Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Mol Med ; 11(12): e10489, 2019 12.
Article in English | MEDLINE | ID: mdl-31660701

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is an inborn error of CoA metabolism causing dystonia, parkinsonism, and brain iron accumulation. Lack of a good mammalian model has impeded studies of pathogenesis and development of rational therapeutics. We took a new approach to investigating an existing mouse mutant of Pank2 and found that isolating the disease-vulnerable brain revealed regional perturbations in CoA metabolism, iron homeostasis, and dopamine metabolism and functional defects in complex I and pyruvate dehydrogenase. Feeding mice a CoA pathway intermediate, 4'-phosphopantetheine, normalized levels of the CoA-, iron-, and dopamine-related biomarkers as well as activities of mitochondrial enzymes. Human cell changes also were recovered by 4'-phosphopantetheine. We can mechanistically link a defect in CoA metabolism to these secondary effects via the activation of mitochondrial acyl carrier protein, which is essential to oxidative phosphorylation, iron-sulfur cluster biogenesis, and mitochondrial fatty acid synthesis. We demonstrate the fidelity of our model in recapitulating features of the human disease. Moreover, we identify pharmacodynamic biomarkers, provide insights into disease pathogenesis, and offer evidence for 4'-phosphopantetheine as a candidate therapeutic for PKAN.


Subject(s)
Coenzyme A/metabolism , Dopamine/metabolism , Iron/metabolism , Pantetheine/analogs & derivatives , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Animals , Biomarkers/metabolism , Genotype , Mice , Pantetheine/pharmacology , Pantetheine/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/metabolism
2.
Sci Rep ; 7(1): 11260, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900161

ABSTRACT

Coenzyme A is an essential metabolite known for its central role in over one hundred cellular metabolic reactions. In cells, Coenzyme A is synthesized de novo in five enzymatic steps with vitamin B5 as the starting metabolite, phosphorylated by pantothenate kinase. Mutations in the pantothenate kinase 2 gene cause a severe form of neurodegeneration for which no treatment is available. One therapeutic strategy is to generate Coenzyme A precursors downstream of the defective step in the pathway. Here we describe the synthesis, characteristics and in vivo rescue potential of the acetyl-Coenzyme A precursor S-acetyl-4'-phosphopantetheine as a possible treatment for neurodegeneration associated with pantothenate kinase deficiency.


Subject(s)
Heredodegenerative Disorders, Nervous System/drug therapy , Pantetheine/analogs & derivatives , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Serum/chemistry , Animals , Cell Line , Disease Models, Animal , Drosophila , Humans , Mice , Pantetheine/administration & dosage , Pantetheine/chemical synthesis , Pantetheine/isolation & purification , Pantetheine/pharmacokinetics , Treatment Outcome
3.
Am J Hum Genet ; 99(6): 1229-1244, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27817865

ABSTRACT

Mitochondrial fatty acid synthesis (mtFAS) is an evolutionarily conserved pathway essential for the function of the respiratory chain and several mitochondrial enzyme complexes. We report here a unique neurometabolic human disorder caused by defective mtFAS. Seven individuals from five unrelated families presented with childhood-onset dystonia, optic atrophy, and basal ganglia signal abnormalities on MRI. All affected individuals were found to harbor recessive mutations in MECR encoding the mitochondrial trans-2-enoyl-coenzyme A-reductase involved in human mtFAS. All six mutations are extremely rare in the general population, segregate with the disease in the families, and are predicted to be deleterious. The nonsense c.855T>G (p.Tyr285∗), c.247_250del (p.Asn83Hisfs∗4), and splice site c.830+2_830+3insT mutations lead to C-terminal truncation variants of MECR. The missense c.695G>A (p.Gly232Glu), c.854A>G (p.Tyr285Cys), and c.772C>T (p.Arg258Trp) mutations involve conserved amino acid residues, are located within the cofactor binding domain, and are predicted by structural analysis to have a destabilizing effect. Yeast modeling and complementation studies validated the pathogenicity of the MECR mutations. Fibroblast cell lines from affected individuals displayed reduced levels of both MECR and lipoylated proteins as well as defective respiration. These results suggest that mutations in MECR cause a distinct human disorder of the mtFAS pathway. The observation of decreased lipoylation raises the possibility of a potential therapeutic strategy.


Subject(s)
Dystonic Disorders/genetics , Fatty Acids/biosynthesis , Mitochondria/metabolism , Mutation , Optic Atrophy/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Basal Ganglia/metabolism , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts , Genetic Complementation Test , Humans , Infant , Male , Mitochondrial Diseases/genetics , Models, Molecular , Mutation, Missense/genetics , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pedigree , RNA Splice Sites/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
4.
Mol Genet Metab ; 116(4): 289-97, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26547561

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is a progressive movement disorder that is due to mutations in PANK2. Pathologically, it is a member of a class of diseases known as neurodegeneration with brain iron accumulation (NBIA) and features increased tissue iron and ubiquitinated proteinaceous aggregates in the globus pallidus. We have previously determined that these aggregates represent condensed residue derived from degenerated pallidal neurons. However, the protein content, other than ubiquitin, of these aggregates remains unknown. In the present study, we performed biochemical and immunohistochemical studies to characterize these aggregates and found them to be enriched in apolipoprotein E that is poorly soluble in detergent solutions. However, we did not determine a significant association between APOE genotype and the clinical phenotype of disease in our database of 81 cases. Rather, we frequently identified similar ubiquitin- and apolipoprotein E-enriched lesions in these neurons in non-PKAN patients in the penumbrae of remote infarcts that involve the globus pallidus, and occasionally in other brain sites that contain large γ-aminobutyric acid (GABA)ergic neurons. Our findings, taken together, suggest that tissue or cellular hypoxic/ischemic injury within the globus pallidus may underlie the pathogenesis of PKAN.


Subject(s)
Apolipoproteins E/chemistry , Brain Ischemia/genetics , GABAergic Neurons/chemistry , Pantothenate Kinase-Associated Neurodegeneration/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Aggregation, Pathological/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Case-Control Studies , Child , Female , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression , Globus Pallidus/metabolism , Globus Pallidus/pathology , Humans , Male , Middle Aged , Mutation , Pantothenate Kinase-Associated Neurodegeneration/complications , Pantothenate Kinase-Associated Neurodegeneration/metabolism , Pantothenate Kinase-Associated Neurodegeneration/pathology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Protein Aggregation, Pathological/complications , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/metabolism
5.
Am J Hum Genet ; 94(1): 11-22, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24360804

ABSTRACT

Neurodegeneration with brain iron accumulation (NBIA) comprises a clinically and genetically heterogeneous group of disorders with progressive extrapyramidal signs and neurological deterioration, characterized by iron accumulation in the basal ganglia. Exome sequencing revealed the presence of recessive missense mutations in COASY, encoding coenzyme A (CoA) synthase in one NBIA-affected subject. A second unrelated individual carrying mutations in COASY was identified by Sanger sequence analysis. CoA synthase is a bifunctional enzyme catalyzing the final steps of CoA biosynthesis by coupling phosphopantetheine with ATP to form dephospho-CoA and its subsequent phosphorylation to generate CoA. We demonstrate alterations in RNA and protein expression levels of CoA synthase, as well as CoA amount, in fibroblasts derived from the two clinical cases and in yeast. This is the second inborn error of coenzyme A biosynthesis to be implicated in NBIA.


Subject(s)
Brain/drug effects , Exome , Iron/metabolism , Nerve Degeneration/pathology , Brain/pathology , Cloning, Molecular , Coenzyme A/metabolism , Escherichia coli/genetics , Female , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Male , Mitochondria/enzymology , Mitochondria/genetics , Mutation, Missense , Pantetheine/analogs & derivatives , Pantetheine/metabolism , Pedigree , Phosphorylation , Saccharomyces cerevisiae/genetics , Transferases/genetics , Transferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...