Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165682, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31931102

RESUMO

Propionic acidemia is caused by lack of propionyl-CoA carboxylase activity. It is biochemically characterized by accumulation of propionic (PA) and 3-hydroxypropionic (3OHPA) acids and clinically by severe encephalopathy and cardiomyopathy. High urinary excretion of maleic acid (MA) and 2-methylcitric acid (2MCA) is also found in the affected patients. Considering that the underlying mechanisms of cardiac disease in propionic acidemia are practically unknown, we investigated the effects of PA, 3OHPA, MA and 2MCA (0.05-5 mM) on important mitochondrial functions in isolated rat heart mitochondria, as well as in crude heart homogenates and cultured cardiomyocytes. MA markedly inhibited state 3 (ADP-stimulated), state 4 (non-phosphorylating) and uncoupled (CCCP-stimulated) respiration in mitochondria supported by pyruvate plus malate or α-ketoglutarate associated with reduced ATP production, whereas PA and 3OHPA provoked less intense inhibitory effects and 2MCA no alterations at all. MA-induced impaired respiration was attenuated by coenzyme A supplementation. In addition, MA significantly inhibited α-ketoglutarate dehydrogenase activity. Similar data were obtained in heart crude homogenates and permeabilized cardiomyocytes. MA, and PA to a lesser degree, also decreased mitochondrial membrane potential (ΔΨm), NAD(P)H content and Ca2+ retention capacity, and caused swelling in Ca2+-loaded mitochondria. Noteworthy, ΔΨm collapse and mitochondrial swelling were fully prevented or attenuated by cyclosporin A and ADP, indicating the involvement of mitochondrial permeability transition. It is therefore proposed that disturbance of mitochondrial energy and calcium homeostasis caused by MA, as well as by PA and 3OHPA to a lesser extent, may be involved in the cardiomyopathy commonly affecting propionic acidemic patients.


Assuntos
Maleatos/metabolismo , Mitocôndrias Cardíacas/patologia , Mioblastos Cardíacos/patologia , Propionatos/metabolismo , Animais , Cálcio/metabolismo , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Fracionamento Celular , Linhagem Celular , Metabolismo Energético , Humanos , Masculino , Mitocôndrias Cardíacas/metabolismo , Dilatação Mitocondrial , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Oxigênio/análise , Oxigênio/metabolismo , Acidemia Propiônica/complicações , Acidemia Propiônica/metabolismo , Acidemia Propiônica/patologia , Ratos
2.
Mol Genet Metab ; 122(1-2): 51-59, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28712602

RESUMO

BACKGROUND: Propionic acidemia is a rare metabolic disorder caused by a deficiency of propionyl- CoA carboxylase, the enzyme converting propionyl-CoA to methylmalonyl-CoA that subsequently enters the citric acid cycle as succinyl-CoA. Patients with propionic acidemia cannot metabolize propionic acid, which combines with oxaloacetate to form methylcitric acid. This, with the defective supply of succinyl-CoA, may lead to a deficiency in citric acid cycle intermediates. PURPOSE: The objective of this study was to determine whether supplements with glutamine (400mg/kg per day), citrate (7.5mEq/kg per day), or ornithine α-ketoglutarate (400mg/kg per day) (anaplerotic agents that could fill up the citric acid cycle) would affect plasma levels of glutamine and ammonia, the urinary excretion of Krebs cycle intermediates, and the clinical outcome in 3 patients with propionic acidemia. METHODS: Each supplement was administered daily for four weeks with a two week washout period between supplements. The supplement that produced the most favorable changes was supplemented for 30 weeks following the initial study period and then for a 2 year extension. RESULTS: The urinary excretion of the Krebs cycle intermediates, α-ketoglutarate, succinate, and fumarate increased significantly compared to baseline during citrate supplementation, but not with the other two supplements. For this reason, citrate supplements were continued in the second part of the study. The urinary excretion of methylcitric acid and 3-hydroxypropionic acid did not change with any intervention. No significant changes in ammonia or glutamine levels were observed with any supplement. However, supplementation with any anaplerotic agents normalized the physiological buffering of ammonia by glutamate, with plasma glutamate and alanine levels significantly increasing, rather than decreasing with increasing ammonia levels. No significant side effects were observed with any therapy and safety labs (blood counts, chemistry and thyroid profile) remained unchanged. Motor and cognitive development was severely delayed before the trial and did not change significantly with therapy. Hospitalizations per year did not change during the trial period, but decreased significantly (p<0.05) in the 2years following the study (when citrate was continued) compared to the 2years before and during the study. CONCLUSIONS: These results indicate that citrate entered the Krebs cycle providing successful anaplerotic therapy by increasing levels of the downstream intermediates of the Krebs cycle: α-ketoglutarate, succinate and fumarate. Citrate supplements were safe and might have contributed to reduce hospitalizations in patients with propionic acidemia.


Assuntos
Ciclo do Ácido Cítrico/efeitos dos fármacos , Ácido Cítrico/administração & dosagem , Suplementos Nutricionais , Glutamina/administração & dosagem , Ornitina/análogos & derivados , Acidemia Propiônica/dietoterapia , Aminoácidos/sangue , Amônia/sangue , Carbono-Carbono Ligases/metabolismo , Criança , Pré-Escolar , Citratos/urina , Ácido Cítrico/efeitos adversos , Suplementos Nutricionais/efeitos adversos , Feminino , Glutamina/efeitos adversos , Glutamina/sangue , Humanos , Ácido Láctico/análogos & derivados , Ácido Láctico/urina , Masculino , Ornitina/administração & dosagem , Acidemia Propiônica/metabolismo , Acidemia Propiônica/fisiopatologia , Resultado do Tratamento
3.
Biochem Biophys Res Commun ; 452(3): 457-61, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25159844

RESUMO

Propionic acidemia (PA), caused by a deficiency of the mitochondrial biotin dependent enzyme propionyl-CoA carboxylase (PCC) is one of the most frequent organic acidurias in humans. Most PA patients present in the neonatal period with metabolic acidosis and hyperammonemia, developing different neurological symptoms, movement disorders and cardiac complications. There is strong evidence indicating that oxidative damage could be a pathogenic factor in neurodegenerative, mitochondrial and metabolic diseases. Recently, we identified an increase in ROS levels in PA patients-derived fibroblasts. Here, we analyze the capability of seven antioxidants to scavenge ROS production in PA patients' cells. Tiron, trolox, resveratrol and MitoQ significantly reduced ROS content in patients and controls' fibroblasts. In addition, changes in the expression of two antioxidant enzymes, superoxide dismutase and glutathione peroxidase, were observed in PA patients-derived fibroblasts after tiron and resveratrol treatment. Our results in PA cellular models establish the proof of concept of the potential of antioxidants as an adjuvant therapy for PA and pave the way for future assessment of antioxidant strategies in the murine model of PA.


Assuntos
Antioxidantes/farmacologia , Fibroblastos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Sal Dissódico do Ácido 1,2-Di-Hidroxibenzeno-3,5 Dissulfônico/farmacologia , Cromanos/farmacologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Metilmalonil-CoA Descarboxilase/genética , Mitocôndrias/metabolismo , Mutação , Compostos Organofosforados/farmacologia , Cultura Primária de Células , Acidemia Propiônica/genética , Acidemia Propiônica/metabolismo , Acidemia Propiônica/patologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Resveratrol , Estilbenos/farmacologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia , Glutationa Peroxidase GPX1
4.
J Inherit Metab Dis ; 36(5): 731-40, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23053474

RESUMO

Propionic acidemia (PA), caused by a deficiency of the mitochondrial biotin dependent enzyme propionyl-CoA carboxylase (PCC) is one of the most frequent organic acidurias in humans. PA is caused by mutations in either the PCCA or PCCB genes encoding the α- and ß-subunits of the PCC enzyme which are assembled as an α6ß6 dodecamer. In this study we have investigated the molecular basis of the defect in ten fibroblast samples from PA patients. Using homology modeling with the recently solved crystal structure of the PCC holoenzyme and a eukaryotic expression system we have analyzed the structural and functional effect of novel point mutations, also revealing a novel splice defect by minigene analysis. In addition, we have investigated the contribution of oxidative stress to cellular damage measuring reactive oxygen species (ROS) levels and apoptosis parameters in patient fibroblasts, as recent studies point to a secondary mitochondrial dysfunction as pathophysiological mechanism in this disorder. The results show an increase in intracellular ROS content compared to controls, correlating with the activation of the JNK and p38 signaling pathways. Highest ROS levels were present in cells harboring functionally null mutations, including one severe missense mutation. This work provides molecular insight into the pathogenicity of PA variants and indicates that oxidative stress may be a major contributing factor to the cellular damage, supporting the proposal of antioxidant strategies as novel supplementary therapy in this rare disease.


Assuntos
Estresse Oxidativo/genética , Mutação Puntual , Acidemia Propiônica/genética , Acidemia Propiônica/metabolismo , Apoptose/genética , Fibroblastos/metabolismo , Estudos de Associação Genética , Genótipo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mutação de Sentido Incorreto , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA