Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Orphanet J Rare Dis ; 18(1): 80, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37046296

RESUMEN

BACKGROUND: Neurodegeneration with brain iron accumulation (NBIA) disorders are a group of neurodegenerative diseases that have in common the accumulation of iron in the basal nuclei of the brain which are essential components of the extrapyramidal system. Frequent symptoms are progressive spasticity, dystonia, muscle rigidity, neuropsychiatric symptoms, and retinal degeneration or optic nerve atrophy. One of the most prevalent subtypes of NBIA is Pantothenate kinase-associated neurodegeneration (PKAN). It is caused by pathogenic variants in the gene of pantothenate kinase 2 (PANK2) which encodes the enzyme responsible for the first reaction on the coenzyme A (CoA) biosynthesis pathway. Thus, deficient PANK2 activity induces CoA deficiency as well as low expression levels of 4'-phosphopantetheinyl proteins which are essential for mitochondrial metabolism. METHODS: This study is aimed at evaluating the role of alpha-lipoic acid (α-LA) in reversing the pathological alterations in fibroblasts and induced neurons derived from PKAN patients. Iron accumulation, lipid peroxidation, transcript and protein expression levels of PANK2, mitochondrial ACP (mtACP), 4''-phosphopantetheinyl and lipoylated proteins, as well as pyruvate dehydrogenase (PDH) and Complex I activity were examined. RESULTS: Treatment with α-LA was able to correct all pathological alterations in responsive mutant fibroblasts with residual PANK2 enzyme expression. However, α-LA had no effect on mutant fibroblasts with truncated/incomplete protein expression. The positive effect of α-LA in particular pathogenic variants was also confirmed in induced neurons derived from mutant fibroblasts. CONCLUSIONS: Our results suggest that α-LA treatment can increase the expression levels of PANK2 and reverse the mutant phenotype in PANK2 responsive pathogenic variants. The existence of residual enzyme expression in some affected individuals raises the possibility of treatment using high dose of α-LA.


Asunto(s)
Enfermedades Neurodegenerativas , Neurodegeneración Asociada a Pantotenato Quinasa , Ácido Tióctico , Humanos , Suplementos Dietéticos , Hierro/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/genética , Neurodegeneración Asociada a Pantotenato Quinasa/tratamiento farmacológico , Neurodegeneración Asociada a Pantotenato Quinasa/genética , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ácido Tióctico/uso terapéutico , Ácido Tióctico/metabolismo
2.
Hum Mol Genet ; 21(18): 4049-59, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22692681

RESUMEN

Pantothenate kinase-associated neurodegeneration (PKAN) is a neurodegenerative disease belonging to the group of neurodegeneration with brain iron accumulation disorders. It is characterized by progressive impairments in movement, speech and cognition. The disease is inherited in a recessive manner due to mutations in the Pantothenate Kinase-2 (PANK2) gene that encodes a mitochondrial protein involved in Coenzyme A synthesis. To investigate the link between a PANK2 gene defect and iron accumulation, we analyzed primary skin fibroblasts from three PKAN patients and three unaffected subjects. The oxidative status of the cells and their ability to respond to iron were analyzed in both basal and iron supplementation conditions. In basal conditions, PKAN fibroblasts show an increase in carbonylated proteins and altered expression of antioxidant enzymes with respect to the controls. After iron supplementation, the PKAN fibroblasts had a defective response to the additional iron. Under these conditions, ferritins were up-regulated and Transferrin Receptor 1 (TfR1) was down-regulated to a minor extent in patients compared with the controls. Analysis of iron regulatory proteins (IRPs) reveals that, with respect to the controls, PKAN fibroblasts have a reduced amount of membrane-associated mRNA-bound IRP1, which responds imperfectly to iron. This accounts for the defective expression of ferritin and TfR1 in patients' cells. The inaccurate quantity of these proteins produced a higher bioactive labile iron pool and consequently increased iron-dependent reactive oxygen species formation. Our results suggest that Pank2 deficiency promotes an increased oxidative status that is further enhanced by the addition of iron, potentially causing damage in cells.


Asunto(s)
Fibroblastos/metabolismo , Hierro/metabolismo , Neurodegeneración Asociada a Pantotenato Quinasa/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Piel/patología , Estudios de Casos y Controles , Catalasa/metabolismo , Células Cultivadas , Ferritinas/metabolismo , Fibroblastos/enzimología , Humanos , Proteínas Reguladoras del Hierro/metabolismo , Mutación Missense , Oxidación-Reducción , Estrés Oxidativo , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Unión Proteica , Carbonilación Proteica , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
3.
Mol Genet Metab ; 105(3): 463-71, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22221393

RESUMEN

Pantothenate kinase-associated neurodegeneration (PKAN) is a rare, inborn error of metabolism characterized by iron accumulation in the basal ganglia and by the presence of dystonia, dysarthria, and retinal degeneration. Mutations in pantothenate kinase 2 (PANK2), the rate-limiting enzyme in mitochondrial coenzyme A biosynthesis, represent the most common genetic cause of this disorder. How mutations in this core metabolic enzyme give rise to such a broad clinical spectrum of pathology remains a mystery. To systematically explore its pathogenesis, we performed global metabolic profiling on plasma from a cohort of 14 genetically defined patients and 18 controls. Notably, lactate is elevated in PKAN patients, suggesting dysfunctional mitochondrial metabolism. As predicted, but never previously reported, pantothenate levels are higher in patients with premature stop mutations in PANK2. Global metabolic profiling and follow-up studies in patient-derived fibroblasts also reveal defects in bile acid conjugation and lipid metabolism, pathways that require coenzyme A. These findings raise a novel therapeutic hypothesis, namely, that dietary fats and bile acid supplements may hold potential as disease-modifying interventions. Our study illustrates the value of metabolic profiling as a tool for systematically exploring the biochemical basis of inherited metabolic diseases.


Asunto(s)
Coenzima A/deficiencia , Mitocondrias/enzimología , Distrofias Neuroaxonales/metabolismo , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adolescente , Adulto , Ácidos y Sales Biliares/metabolismo , Niño , Preescolar , Codón sin Sentido , Coenzima A/biosíntesis , Coenzima A/genética , Estudios de Cohortes , Femenino , Humanos , Trastornos del Metabolismo del Hierro , Ácido Láctico/sangre , Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/metabolismo , Masculino , Metaboloma , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Distrofias Neuroaxonales/diagnóstico , Distrofias Neuroaxonales/enzimología , Neurodegeneración Asociada a Pantotenato Quinasa/enzimología , Neurodegeneración Asociada a Pantotenato Quinasa/genética , Ácido Pantoténico/sangre , Esfingomielinas/sangre , Adulto Joven
4.
Cell Mol Biol (Noisy-le-grand) ; 46(4): 743-60, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10875437

RESUMEN

In several neurodegenerative diseases, iron accumulates at sites of brain pathology. Since post-mortem examination cannot distinguish whether iron accumulation caused the damage or resulted from damage, it is necessary to manipulate iron in animal and tissue culture models to assess its causal role(s). However, only in models of Parkinson's disease and of global ischemia, iron deprivation (ID) or iron-chelators have been used to protect from damage. In these studies, documentation of microgliosis was not performed even though several lines of evidence converge to suggest that activation of microglia is an important source of oxidative stress. In the kainate model of epilepsy, we found that ID protected the olfactory cortex, thalamus and hippocampus and attenuated microgliosis, whereas iron supplementation to ID rats increased damage and microgliosis in the above regions. In the hilus of the hippocampal dentate gyrus, even though no cell loss was observed, ID attenuated microgliosis and iron-supplementation increased it. Thus there is a tight relationship between iron and microgliosis. In addition, iron+zinc supplementation dramatically increased damage to hippocampal CA1 whereas zinc supplementation alone had no effect. This study demonstrates an anatomically unique interaction of iron and zinc, which may lead to new insights to neurodegeneration in epilepsy.


Asunto(s)
Hierro/metabolismo , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Dieta , Epilepsia/metabolismo , Ataxia de Friedreich/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Masculino , Microglía/patología , Esclerosis Múltiple/metabolismo , Enfermedades Neurodegenerativas/etiología , Neuronas/patología , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA