Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Orphanet J Rare Dis ; 18(1): 80, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37046296

RESUMO

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.


Assuntos
Doenças Neurodegenerativas , Neurodegeneração Associada a Pantotenato-Quinase , Ácido Tióctico , Humanos , Suplementos Nutricionais , Ferro/metabolismo , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/genética , Neurodegeneração Associada a Pantotenato-Quinase/tratamento farmacológico , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ácido Tióctico/uso terapêutico , Ácido Tióctico/metabolismo
2.
Orphanet J Rare Dis ; 17(1): 111, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35246191

RESUMO

BACKGROUND: Neurodegeneration with brain iron accumulation describes a group of rare heterogeneous progressive neurodegenerative disorders characterized by excessive iron accumulation in the basal ganglia region. Pantothenate kinase-associated neurodegeneration (PKAN) is a major form of this disease. RESULTS: A total of 7 unrelated patients were diagnosed with PKAN in a single tertiary center from August 2009 to February 2018. Ten variants in PANK2 including three novel sequence variants and one large exonic deletion were detected. Sequencing of the breakpoint was performed to predict the mechanism of large deletion and AluSx3 and AluSz6 were found with approximately 97.3% sequence homology. CONCLUSION: The findings support the disease-causing role of PANK2 and indicate the possibility that exonic deletion of PANK2 found in PKAN is mediated through Alu-mediated homologous recombination.


Assuntos
Mutação , Neurodegeneração Associada a Pantotenato-Quinase , Humanos , Ferro/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
3.
Cell Death Dis ; 13(2): 185, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217637

RESUMO

Neurodegeneration associated with defective pantothenate kinase-2 (PKAN) is an early-onset monogenic autosomal-recessive disorder. The hallmark of the disease is the massive accumulation of iron in the globus pallidus brain region of patients. PKAN is caused by mutations in the PANK2 gene encoding the mitochondrial enzyme pantothenate kinase-2, whose function is to catalyze the first reaction of the CoA biosynthetic pathway. To date, the way in which this alteration leads to brain iron accumulation has not been elucidated. Starting from previously obtained hiPS clones, we set up a differentiation protocol able to generate inhibitory neurons. We obtained striatal-like medium spiny neurons composed of approximately 70-80% GABAergic neurons and 10-20% glial cells. Within this mixed population, we detected iron deposition in both PKAN cell types, however, the viability of PKAN GABAergic neurons was strongly affected. CoA treatment was able to reduce cell death and, notably, iron overload. Further differentiation of hiPS clones in a pure population of astrocytes showed particularly evident iron accumulation, with approximately 50% of cells positive for Perls staining. The analysis of these PKAN astrocytes indicated alterations in iron metabolism, mitochondrial morphology, respiratory activity, and oxidative status. Moreover, PKAN astrocytes showed signs of ferroptosis and were prone to developing a stellate phenotype, thus gaining neurotoxic features. This characteristic was confirmed in iPS-derived astrocyte and glutamatergic neuron cocultures, in which PKAN glutamatergic neurons were less viable in the presence of PKAN astrocytes. This newly generated astrocyte model is the first in vitro disease model recapitulating the human phenotype and can be exploited to deeply clarify the pathogenetic mechanisms underlying the disease.


Assuntos
Astrócitos , Neurodegeneração Associada a Pantotenato-Quinase , Astrócitos/metabolismo , Coenzima A/genética , Coenzima A/metabolismo , Humanos , Ferro/metabolismo , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
4.
J Biol Chem ; 298(3): 101577, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35041826

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is an incurable rare genetic disorder of children and young adults caused by mutations in the PANK2 gene, which encodes an enzyme critical for the biosynthesis of coenzyme A. Although PKAN affects only a small number of patients, it shares several hallmarks of more common neurodegenerative diseases of older adults such as Alzheimer's disease and Parkinson's disease. Advances in etiological understanding and treatment of PKAN could therefore have implications for our understanding of more common diseases and may shed new lights on the physiological importance of coenzyme A, a cofactor critical for the operation of various cellular metabolic processes. The large body of knowledge that accumulated over the years around PKAN pathology, including but not limited to studies of various PKAN models and therapies, has contributed not only to progress in our understanding of the disease but also, importantly, to the crystallization of key questions that guide future investigations of the disease. In this review, we will summarize this knowledge and demonstrate how it forms the backdrop to new avenues of research.


Assuntos
Doenças Neurodegenerativas , Neurodegeneração Associada a Pantotenato-Quinase , Animais , Coenzima A/genética , Coenzima A/metabolismo , Humanos , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Neurodegeneração Associada a Pantotenato-Quinase/terapia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
5.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36613877

RESUMO

Human PANK1, PANK2, and PANK3 genes encode several pantothenate kinase isoforms that catalyze the phosphorylation of vitamin B5 (pantothenic acid) to phosphopantothenate, a critical step in the biosynthesis of the major cellular cofactor, Coenzyme A (CoA). Mutations in the PANK2 gene, which encodes the mitochondrial pantothenate kinase (PanK) isoform, have been linked to pantothenate-kinase associated neurodegeneration (PKAN), a debilitating and often fatal progressive neurodegeneration of children and young adults. While the biochemical properties of these enzymes have been well-characterized in vitro, their expression in a model organism such as yeast in order to probe their function under cellular conditions have never been achieved. Here we used three yeast mutants carrying missense mutations in the yeast PanK gene, CAB1, which are associated with defective growth at high temperature and iron, mitochondrial dysfunction, increased iron content, and oxidative stress, to assess the cellular function of human PANK genes and functional conservation of the CoA-controlled processes between humans and yeast. Overexpression of human PANK1 and PANK3 in these mutants restored normal cellular activity whereas complementation with PANK2 was partial and could only be achieved with an isoform, PanK2mtmΔ, lacking the mitochondrial transit peptide. These data, which demonstrate functional conservation of PanK activity between humans and yeast, set the stage for the use of yeast as a model system to investigate the impact of PKAN-associated mutations on the metabolic pathways altered in this disease.


Assuntos
Estresse Oxidativo , Neurodegeneração Associada a Pantotenato-Quinase , Saccharomyces cerevisiae , Humanos , Homeostase , Ferro/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Estresse Oxidativo/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Ácido Pantotênico , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
6.
Bull Exp Biol Med ; 170(1): 58-63, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33237527

RESUMO

Mutations in pank2 gene encoding pantothenate kinase 2 determine a pantothenate kinase-associated neurodegeneration, a rare disorder characterized by iron deposition in the globus pallidus. To extend our previous work, we performed microinjections of a new pank2-specific morpholino to zebrafish embryos and thoroughly analyzed vasculature development. Vessels development was severely perturbed in the head, trunk, and tail, where blood accumulation was remarkable and associated with dilation of the posterior cardinal vein. This phenotype was specific as confirmed by p53 expression analysis and injection of the same morpholino in pank2-mutant embryos. We can conclude that pank2 gene is involved in vasculature development in zebrafish embryos. The comprehension of the underlining mechanisms could be of relevance for understanding of pantothenate kinase-associated neurodegeneration.


Assuntos
Vasos Sanguíneos/metabolismo , Coenzima A/farmacologia , Globo Pálido/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/prevenção & controle , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Globo Pálido/irrigação sanguínea , Globo Pálido/efeitos dos fármacos , Globo Pálido/patologia , Cabeça/irrigação sanguínea , Cabeça/crescimento & desenvolvimento , Humanos , Morfolinos/administração & dosagem , Morfolinos/genética , Morfolinos/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Cauda/irrigação sanguínea , Cauda/crescimento & desenvolvimento , Cauda/metabolismo , Tronco/irrigação sanguínea , Tronco/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra
7.
Int J Mol Sci ; 21(10)2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32456086

RESUMO

Pantothenate Kinase-associated Neurodegeneration (PKAN) belongs to a wide spectrum of diseases characterized by brain iron accumulation and extrapyramidal motor signs. PKAN is caused by mutations in PANK2, encoding the mitochondrial pantothenate kinase 2, which is the first enzyme of the biosynthesis of Coenzyme A. We established and characterized glutamatergic neurons starting from previously developed PKAN Induced Pluripotent Stem Cells (iPSCs). Results obtained by inductively coupled plasma mass spectrometry indicated a higher amount of total cellular iron in PKAN glutamatergic neurons with respect to controls. PKAN glutamatergic neurons, analyzed by electron microscopy, exhibited electron dense aggregates in mitochondria that were identified as granules containing calcium phosphate. Calcium homeostasis resulted compromised in neurons, as verified by monitoring the activity of calcium-dependent enzyme calpain1, calcium imaging and voltage dependent calcium currents. Notably, the presence of calcification in the internal globus pallidus was confirmed in seven out of 15 genetically defined PKAN patients for whom brain CT scan was available. Moreover, we observed a higher prevalence of brain calcification in females. Our data prove that high amount of iron coexists with an impairment of cytosolic calcium in PKAN glutamatergic neurons, indicating both, iron and calcium dys-homeostasis, as actors in pathogenesis of the disease.


Assuntos
Cálcio/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Adolescente , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Cálcio/efeitos adversos , Calpaína/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Citoplasma/fisiologia , Feminino , Homeostase , Humanos , Células-Tronco Pluripotentes Induzidas , Lactente , Ferro/efeitos adversos , Imageamento por Ressonância Magnética , Masculino , Espectrometria de Massas , Microscopia Eletrônica , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool) , Tomografia Computadorizada por Raios X , Adulto Jovem
8.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33396642

RESUMO

Mutations in the pantothenate kinase 2 gene (PANK2) are the cause of pantothenate kinase-associated neurodegeneration (PKAN), the most common form of neurodegeneration with brain iron accumulation. Although different disease models have been created to investigate the pathogenic mechanism of PKAN, the cascade of molecular events resulting from CoA synthesis impairment is not completely understood. Moreover, for PKAN disease, only symptomatic treatments are available. Despite the lack of a neural system, Saccharomyces cerevisiae has been successfully used to decipher molecular mechanisms of many human disorders including neurodegenerative diseases as well as iron-related disorders. To gain insights into the molecular basis of PKAN, a yeast model of this disease was developed: a yeast strain with the unique gene encoding pantothenate kinase CAB1 deleted, and expressing a pathological variant of this enzyme. A detailed functional characterization demonstrated that this model recapitulates the main phenotypes associated with human disease: mitochondrial dysfunction, altered lipid metabolism, iron overload, and oxidative damage suggesting that the yeast model could represent a tool to provide information on pathophysiology of PKAN. Taking advantage of the impaired oxidative growth of this mutant strain, a screening for molecules able to rescue this phenotype was performed. Two molecules in particular were able to restore the multiple defects associated with PKAN deficiency and the rescue was not allele-specific. Furthermore, the construction and characterization of a set of mutant alleles, allowing a quick evaluation of the biochemical consequences of pantothenate kinase (PANK) protein variants could be a tool to predict genotype/phenotype correlation.


Assuntos
Predisposição Genética para Doença/genética , Modelos Genéticos , Mutação , Neurodegeneração Associada a Pantotenato-Quinase/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Saccharomyces cerevisiae/genética , Humanos , Ferro/metabolismo , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/genética , Peroxidação de Lipídeos/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oxirredução , Neurodegeneração Associada a Pantotenato-Quinase/tratamento farmacológico , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/metabolismo
9.
EMBO Mol Med ; 11(12): e10489, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31660701

RESUMO

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.


Assuntos
Coenzima A/metabolismo , Dopamina/metabolismo , Ferro/metabolismo , Panteteína/análogos & derivados , Neurodegeneração Associada a Pantotenato-Quinase/tratamento farmacológico , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Animais , Biomarcadores/metabolismo , Genótipo , Camundongos , Panteteína/farmacologia , Panteteína/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
10.
PLoS One ; 13(3): e0192028, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29522513

RESUMO

In cells, phosphorylation of pantothenic acid to generate phosphopantothenic acid by the pantothenate kinase enzymes is the first step in coenzyme A synthesis. Pantothenate kinase 2, the isoform localized in neuronal cell mitochondria, is dysfunctional in patients with pantothenate kinase-associated neurodegeneration. Fosmetpantotenate is a phosphopantothenic acid prodrug in clinical development for treatment of pantothenate kinase-associated neurodegeneration, which aims to replenish phosphopantothenic acid in patients. Fosmetpantotenate restored coenzyme A in short-hairpin RNA pantothenate kinase 2 gene-silenced neuroblastoma cells and was permeable in a blood-brain barrier model. The rate of fosmetpantotenate metabolism in blood is species-dependent. Following up to 700 mg/kg orally, blood exposure to fosmetpantotenate was negligible in rat and mouse, but measurable in monkey. Consistent with the difference in whole blood half-life, fosmetpantotenate dosed orally was found in the brains of the monkey (striatal dialysate) but was absent in mice. Following administration of isotopically labeled-fosmetpantotenate to mice, ~40% of liver coenzyme A (after 500 mg/kg orally) and ~50% of brain coenzyme A (after 125 µg intrastriatally) originated from isotopically labeled-fosmetpantotenate. Additionally, 10-day dosing of isotopically labeled-fosmetpantotenate, 12.5 µg, intracerebroventricularly in mice led to ~30% of brain coenzyme A containing the stable isotopic labels. This work supports the hypothesis that fosmetpantotenate acts to replace reduced phosphopantothenic acid in pantothenate kinase 2-deficient tissues.


Assuntos
Coenzima A/metabolismo , Modelos Animais de Doenças , Neurodegeneração Associada a Pantotenato-Quinase/tratamento farmacológico , Ácido Pantotênico/análogos & derivados , Pró-Fármacos/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Humanos , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Ácido Pantotênico/farmacocinética , Ácido Pantotênico/farmacologia , Ácido Pantotênico/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Interferência de RNA , Ratos Sprague-Dawley , Especificidade da Espécie
11.
Mol Genet Metab ; 116(4): 289-97, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26547561

RESUMO

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.


Assuntos
Apolipoproteínas E/química , Isquemia Encefálica/genética , Neurônios GABAérgicos/química , Neurodegeneração Associada a Pantotenato-Quinase/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Agregação Patológica de Proteínas/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Estudos de Casos e Controles , Criança , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Expressão Gênica , Globo Pálido/metabolismo , Globo Pálido/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neurodegeneração Associada a Pantotenato-Quinase/complicações , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Agregação Patológica de Proteínas/complicações , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/metabolismo
12.
PLoS One ; 10(6): e0130013, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26052948

RESUMO

BACKGROUND: Pantothenate kinase-associated neurodegeneration, PKAN, is an inherited disorder characterized by progressive impairment in motor coordination and caused by mutations in PANK2, a human gene that encodes one of four pantothenate kinase (PanK) isoforms. PanK initiates the synthesis of coenzyme A (CoA), an essential cofactor that plays a key role in energy metabolism and lipid synthesis. Most of the mutations in PANK2 reduce or abolish the activity of the enzyme. This evidence has led to the hypothesis that lower CoA might be the underlying cause of the neurodegeneration in PKAN patients; however, no mouse model of the disease is currently available to investigate the connection between neuronal CoA levels and neurodegeneration. Indeed, genetic and/or dietary manipulations aimed at reducing whole-body CoA synthesis have not produced a desirable PKAN model, and this has greatly hindered the discovery of a treatment for the disease. OBJECTIVE, METHODS, RESULTS AND CONCLUSIONS: Cellular CoA levels are tightly regulated by a balance between synthesis and degradation. CoA degradation is catalyzed by two peroxisomal nudix hydrolases, Nudt7 and Nudt19. In this study we sought to reduce neuronal CoA in mice through the alternative approach of increasing Nudt7-mediated CoA degradation. This was achieved by combining the use of an adeno-associated virus-based expression system with the synapsin (Syn) promoter. We show that mice with neuronal overexpression of a cytosolic version of Nudt7 (scAAV9-Syn-Nudt7cyt) exhibit a significant decrease in brain CoA levels in conjunction with a reduction in motor coordination. These results strongly support the existence of a link between CoA levels and neuronal function and show that scAAV9-Syn-Nudt7cyt mice can be used to model PKAN.


Assuntos
Coenzima A/metabolismo , Atividade Motora , Neurônios/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Pirofosfatases/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Neurodegeneração Associada a Pantotenato-Quinase/fisiopatologia , Pirofosfatases/metabolismo , Nudix Hidrolases
13.
PLoS One ; 10(5): e0125580, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25933379

RESUMO

BACKGROUND: Panthothenate kinase-associated neurodegeneration (PKAN) belongs to a group of hereditary neurodegenerative disorders known as neuroacanthocytosis (NA). This genetically heterogeneous group of diseases is characterized by degeneration of neurons in the basal ganglia and by the presence of deformed red blood cells with thorny protrusions, acanthocytes, in the circulation. OBJECTIVE: The goal of our study is to elucidate the molecular mechanisms underlying this aberrant red cell morphology and the corresponding functional consequences. This could shed light on the etiology of the neurodegeneration. METHODS: We performed a qualitative and semi-quantitative morphological, immunofluorescent, biochemical and functional analysis of the red cells of several patients with PKAN and, for the first time, of the red cells of their family members. RESULTS: We show that the blood of patients with PKAN contains not only variable numbers of acanthocytes, but also a wide range of other misshapen red cells. Immunofluorescent and immunoblot analyses suggest an altered membrane organization, rather than quantitative changes in protein expression. Strikingly, these changes are not limited to the red blood cells of PKAN patients, but are also present in the red cells of heterozygous carriers without neurological problems. Furthermore, changes are not only present in acanthocytes, but also in other red cells, including discocytes. The patients' cells, however, are more fragile, as observed in a spleen-mimicking device. CONCLUSION: These morphological, molecular and functional characteristics of red cells in patients with PKAN and their family members offer new tools for diagnosis and present a window into the pathophysiology of neuroacanthocytosis.


Assuntos
Acantócitos/patologia , Membrana Eritrocítica/patologia , Neuroacantocitose/patologia , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Acantócitos/metabolismo , Acantócitos/ultraestrutura , Adulto , Idoso , Proteína 1 de Troca de Ânion do Eritrócito/genética , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Estudos de Casos e Controles , Forma Celular , Criança , Contagem de Eritrócitos , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/ultraestrutura , Feminino , Expressão Gênica , Heterozigoto , Homozigoto , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Neuroacantocitose/genética , Neuroacantocitose/metabolismo , Neuroacantocitose/fisiopatologia , Fragilidade Osmótica , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/fisiopatologia , Linhagem , Espectrina/genética , Espectrina/metabolismo
14.
J Inherit Metab Dis ; 38(1): 123-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25300979

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) is a group of devastating and life threatening rare diseases. Adult and early-onset NBIA syndromes are inherited as X-chromosomal, autosomal dominant or recessive traits and several genes have been identified as responsible for these disorders. Among the identified disease genes, only two code for proteins directly involved in iron metabolism while the remaining NBIA genes encode proteins with a wide variety of functions ranging from fatty acid metabolism and autophagy to still unknown activities. It is becoming increasingly evident that many neurodegenerative diseases are associated with metabolic dysfunction that often involves altered lipid metabolism. This is not surprising since neurons have a peculiar and heterogeneous lipid composition critical for the development and correct functioning of the nervous system. This review will focus on specific NBIA forms, namely PKAN, CoPAN, PLAN, FAHN and MPAN, which display an interesting link between neurodegeneration and alteration of phospholipids and sphingolipids metabolism, mitochondrial morphology and membrane remodelling.


Assuntos
Encéfalo/metabolismo , Distúrbios do Metabolismo do Ferro/metabolismo , Ferro/química , Metabolismo dos Lipídeos , Distrofias Neuroaxonais/metabolismo , Doenças Neurodegenerativas/metabolismo , Animais , Ácidos Graxos/metabolismo , Humanos , Distúrbios do Metabolismo do Ferro/genética , Mitocôndrias/metabolismo , Oxigenases de Função Mista/metabolismo , Distrofias Neuroaxonais/genética , Doenças Neurodegenerativas/genética , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fosfolipídeos/metabolismo , Esfingolipídeos/metabolismo , Síndrome
15.
Biochem Soc Trans ; 42(4): 1063-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25110003

RESUMO

Inborn errors of CoA (coenzyme A) biosynthesis lead to neurodegenerative disorders in humans. PKAN (pantothenate kinase-associated neurodegeneration) manifests with damage to brain, retina and testis and is caused by mutations in PANK2, the gene encoding the mitochondrial form of pantothenate kinase, a key regulatory enzyme in CoA synthesis. Further attention has been focused on this pathway by the recent discovery that mutations in the gene encoding CoA synthase lead to a similar neurodegenerative disorder, raising the spectre of a common mechanism of pathogenesis. How do defects in CoA production result in neurodegeneration? Why are certain tissues and cell types selectively vulnerable? And what is the underlying neurodegenerative process? Answers to some of these questions have come from animal models of disease, including flies and mice, as well as directly from humans. The damaged tissue types share key features that are likely to contribute to their selective vulnerability. These include the presence of a blood-tissue barrier, the milieu with respect to oxidative stress, tissue metabolic demand, relative expression of genes encoding similar proteins in these tissues and cell membrane composition. Substantial progress in understanding these important neurometabolic disorders has been made since the first gene discovery more than a decade ago. With rational therapeutics now in development for PKAN, we foresee prevention of neurodegeneration and hope for neuroregeneration or neuro-rescue.


Assuntos
Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Ácido Pantotênico/metabolismo , Animais , Coenzima A/metabolismo , Humanos , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
16.
Curr Neurol Neurosci Rep ; 13(12): 413, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24142851

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) comprises a group of brain iron deposition syndromes that lead to mixed extrapyramidal features and progressive dementia. Historically, there has not been a clearly identifiable molecular cause for many patients with clinical and radiologic features of NBIA. Recent discoveries have shown that mutations in C19orf12 or WDR45 can lead to NBIA. C19orf12 mutations are inherited in an autosomal recessive manner, and lead to a syndrome similar to that caused by mutations in PANK2 or PLA2G6. In contrast, WDR45 mutations lead to a distinct form of NBIA characterized by spasticity and intellectual disability in childhood followed by the subacute onset of dystonia-parkinsonism in adulthood. WDR45 mutations act in an X-linked dominant manner. Although the function of C19orf12 is largely unknown, WDR45 plays a key role in autophagy. Each of these new forms of NBIA thus leads to a distinct clinical syndrome, and together they implicate new cellular pathways in the pathogenesis of these disorders.


Assuntos
Química Encefálica/fisiologia , Ferro/metabolismo , Doenças Neurodegenerativas/metabolismo , Encéfalo/patologia , Química Encefálica/genética , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Humanos , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neuroimagem , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo
17.
PLoS One ; 8(10): e76715, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098554

RESUMO

Neuroacanthocytosis (NA) refers to a group of heterogenous, rare genetic disorders, namely chorea acanthocytosis (ChAc), McLeod syndrome (MLS), Huntington's disease-like 2 (HDL2) and pantothenate kinase associated neurodegeneration (PKAN), that mainly affect the basal ganglia and are associated with similar neurological symptoms. PKAN is also assigned to a group of rare neurodegenerative diseases, known as NBIA (neurodegeneration with brain iron accumulation), associated with iron accumulation in the basal ganglia and progressive movement disorder. Acanthocytosis, the occurrence of misshaped erythrocytes with thorny protrusions, is frequently observed in ChAc and MLS patients but less prevalent in PKAN (about 10%) and HDL2 patients. The pathological factors that lead to the formation of the acanthocytic red blood cell shape are currently unknown. The aim of this study was to determine whether NA/NBIA acanthocytes differ in their functionality from normal erythrocytes. Several flow-cytometry-based assays were applied to test the physiological responses of the plasma membrane, namely drug-induced endocytosis, phosphatidylserine exposure and calcium uptake upon treatment with lysophosphatidic acid. ChAc red cell samples clearly showed a reduced response in drug-induced endovesiculation, lysophosphatidic acid-induced phosphatidylserine exposure, and calcium uptake. Impaired responses were also observed in acanthocyte-positive NBIA (PKAN) red cells but not in patient cells without shape abnormalities. These data suggest an "acanthocytic state" of the red cell where alterations in functional and interdependent membrane properties arise together with an acanthocytic cell shape. Further elucidation of the aberrant molecular mechanisms that cause this acanthocytic state may possibly help to evaluate the pathological pathways leading to neurodegeneration.


Assuntos
Acantócitos/patologia , Gânglios da Base/patologia , Membrana Eritrocítica/patologia , Neuroacantocitose/patologia , Neurodegeneração Associada a Pantotenato-Quinase/patologia , Acantócitos/efeitos dos fármacos , Acantócitos/metabolismo , Adolescente , Adulto , Gânglios da Base/metabolismo , Cálcio/metabolismo , Estudos de Casos e Controles , Cátions Bivalentes , Criança , Clorpromazina/farmacologia , Endocitose , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Feminino , Humanos , Imipramina/farmacologia , Transporte de Íons , Lisofosfolipídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Neuroacantocitose/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Primaquina/farmacologia
18.
Orv Hetil ; 154(29): 1156-64, 2013 Jul 21.
Artigo em Húngaro | MEDLINE | ID: mdl-23853349

RESUMO

Iron-overload diseases are typically insidious, causing progressive and irreversible organ injury before clinical symptoms develop. Some iron-overload diseases as HFE-associated hemochromatosis and beta-thalassemia are quite common, whereas others are very rare. Early diagnosis is important since iron toxicity can be attenuated or prevented. Significant progress of our knowledge on iron metabolism developed in the past years. We learned a lot about HFE gene mutations, function of ferroportin and hepcidin, the hypoferremia hormone produced by the liver. However, many questions are still open. Special forms of localized iron overload are the Hallervorden-Spatz syndrome and pantothenate kinase gene mutation associated neurodegeneration causing progressive extrapyramidal movement disorders. Neonatal hemochromatosis is a severe systemic iron-overload disorder due to gestational alloimmune liver disease caused by transplacental maternal IgG directed against the fetal liver. This review article gives an overview on iron metabolism and iron-overload disease. Pathomechanism, diagnosis and treatment of hereditary hemochromatosis are discussed.


Assuntos
Hemocromatose , Hepatócitos/metabolismo , Ferro/metabolismo , Mutação , Neurodegeneração Associada a Pantotenato-Quinase , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Quelantes/uso terapêutico , Dieta , Progressão da Doença , Diagnóstico Precoce , Feminino , Hemocromatose/complicações , Hemocromatose/diagnóstico , Hemocromatose/genética , Hemocromatose/metabolismo , Hemocromatose/terapia , Hepcidinas , Humanos , Imunoglobulina G/imunologia , Recém-Nascido , Macrófagos/metabolismo , Neurodegeneração Associada a Pantotenato-Quinase/diagnóstico , Neurodegeneração Associada a Pantotenato-Quinase/genética , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Flebotomia , Gravidez , Complicações na Gravidez/imunologia
19.
Mol Genet Metab ; 110(3): 336-41, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23891537

RESUMO

Pantothenate kinase-associated neurodegeneration (PKAN) is an autosomal recessive disorder of coenzyme A homeostasis caused by defects in the mitochondrial pantothenate kinase 2. Patients with PKAN present with a progressive neurological decline and brain iron accumulation, but general energy balance and nutrition status among these patients has not been reported. To determine if defects in PANK2 change basic energy metabolism in humans, we measured body composition, resting energy expenditure, dietary intake, and blood metabolites among 16 subjects with PKAN. Subjects had a broad range of disease severity but, despite the essential role of coenzyme A in energy metabolism, the subjects had remarkably normal body composition, dietary intake and energy metabolism compared to population normal values. We did observe increased resting energy expenditure associated with disease severity, suggesting increased energy needs later in the disease process, and elevated urinary mevalonate levels.


Assuntos
Metabolismo Energético , Neurodegeneração Associada a Pantotenato-Quinase/metabolismo , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Composição Corporal , Estudos de Casos e Controles , Criança , Feminino , Humanos , Lipídeos/sangue , Masculino , Ácido Mevalônico/urina , Pessoa de Meia-Idade , Neurodegeneração Associada a Pantotenato-Quinase/sangue , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA