA Dehydrogenase/deficiency , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Amino Acid Metabolism, Inborn Errors/genetics , Animals , Brain Diseases, Metabolic/genetics , Carnitine/analogs & derivatives , Carnitine/metabolism , Cathepsin D/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Grooming/drug effects , Inflammation/genetics , Interleukin-1beta/metabolism , Locomotion/drug effects , Lysine/pharmacology , Mice, Knockout , Open Field Test/drug effects , Transforming Growth Factor beta/metabolism
4.
J Inherit Metab Dis ; 40(1): 75-101, 2017 01.
Article in English | MEDLINE | ID: mdl-27853989

ABSTRACT

Glutaric aciduria type I (GA-I; synonym, glutaric acidemia type I) is a rare inherited metabolic disease caused by deficiency of glutaryl-CoA dehydrogenase located in the catabolic pathways of L-lysine, L-hydroxylysine, and L-tryptophan. The enzymatic defect results in elevated concentrations of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutaryl carnitine in body tissues, which can be reliably detected by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Most untreated individuals with GA-I experience acute encephalopathic crises during the first 6 years of life that are triggered by infectious diseases, febrile reaction to vaccinations, and surgery. These crises result in striatal injury and consequent dystonic movement disorder; thus, significant mortality and morbidity results. In some patients, neurologic disease may also develop without clinically apparent crises at any age. Neonatal screening for GA-I us being used in a growing number of countries worldwide and is cost effective. Metabolic treatment, consisting of low lysine diet, carnitine supplementation, and intensified emergency treatment during catabolism, is effective treatment and improves neurologic outcome in those individuals diagnosed early; treatment after symptom onset, however, is less effective. Dietary treatment is relaxed after age 6 years and should be supervised by specialized metabolic centers. The major aim of this second revision of proposed recommendations is to re-evaluate the previous recommendations (Kölker et al. J Inherit Metab Dis 30:5-22, 2007b; J Inherit Metab Dis 34:677-694, 2011) and add new research findings, relevant clinical aspects, and the perspective of affected individuals.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Glutaryl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Dietary Supplements , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Lysine/metabolism
5.
Am J Hum Genet ; 89(6): 806-12, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152682

ABSTRACT

Thiamine pyrophosphate (TPP) is an essential cofactor of the cytosolic transketolase and of three mitochondrial enzymes involved in the oxidative decarboxylation of either pyruvate, α-ketoglutarate or branched chain amino acids. Thiamine is taken up by specific transporters into the cell and converted to the active TPP by thiamine pyrophosphokinase (TPK) in the cytosol from where it can be transported into mitochondria. Here, we report five individuals from three families presenting with variable degrees of ataxia, psychomotor retardation, progressive dystonia, and lactic acidosis. Investigation of the mitochondrial energy metabolism showed reduced oxidation of pyruvate but normal pyruvate dehydrogenase complex activity in the presence of excess TPP. A reduced concentration of TPP was found in the muscle and blood. Mutation analysis of TPK1 uncovered three missense, one splice-site, and one frameshift mutation resulting in decreased TPK protein levels.


Subject(s)
Abnormalities, Multiple/enzymology , Brain Diseases, Metabolic/enzymology , Metabolic Networks and Pathways/genetics , Pyruvic Acid/metabolism , Thiamin Pyrophosphokinase/deficiency , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/genetics , Acidosis, Lactic/enzymology , Acidosis, Lactic/genetics , Adolescent , Amino Acid Sequence , Base Sequence , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/genetics , Child , DNA Mutational Analysis , Enzyme Assays , Fatal Outcome , Female , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Mutation , Oxidation-Reduction , Pedigree , Thiamin Pyrophosphokinase/genetics , Thiamine/blood , Thiamine/metabolism , Thiamine/therapeutic use
6.
Neuropediatrics ; 45(1): 64-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24297574

ABSTRACT

Pyridoxal-5'-phosphate oxidase (PNPO) deficiency is a rare autosomal recessive, vitamin-responsive metabolic disorder causing refractory neonatal seizures that respond to the administration of pyridoxal-5'-phosphate (PLP). There are currently few case studies that have documented the functional outcome in PNPO deficiency, which remains poor in the majority of cases. We present the case of a male infant born at 35 weeks gestation who promptly responded to oral administration of PLP, following resistance to common anticonvulsive therapy and to a pyridoxine trial. Neurological outcome at 21 months is favorable and illustrates the importance of standardized vitamin trials in the acute setting of "therapy-resistant" neonatal seizures. Early recognition of PNPO deficiency and appropriate intervention might be associated with a more favorable outcome than initially considered.


Subject(s)
Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Hypoxia-Ischemia, Brain/diagnosis , Hypoxia-Ischemia, Brain/drug therapy , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/drug therapy , Phosphoric Monoester Hydrolases/therapeutic use , Pyridoxaminephosphate Oxidase/deficiency , Seizures/diagnosis , Seizures/drug therapy , Humans , Infant, Premature , Male , Treatment Outcome
7.
Dev Med Child Neurol ; 56(5): 498-502, 2014 May.
Article in English | MEDLINE | ID: mdl-24266778

ABSTRACT

Pyridox(am)ine phosphate oxidase (PNPO) deficiency causes severe early infantile epileptic encephalopathy and has been characterized as responding to pyridoxal-5'-phosphate but not to pyridoxine. Two males with PNPO deficiency and novel PNPO mutations are reported and their clinical, metabolic, and video-electroencephalographic (EEG) findings described. The first child showed electro-clinical responses to pyridoxine and deterioration when pyridoxine was withheld. At last review, he has well-controlled epilepsy with pyridoxal-5'-phosphate monotherapy and an autism spectrum disorder. The second child had a perinatal middle cerebral artery infarct and a myoclonic encephalopathy. He failed to respond to pyridoxine but responded well to pyridoxal-5'-phosphate. At the age of 21 months he has global developmental delay and hemiparesis but is seizure-free with pyridoxal-5'-phosphate monotherapy. Plasma and cerebrospinal fluid pyridoxamine levels were increased in both children during treatment with pyridoxine or pyridoxal-5'-phosphate. These observations indicate that differential responses to pyridoxine and pyridoxal-5'-phosphate treatment cannot be relied upon to diagnose PNPO deficiency.


Subject(s)
Brain Diseases, Metabolic , Hypoxia-Ischemia, Brain , Pyridoxal Phosphate/therapeutic use , Pyridoxamine/blood , Pyridoxamine/cerebrospinal fluid , Pyridoxaminephosphate Oxidase/deficiency , Seizures , Vitamin B Complex/therapeutic use , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/physiopathology , Child , Child, Preschool , Electroencephalography , Humans , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Male , Pyridoxaminephosphate Oxidase/metabolism , Seizures/drug therapy , Seizures/metabolism , Seizures/physiopathology
8.
Klin Med (Mosk) ; 92(5): 46-50, 2014.
Article in Russian | MEDLINE | ID: mdl-25782306

ABSTRACT

Effects of pirebedil used to prevent falls in elderly patients with metabolic syndrome are discussed. A prospective controlled study showed that therapy with pirebedil significantly decreases the frequency of falls, reduces severity of pro-inflammatory and pro-oxidative activities, improves cognitive abilities. Prevention of falls by virtue of improved cognitive abilities is a new clinical effect of pirebedil and gives reason to recommend it for the treatment of geriatric patients with metabolic syndrome.


Subject(s)
Accidental Falls/prevention & control , Brain Diseases, Metabolic , Diabetes Mellitus, Type 2 , Mental Competency , Piribedil , Aged , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/etiology , Brain Diseases, Metabolic/physiopathology , Brain Diseases, Metabolic/psychology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Dopamine Agonists/administration & dosage , Dopamine Agonists/pharmacokinetics , Female , Geriatric Assessment/methods , Humans , Male , Middle Aged , Nootropic Agents/administration & dosage , Nootropic Agents/pharmacokinetics , Piribedil/administration & dosage , Piribedil/pharmacokinetics , Prospective Studies , Treatment Outcome
9.
ACS Chem Biol ; 19(7): 1544-1553, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38915184

ABSTRACT

Glutaric Aciduria Type 1 (GA1) is a serious inborn error of metabolism with no pharmacological treatments. A novel strategy to treat this disease is to divert the toxic biochemical intermediates to less toxic or nontoxic metabolites. Here, we report a putative novel target, succinyl-CoA:glutarate-CoA transferase (SUGCT), which we hypothesize suppresses the GA1 metabolic phenotype through decreasing glutaryl-CoA and the derived 3-hydroxyglutaric acid. SUGCT is a type III CoA transferase that uses succinyl-CoA and glutaric acid as substrates. We report the structure of SUGCT, develop enzyme- and cell-based assays, and identify valsartan and losartan carboxylic acid as inhibitors of the enzyme in a high-throughput screen of FDA-approved compounds. The cocrystal structure of SUGCT with losartan carboxylic acid revealed a novel pocket in the active site and further validated the high-throughput screening approach. These results may form the basis for the future development of new pharmacological intervention to treat GA1.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Humans , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/enzymology , Glutarates/metabolism , Glutarates/chemistry , Losartan/pharmacology , Losartan/chemistry , Coenzyme A-Transferases/metabolism , Coenzyme A-Transferases/antagonists & inhibitors , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/chemistry , Valsartan , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Crystallography, X-Ray , Catalytic Domain , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/chemistry , Models, Molecular , High-Throughput Screening Assays , Glutaryl-CoA Dehydrogenase/deficiency
10.
Gan To Kagaku Ryoho ; 40(13): 2581-4, 2013 Dec.
Article in Japanese | MEDLINE | ID: mdl-24335375

ABSTRACT

We report a rare case of a patient with metastatic colorectal cancer who experienced hyperammonemic encephalopathy induced by 5 -fluorouracil(5-FU)and was continuously treated with capecitabine plus oxaliplatin(XELOX)therapy. A 60 years man with anorexia and weight loss was diagnosed with Stage IV rectal cancer, and chemotherapy with XELOX was initiated. When the second course of XELOX therapy was administered, the patient found it difficult to take oral capecitabine. Subsequently, modified FOLFOX6 was administered. Complications such as nausea and vomiting were observed on day 2, with confusion and cognitive disturbances on day 3 . Laboratory examination revealed hyperammonemia, and therefore, branched-chain amino acid solutions were administered as treatment. The patient's symptoms disappeared entirely on day 4. He is currently receiving XELOX therapy.


Subject(s)
Amino Acids, Branched-Chain/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Diseases, Metabolic/etiology , Fluorouracil/adverse effects , Hyperammonemia/drug therapy , Liver Neoplasms/drug therapy , Rectal Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Diseases, Metabolic/drug therapy , Capecitabine , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Humans , Hyperammonemia/chemically induced , Liver Neoplasms/secondary , Male , Oxaloacetates , Rectal Neoplasms/pathology
11.
Eksp Klin Farmakol ; 76(11): 40-3, 2013.
Article in Russian | MEDLINE | ID: mdl-24555233

ABSTRACT

The effect of cytoflavin upon bolus intravenous administration on the state of hemodynamic values and liquid sectors of the organism was studied in patients with toxicohypoxic encephalopathy caused by heavy acute poisoning. It is established that the bolus administration of cytoflavin is less effective than dropwise intravenous injection.


Subject(s)
Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/pathology , Brain Diseases, Metabolic/physiopathology , Flavin Mononucleotide/administration & dosage , Hemodynamics/drug effects , Inosine Diphosphate/administration & dosage , Niacinamide/administration & dosage , Poisoning/drug therapy , Poisoning/physiopathology , Succinates/administration & dosage , Acute Disease , Adult , Brain Diseases, Metabolic/etiology , Drug Combinations , Female , Humans , Male , Middle Aged , Poisoning/complications
12.
J Neurosci Res ; 90(9): 1723-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22535575

ABSTRACT

Glutaric acidemia type I (GA-I) is an inherited metabolic disease characterized by accumulation of glutaric acid (GA) and seizures. The intrastriatal GA administration in rats has been used as an animal model to mimic seizures presented by glutaric acidemic patients. m-Trifluoromethyl diphenyl diselenide, (m-CF(3) -C(6) H(4) Se)(2) , is an organoselenium compound that protects against seizures induced by pentylenetetrazole in mice. Thus, the aim of this study was to investigate whether (m-CF(3) -C(6) H(4) Se)(2) is effective against GA-induced seizures and oxidative stress in rat pups 21 days of age. Our findings demonstrate that (m-CF(3) -C(6) H(4) Se)(2) preadministration (50 mg/kg; p.o.) protected against the reduction in latency and the increased duration of GA (1.3 µmol/right striatum)-induced seizures in rat pups. In addition, (m-CF(3) -C(6) H(4) Se)(2) protected against the increase in reactive species generation and the reduction in antioxidant defenses glutathione peroxidase and glutathione S-transferase activities induced by GA. By contrast, no change in glutathione reductase or catalase activities was found. In addition, (m-CF(3) -C(6) H(4) Se)(2) was effective in protecting against inhibition of Na(+) ,K(+) -ATPase activity caused by GA in striatum of rat pups. This study showed for the first time that GA administration caused an increase in [(3) H]GABA uptake from striatum slices of rat pups and that (m-CF(3) -C(6) H(4) Se)(2) preadministration protected against this increase. A positive correlation between duration of seizures and [(3) H]GABA uptake levels was demonstrated. The results indicate that (m-CF(3) -C(6) H(4) Se)(2) protected against GA-induced seizures. Moreover, these findings suggest that the protection against oxidative stress, the inhibition of Na(+) ,K(+) -ATPase activity, and the increase in [(3) H]GABA uptake are possible mechanisms for the potential anticonvulsant action of (m-CF(3) -C(6) H(4) Se)(2).


Subject(s)
Anticonvulsants/pharmacology , Glutarates/toxicity , Organosilicon Compounds/pharmacology , Oxidative Stress/drug effects , Seizures/drug therapy , Seizures/metabolism , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Disease Models, Animal , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/metabolism , Male , Rats , Rats, Wistar , Seizures/chemically induced , gamma-Aminobutyric Acid
13.
Mol Genet Metab ; 107(1-2): 81-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22578804

ABSTRACT

Glutaric acidemia type I (GA I) is an inherited neurometabolic disorder caused by a severe deficiency of the mitochondrial glutaryl-CoA dehydrogenase activity leading to accumulation of predominantly glutaric (GA) and 3-hydroxyglutaric (3HGA) acids in the brain and other tissues. Affected patients usually present with hypotonia and brain damage and acute encephalopathic episodes whose pathophysiology is not yet fully established. In this study we investigated important parameters of cellular bioenergetics in brain, heart and skeletal muscle from 15-day-old glutaryl-CoA dehydrogenase deficient mice (Gcdh(-/-)) submitted to a single intra-peritoneal injection of saline (Sal) or lysine (Lys - 8 µmol/g) as compared to wild type (WT) mice. We evaluated the activities of the respiratory chain complexes II, II-III and IV, α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and synaptic Na(+), K(+)-ATPase. No differences of all evaluated parameters were detected in the Gcdh(-/-) relatively to the WT mice injected at baseline (Sal). Furthermore, mild increases of the activities of some respiratory chain complexes (II-III and IV) were observed in heart and skeletal muscle of Gcdh(-/-) and WT mice after Lys administration. However, the most marked effects provoked by Lys administration were marked decreases of the activities of Na(+), K(+)-ATPase in brain and CK in brain and skeletal muscle of Gcdh(-/-) mice. In contrast, brain α-KGDH activity was not altered in WT and Gcdh(-/-) injected with Sal or Lys. Our results demonstrate that reduction of Na(+), K(+)-ATPase and CK activities may play an important role in the pathogenesis of the neurodegenerative changes in GA I.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Creatine Kinase/metabolism , Lysine/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Metabolism, Inborn Errors/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Brain Diseases, Metabolic/drug therapy , Disease Models, Animal , Electron Transport/drug effects , Electron Transport/physiology , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/metabolism , Heart/drug effects , Ketoglutarate Dehydrogenase Complex/metabolism , Lysine/administration & dosage , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocardium/metabolism
14.
Bioorg Med Chem Lett ; 22(19): 6173-80, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22959249

ABSTRACT

Analogs of SLV-319 (Ibipinibant), a CB1 receptor inverse agonist, were synthesized with functionality intended to limit brain exposure while maintaining the receptor affinity and selectivity of the parent compound. Structure activity relationships of this series, and pharmacology of two lead compounds, 16 (JD-5006) and 23 (JD-5037) showing little brain presence as indicated by tissue distribution and receptor occupancy studies, are described. Effects with one of these compounds on plasma triglyceride levels, liver weight and enzymes, glucose tolerance and insulin sensitivity support the approach that blockade of peripheral CB(1) receptors is sufficient to produce many of the beneficial metabolic effects of globally active CB(1) blockers. Thus, PR CB(1) inverse agonists may indeed represent a safer alternative to highly brain-penetrant agents for the treatment of metabolic disorders, including diabetes, liver diseases, dyslipidemias, and obesity.


Subject(s)
Amidines/pharmacology , Brain Diseases, Metabolic/drug therapy , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Amidines/chemical synthesis , Amidines/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship , Sulfonamides
15.
Mol Biol Rep ; 39(10): 9311-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22810648

ABSTRACT

In this study, we determined the neuroprotective effect of aucubin on diabetes and diabetic encephalopathy. With the exception of the control group, all rats received intraperitoneal injections of streptozotocin (STZ; 60 mg/kg) to induce type 1 diabetes mellitus (DM). Aucubin (1, 5, 10 mg/kg ip) was used after induction of DM (immediately) and diabetic encephalopathy (65 days after the induction of diabetes). The diabetic encephalopathy treatment groups were divided into short-term and long-term treatment groups. Treatment responses to all parameters were examined (body weight, plasma glucose, Y-maze error rates and proportion of apoptotic cells). In diabetic rats, aucubin controlled blood glucose levels effectively, prevented complications, and improved the quality of life of diabetic rats. In diabetic encephalopathy, aucubin significantly rescued neurons in the hippocampal CA1 subfield and reduced working errors during behavioral testing. The significant neuroprotective effect of aucubin could be seen not only in the short term (15 days) but also in the long term (45 days), which was a highly encouraging finding. These data suggest that aucubin may be a potential neuroprotective agent.


Subject(s)
Brain Diseases, Metabolic/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Iridoid Glucosides/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Blood Glucose , Body Weight/drug effects , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/prevention & control , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cell Survival/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Iridoid Glucosides/pharmacology , Male , Neuroprotective Agents/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats , Rats, Wistar
17.
J Endocrinol Invest ; 35(3): 312-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21646859

ABSTRACT

Thyroid hormone insufficiency in adulthood causes a wide range of brain impairments, including altered synaptic proteins in the prefrontal cortex (PFC). The present study investigated whether adult-onset hypothyroidism altered the expression of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes and synaptotagmin-1 (syt-1) in the PFC of rats. Sprague-Dawley rats were randomly divided into 4 groups: control, hypothyroid, and hypothyroid treated with T(4) [5 or 20 µg/100 g body weight (BW)]. Adult-onset hypothyroidism was induced in rats with the antithyroid drug 6-n-propyl-2-thiouracil (ip injection). PFC levels of synaptosomal-associated protein of 25 kDa (SNAP-25), syntaxin-1, vesicle-associated membrane protein 2 (VAMP-2) and syt-1 were determined by immunohistochemistry and western blot analyses. The results showed that syntaxin-1 and syt-1 were expressed at significantly lower levels in hypothyroid rats, VAMP-2 levels were not altered, and SNAP-25 levels were much higher compared to controls. A 2-week treatment with 5 µg T(4)/100 g BW partially normalized levels of SNARE complex and syt-1, and 20 µg T(4)/100 g BW restored these proteins closer to normal levels. Our findings indicate that dysregulation of SNARE complex and syt-1 in PFC of adult-onset hypothyroidism can be restored by T(4) treatment.


Subject(s)
Brain Diseases, Metabolic/metabolism , Hypothyroidism/metabolism , Prefrontal Cortex/metabolism , SNARE Proteins/metabolism , Synaptotagmin I/metabolism , Thyroxine/metabolism , Animals , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/etiology , Hypothyroidism/complications , Hypothyroidism/drug therapy , Male , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Thyroxine/blood , Thyroxine/pharmacology , Triiodothyronine/blood , Vesicle-Associated Membrane Protein 2/metabolism
18.
Mol Genet Metab ; 104(1-2): 93-106, 2011.
Article in English | MEDLINE | ID: mdl-21820344

ABSTRACT

Striatal degeneration from glutaryl-CoA dehydrogenase deficiency (glutaric aciduria type 1, GA1) is associated with cerebral formation and entrapment of glutaryl-CoA and its derivatives that depend on cerebral lysine influx. In 2006 we designed a lysine-free study formula enriched with arginine to selectively block lysine transport across cerebral endothelia and thereby limit glutaryl-CoA production by brain. Between 2006 and present, we treated twelve consecutive children with study formula (LYSx group) while holding all other treatment practices constant. Clinical and biochemical outcomes were compared to 25 GA1 patients (PROx group) treated between 1995 and 2005 with natural protein restriction (dietary lysine/arginine ratio of 1.7±0.3 mg:mg). We used published kinetic parameters of the y+and LAT1 blood-brain barrier transporters to model the influx of amino acids into the brain. Arginine fortification to achieve a mean dietary lysine/arginine ratio of 0.7±0.2 mg:mg was neuroprotective. All 12 LYSx patients are physically and neurologically healthy after 28 aggregate patient-years of follow up (current ages 28±21 months) and there were no adverse events related to formula use. This represents a 36% reduction of neurological risk (95% confidence interval 14-52%, p=0.018) that we can directly attribute to altered amino acid intake. During the first year of life, 20% lower lysine intake and two-fold higher arginine intake by LYSx patients were associated with 50% lower plasma lysine, 3-fold lower plasma lysine/arginine concentration ratio, 42% lower mean calculated cerebral lysine influx, 54% higher calculated cerebral arginine influx, 15-26% higher calculated cerebral influx of several anaplerotic precursors (isoleucine, threonine, methionine, and leucine), 50% less 3-hydroxyglutarate excretion, and a 3-fold lower hospitalization rate (0.8 versus 2.3 hospitalizations per patient per year). The relationship between arginine fortification and plasma lysine indicates that transport competition exists at both cerebrovascular and gastrointestinal barriers, suggesting their co-administration is key to efficacy. Monitoring the ratio between lysine and arginine in diet and plasma may prove a useful strategy for treating children with GA1.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Arginine/adverse effects , Arginine/therapeutic use , Brain Diseases, Metabolic/drug therapy , Brain/metabolism , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/physiopathology , Arginine/blood , Brain/pathology , Brain Diseases, Metabolic/blood , Brain Diseases, Metabolic/physiopathology , Carnitine/administration & dosage , Carnitine/therapeutic use , Child, Preschool , Dietary Proteins/metabolism , Dose-Response Relationship, Drug , Female , Glutaryl-CoA Dehydrogenase/blood , Glutaryl-CoA Dehydrogenase/deficiency , Growth and Development , Homeostasis , Hospitalization , Humans , Immunization , Infant , Infant, Newborn , Lysine/blood , Male , Nutritional Physiological Phenomena , Treatment Outcome
19.
Mol Genet Metab ; 104(3): 265-72, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21816645

ABSTRACT

Influenza-associated encephalopathy (IAE) is characterized by persistently high fever, febrile convulsions, severe brain edema and high mortality. We reported previously that a large proportion of patients with disabling or fatal IAE exhibit a thermolabile phenotype of compound variants for [1055T>G/F352C] and [1102G>A/V368I] of carnitine palmitoyltransferase II (CPT II) and mitochondrial energy crisis during high fever. In the present study, we studied the effect of bezafibrate, a hypolipidemic pan-agonist of peroxisome proliferator-activated receptor (PPAR), on CPT II expression and mitochondrial energy metabolism in fibroblasts of IAE patients and wild type (WT) fibroblasts from a healthy volunteer at 37°C and 41°C. Although heat stress markedly upregulated CPT II, CPT IA and PPAR-δ mRNA expression levels, CPT II activity, ß-oxidation and ATP levels in WT and IAE fibroblasts at 41°C were paradoxically downregulated probably due to the thermal instability of the corresponding enzymes. Bezafibrate significantly enhanced the expression levels of the above mRNAs and cellular functions of these enzymes in fibroblasts at 37°C. Bezafibrate-induced increase in CPT II activity also tended to restore the downregulated ATP levels, though moderately, and improved mitochondrial membrane potential even at 41°C to the levels at 37°C in fibroblasts of IAE patients. L-carnitine, a substrate of CPT II, boosted the effects of bezafibrate on cellular ATP levels in WT and IAE fibroblasts, even in severe IAE fibroblasts with thermolabile compound variations of F352C+V368I at 37°C and 41°C. The results suggest the potential usefulness of bezafibrate for the treatment of IAE.


Subject(s)
Bezafibrate/pharmacology , Brain Diseases, Metabolic/drug therapy , Carnitine O-Palmitoyltransferase/metabolism , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Influenza, Human/complications , Mitochondria/drug effects , RNA, Messenger/metabolism , Adenosine Triphosphate/metabolism , Base Sequence , Bezafibrate/therapeutic use , Blotting, Western , Brain Diseases, Metabolic/etiology , Brain Diseases, Metabolic/metabolism , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/genetics , DNA Primers/genetics , Fibroblasts/metabolism , Genomics/methods , Hot Temperature , Humans , Japan , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Microscopy, Fluorescence , Mitochondria/metabolism , Molecular Sequence Data , Peroxisome Proliferator-Activated Receptors/agonists , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Time Factors
20.
Med Sci Monit ; 17(7): PH55-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21709643

ABSTRACT

BACKGROUND: Glutaric aciduria type I (GA I; MIM 231670) is a rare autosomal recessive disorder resulting from glutaryl-CoA dehydrogenase deficiency. This article reports our experience in the diagnosis, treatment and outcome of GA I patients in Zhejiang Province, China. MATERIAL/METHODS: A total of 129,415 newborns (accounting for approximately one-tenth of the annual births in Zhejiang Province) and 9640 high-risk infants were screened for inborn errors of metabolism in the Neonatal Screening Center of Zhejiang Province during a 3-year period. Tandem mass spectrometry and gas chromatography-mass spectrometry were used for diagnosis of the patients. Dietary modification, carnitine supplementation and aggressive treatment of intercurrent illnesses were adapted for GA I patients. RESULTS: Three infants were diagnosed with GA I by high-risk screening (detection rate: 1/3,213) and 2 were diagnosed by newborn screening (incidence: 1/64,708). Four patients (3 by high-risk screening and 1 by neonatal screening) undergoing MRI examination showed remarkable changes on T2-weighted image. Four patients accepted timely treatment, and in the patient diagnosed by neonatal screening, treatment was delayed until hypotonia appeared 3 months later. Neuropsychological assessment showed mental and motor retardation in 3 patients after treatment, including the patient diagnosed by neonatal screening. CONCLUSIONS: Individualized timely treatment and close monitoring of GA I patients needs to be optimized in China. Appropriate communication with parents may help to achieve successful management of GA I patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/epidemiology , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/epidemiology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Carnitine/therapeutic use , China/epidemiology , Female , Gas Chromatography-Mass Spectrometry , Genes, Recessive , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Humans , Incidence , Infant , Infant, Newborn , Male , Mass Screening , Neuropsychological Tests , Riboflavin/therapeutic use , Tandem Mass Spectrometry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL