Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neuromuscul Dis ; 7(3): 301-308, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32444556

RESUMO

BACKGROUND: In 2009, we identified TACO1 as a novel mitochondrial disease gene in a single family, however no second family has been described to confirm the role of TACO1 in mitochondrial disease. OBJECTIVE: In this report, we describe two independent consanguineous families carrying pathogenic variants in TACO1, confirming the phenotype. METHODS: Detailed clinical investigations and whole exome sequencing with haplotype analysis have been performed in several members of the two reported families. RESULTS: Clinical phenotype of the patients confirms the originally reported phenotype of a childhood-onset progressive cerebellar and pyramidal syndrome with optic atrophy and learning difficulties. Brain MRI showed periventricular white matter lesions with multiple cystic defects, suggesting leukoencephalopathy in both patients. One patient carried the previously described homozygous TACO1 variant (p.His158ProfsTer8) and haplotype analysis suggested that this variant is a rare founder mutation. The second patient from another family carried a homozygous novel frame shift variant (p.Cys85PhefsTer15). CONCLUSIONS: The identification of two Turkish families with similar characteristic clinical presentation and an additional homozygous nonsense mutation confirms that TACO1 is a human mitochondrial disease gene. Although most patients with this clinical presentation undergo next generation sequencing analysis, screening for selected founder mutations in the Turkish population based on the precise clinical presentation may reduce time and cost of finding the genetic diagnosis even in the era of massively parallel sequencing.


Assuntos
Doença de Leigh/genética , Proteínas Mitocondriais/genética , Fatores de Transcrição/genética , Adolescente , Adulto , Consanguinidade , Feminino , Humanos , Doença de Leigh/diagnóstico por imagem , Doença de Leigh/patologia , Doença de Leigh/fisiopatologia , Masculino , Linhagem , Turquia
2.
Mov Disord ; 33(7): 1119-1129, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29603387

RESUMO

BACKGROUND: Spinocerebellar ataxia type 14 is a rare form of autosomal dominant cerebellar ataxia caused by mutations in protein kinase Cγ gene. Clinically, it presents with a slowly progressive, mainly pure cerebellar ataxia. METHODS: Using next generation sequencing, we screened 194 families with autosomal dominant cerebellar ataxia and normal polyglutamine repeats. In-depth phenotyping was performed using validated clinical rating scales neuroimaging and electrophysiological investigations. RESULTS: We identified 25 individuals from 13 families carrying pathogenic mutations in protein kinase Cγ gene. A total of 10 unique protein kinase Cγ gene mutations have been confirmed of which 5 are novel and 5 were previously described. Our data suggest that the age at onset is highly variable; disease course is slowly progressive and rarely associated with severe disability. However, one third of patients presented with a complex ataxia comprising severe focal and/or task-induced dystonia, peripheral neuropathy, parkinsonism, myoclonus, and pyramidal syndrome. The most complex phenotype is related to a missense mutation in the catalytic domain in exon 11. CONCLUSION: We present one of the largest genetically confirmed spinocerebellar ataxia type 14 cohorts contributing novel variants and clinical characterisation. We show that although protein kinase Cγ gene mutations present mainly as slowly progressive pure ataxia, more than a third of cases had a complex phenotype. Overall, our case series extends the phenotype and suggests that protein kinase Cγ gene mutations should be considered in patients with slowly progressive autosomal dominant cerebellar ataxia, particularly when myoclonus, dystonia, or mild cognitive impairment are present in the absence of polyglutamine expansion. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Distonia/etiologia , Mutação de Sentido Incorreto/genética , Peptídeos/genética , Proteína Quinase C/genética , Ataxias Espinocerebelares/complicações , Ataxias Espinocerebelares/genética , Adulto , Idade de Início , Idoso , Pré-Escolar , Estudos de Coortes , Cisteína/genética , Progressão da Doença , Saúde da Família , Feminino , Estudos de Associação Genética , Testes Genéticos , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Fenótipo , Ataxias Espinocerebelares/diagnóstico por imagem , Adulto Jovem
3.
Handb Clin Neurol ; 145: 157-166, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28987166

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) describes a heterogeneous group of inherited rare clinical and genetic entities. Clinical core symptoms comprise a combination of early-onset dystonia, pyramidal and extrapyramidal signs with ataxia, cognitive decline, behavioral abnormalities, and retinal and axonal neuropathy variably accompanying these core features. Increased nonphysiologic, nonaging-associated brain iron, most pronounced in the basal ganglia, is often termed the unifying characteristic of these clinically variable disorders, though occurrence and extent can be fluctuating or even absent. Neuropathologically, NBIA disorders usually are associated with widespread axonal spheroids and local iron accumulation in the basal ganglia. Postmortem, Lewy body, TDP-43, or tau pathology has been observed. Genetics have fostered ongoing progress in elucidating underlying pathophysiologic mechanisms of NBIA disorders. Ten associated genes have been established, with many more being suggested as new technologies and data emerge. Clinically, certain symptom combinations can suggest a specific genetic defect. Genetic tests, combined with postmortem neuropathology, usually make for the final disease confirmation. Despite these advances, treatment to date remains mainly symptomatic. This chapter reviews the established genetic defects leading to different NBIA subtypes, highlights phenotypic presentations to direct genetic testing, and briefly discusses the scarce available treatment options and upcoming challenges and future hopes of the field.


Assuntos
Encéfalo/metabolismo , Distúrbios do Metabolismo do Ferro , Ferro/metabolismo , Mutação/genética , Distrofias Neuroaxonais , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Humanos , Distúrbios do Metabolismo do Ferro/genética , Distúrbios do Metabolismo do Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Distrofias Neuroaxonais/genética , Distrofias Neuroaxonais/metabolismo , Distrofias Neuroaxonais/patologia
4.
PLoS One ; 12(9): e0184104, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28863176

RESUMO

Mutations in PANK2 lead to neurodegeneration with brain iron accumulation. PANK2 has a role in the biosynthesis of coenzyme A (CoA) from dietary vitamin B5, but the neuropathological mechanism and reasons for iron accumulation remain unknown. In this study, atypical patient-derived fibroblasts were reprogrammed into induced pluripotent stem cells (iPSCs) and subsequently differentiated into cortical neuronal cells for studying disease mechanisms in human neurons. We observed no changes in PANK2 expression between control and patient cells, but a reduction in protein levels was apparent in patient cells. CoA homeostasis and cellular iron handling were normal, mitochondrial function was affected; displaying activated NADH-related and inhibited FADH-related respiration, resulting in increased mitochondrial membrane potential. This led to increased reactive oxygen species generation and lipid peroxidation in patient-derived neurons. These data suggest that mitochondrial deficiency is an early feature of the disease process and can be explained by altered NADH/FADH substrate supply to oxidative phosphorylation. Intriguingly, iron chelation appeared to exacerbate the mitochondrial phenotype in both control and patient neuronal cells. This raises caution for the use iron chelation therapy in general when iron accumulation is absent.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Doenças Mitocondriais/fisiopatologia , Neurodegeneração Associada a Pantotenato-Quinase/fisiopatologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Acetilcoenzima A/química , Adolescente , Biópsia , Encéfalo/metabolismo , Diferenciação Celular , Criança , Coenzima A/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Ferro/química , Cariotipagem , Peroxidação de Lipídeos , Masculino , Potencial da Membrana Mitocondrial , Mitocôndrias/patologia , Mutação , NAD/química , Neurônios/metabolismo , Ácido Pantotênico/química , Fenótipo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Plasmídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
5.
J Neurol Neurosurg Psychiatry ; 88(7): 575-585, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28501821

RESUMO

OBJECTIVES: To analyse and describe the clinical and genetic spectrum of Charcot-Marie-Tooth disease (CMT) caused by mutations in the neurofilament light polypeptide gene (NEFL). METHODS: Combined analysis of newly identified patients with NEFL-related CMT and all previously reported cases from the literature. RESULTS: Five new unrelated patients with CMT carrying the NEFL mutations P8R and N98S and the novel variant L311P were identified. Combined data from these cases and 62 kindreds from the literature revealed four common mutations (P8R, P22S, N98S and E396K) and three mutational hotspots accounting for 37 (55%) and 50 (75%) kindreds, respectively. Eight patients had de novo mutations. Loss of large-myelinated fibres was a uniform feature in a total of 21 sural nerve biopsies and 'onion bulb' formations and/or thin myelin sheaths were observed in 14 (67%) of them. The neurophysiological phenotype was broad but most patients with E90K and N98S had upper limb motor conduction velocities <38 m/s. Age of onset was ≤3 years in 25 cases. Pyramidal tract signs were described in 13 patients and 7 patients were initially diagnosed with or tested for inherited ataxia. Patients with E90K and N98S frequently presented before age 3 years and developed hearing loss or other neurological features including ataxia and/or cerebellar atrophy on brain MRI. CONCLUSIONS: NEFL-related CMT is clinically and genetically heterogeneous. Based on this study, however, we propose mutational hotspots and relevant clinical-genetic associations that may be helpful in the evaluation of NEFL sequence variants and the differential diagnosis with other forms of CMT.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Mutação/genética , Proteínas de Neurofilamentos/genética , Axônios/patologia , Ataxia Cerebelar/genética , Ataxia Cerebelar/patologia , Genótipo , Humanos , Linhagem , Fenótipo , Nervo Sural/patologia
6.
Brain ; 140(4): 940-952, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334956

RESUMO

PRUNE is a member of the DHH (Asp-His-His) phosphoesterase protein superfamily of molecules important for cell motility, and implicated in cancer progression. Here we investigated multiple families from Oman, India, Iran and Italy with individuals affected by a new autosomal recessive neurodevelopmental and degenerative disorder in which the cardinal features include primary microcephaly and profound global developmental delay. Our genetic studies identified biallelic mutations of PRUNE1 as responsible. Our functional assays of disease-associated variant alleles revealed impaired microtubule polymerization, as well as cell migration and proliferation properties, of mutant PRUNE. Additionally, our studies also highlight a potential new role for PRUNE during microtubule polymerization, which is essential for the cytoskeletal rearrangements that occur during cellular division and proliferation. Together these studies define PRUNE as a molecule fundamental for normal human cortical development and define cellular and clinical consequences associated with PRUNE mutation.


Assuntos
Encéfalo/crescimento & desenvolvimento , Proteínas de Transporte/genética , Deficiências do Desenvolvimento/genética , Microcefalia/genética , Adolescente , Diferenciação Celular/genética , Movimento Celular/genética , Córtex Cerebral/crescimento & desenvolvimento , Criança , Pré-Escolar , Citoesqueleto/genética , Citoesqueleto/ultraestrutura , Feminino , Genes Recessivos , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Humanos , Lactente , Masculino , Microtúbulos/genética , Microtúbulos/ultraestrutura , Mutação/genética , Linhagem , Monoéster Fosfórico Hidrolases , Adulto Jovem
7.
Mov Disord Clin Pract ; 4(2): 254-257, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30838262

RESUMO

Neurodegeneration with brain iron accumulation (NBIA) mostly has its disease onset in childhood, adolescence, or early adulthood and usually presents with predominant bulbar and axial dystonia along with signs such as spasticity, indicating an involvement of additional neurological systems. Because of their early onset and presentation with a combination of dystonia plus other neurological symptoms, they are usually not considered as differential diagnosis for late-onset isolated (idiopathic) craniocervical dystonia. In this case series, we present 4 genetically proven cases of NBIA (including neuroferritinopathy, pantothenate-kinase-associated neurodegeneration, and aceruloplasminemia) with late disease onset, which resembled isolated adult-onset craniocervical dystonia at disease onset. We also want to highlight the importance of taking NBIA into consideration when dealing with putatively isolated late-onset dystonias and of picking up unusual signs at later stages of the disease.

8.
Nat Genet ; 49(2): 223-237, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27992417

RESUMO

Histone lysine methylation, mediated by mixed-lineage leukemia (MLL) proteins, is now known to be critical in the regulation of gene expression, genomic stability, cell cycle and nuclear architecture. Despite MLL proteins being postulated as essential for normal development, little is known about the specific functions of the different MLL lysine methyltransferases. Here we report heterozygous variants in the gene KMT2B (also known as MLL4) in 27 unrelated individuals with a complex progressive childhood-onset dystonia, often associated with a typical facial appearance and characteristic brain magnetic resonance imaging findings. Over time, the majority of affected individuals developed prominent cervical, cranial and laryngeal dystonia. Marked clinical benefit, including the restoration of independent ambulation in some cases, was observed following deep brain stimulation (DBS). These findings highlight a clinically recognizable and potentially treatable form of genetic dystonia, demonstrating the crucial role of KMT2B in the physiological control of voluntary movement.


Assuntos
Distonia/genética , Histona-Lisina N-Metiltransferase/genética , Mutação/genética , Adolescente , Proteínas de Ligação a DNA/genética , Feminino , Histona Metiltransferases , Histonas/genética , Humanos , Lisina/genética , Masculino , Metilação , Proteínas Nucleares/genética
9.
Cell Rep ; 16(1): 79-91, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27320912

RESUMO

A CAPN1 missense mutation in Parson Russell Terrier dogs is associated with spinocerebellar ataxia. We now report that homozygous or heterozygous CAPN1-null mutations in humans result in cerebellar ataxia and limb spasticity in four independent pedigrees. Calpain-1 knockout (KO) mice also exhibit a mild form of ataxia due to abnormal cerebellar development, including enhanced neuronal apoptosis, decreased number of cerebellar granule cells, and altered synaptic transmission. Enhanced apoptosis is due to absence of calpain-1-mediated cleavage of PH domain and leucine-rich repeat protein phosphatase 1 (PHLPP1), which results in inhibition of the Akt pro-survival pathway in developing granule cells. Injection of neonatal mice with the indirect Akt activator, bisperoxovanadium, or crossing calpain-1 KO mice with PHLPP1 KO mice prevented increased postnatal cerebellar granule cell apoptosis and restored granule cell density and motor coordination in adult mice. Thus, mutations in CAPN1 are an additional cause of ataxia in mammals, including humans.


Assuntos
Calpaína/genética , Ataxia Cerebelar/genética , Cerebelo/embriologia , Cerebelo/metabolismo , Envelhecimento/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Apoptose , Calpaína/química , Calpaína/metabolismo , Contagem de Células , Ataxia Cerebelar/patologia , Ataxia Cerebelar/fisiopatologia , Cerebelo/patologia , Cerebelo/fisiopatologia , Ativação Enzimática , Feminino , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Deficiência Intelectual/fisiopatologia , Masculino , Camundongos Knockout , Atividade Motora , Espasticidade Muscular/genética , Espasticidade Muscular/patologia , Espasticidade Muscular/fisiopatologia , Mutação/genética , Proteínas Nucleares/metabolismo , Atrofia Óptica/genética , Atrofia Óptica/patologia , Atrofia Óptica/fisiopatologia , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células de Purkinje/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/fisiopatologia , Transmissão Sináptica
10.
Am J Hum Genet ; 98(4): 763-71, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27058447

RESUMO

Chorea is a hyperkinetic movement disorder resulting from dysfunction of striatal medium spiny neurons (MSNs), which form the main output projections from the basal ganglia. Here, we used whole-exome sequencing to unravel the underlying genetic cause in three unrelated individuals with a very similar and unique clinical presentation of childhood-onset chorea and characteristic brain MRI showing symmetrical bilateral striatal lesions. All individuals were identified to carry a de novo heterozygous mutation in PDE10A (c.898T>C [p.Phe300Leu] in two individuals and c.1000T>C [p.Phe334Leu] in one individual), encoding a phosphodiesterase highly and selectively present in MSNs. PDE10A contributes to the regulation of the intracellular levels of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both substitutions affect highly conserved amino acids located in the regulatory GAF-B domain, which, by binding to cAMP, stimulates the activity of the PDE10A catalytic domain. In silico modeling showed that the altered residues are located deep in the binding pocket, where they are likely to alter cAMP binding properties. In vitro functional studies showed that neither substitution affects the basal PDE10A activity, but they severely disrupt the stimulatory effect mediated by cAMP binding to the GAF-B domain. The identification of PDE10A mutations as a cause of chorea further motivates the study of cAMP signaling in MSNs and highlights the crucial role of striatal cAMP signaling in the regulation of basal ganglia circuitry. Pharmacological modulation of this pathway could offer promising etiologically targeted treatments for chorea and other hyperkinetic movement disorders.


Assuntos
Coreia/genética , Corpo Estriado/patologia , Mutação , Diester Fosfórico Hidrolases/genética , Sequência de Aminoácidos , Animais , Criança , Coreia/diagnóstico , Corpo Estriado/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Conformação Proteica , Alinhamento de Sequência , Transdução de Sinais , Adulto Jovem
11.
Neurobiol Dis ; 87: 59-68, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26707700

RESUMO

Aberrant brain iron deposition is observed in both common and rare neurodegenerative disorders, including those categorized as Neurodegeneration with Brain Iron Accumulation (NBIA), which are characterized by focal iron accumulation in the basal ganglia. Two NBIA genes are directly involved in iron metabolism, but whether other NBIA-related genes also regulate iron homeostasis in the human brain, and whether aberrant iron deposition contributes to neurodegenerative processes remains largely unknown. This study aims to expand our understanding of these iron overload diseases and identify relationships between known NBIA genes and their main interacting partners by using a systems biology approach. We used whole-transcriptome gene expression data from human brain samples originating from 101 neuropathologically normal individuals (10 brain regions) to generate weighted gene co-expression networks and cluster the 10 known NBIA genes in an unsupervised manner. We investigated NBIA-enriched networks for relevant cell types and pathways, and whether they are disrupted by iron loading in NBIA diseased tissue and in an in vivo mouse model. We identified two basal ganglia gene co-expression modules significantly enriched for NBIA genes, which resemble neuronal and oligodendrocytic signatures. These NBIA gene networks are enriched for iron-related genes, and implicate synapse and lipid metabolism related pathways. Our data also indicates that these networks are disrupted by excessive brain iron loading. We identified multiple cell types in the origin of NBIA disorders. We also found unforeseen links between NBIA networks and iron-related processes, and demonstrate convergent pathways connecting NBIAs and phenotypically overlapping diseases. Our results are of further relevance for these diseases by providing candidates for new causative genes and possible points for therapeutic intervention.


Assuntos
Gânglios da Base/metabolismo , Perfilação da Expressão Gênica/métodos , Distúrbios do Metabolismo do Ferro/metabolismo , Distrofias Neuroaxonais/metabolismo , Transcriptoma , Adolescente , Idoso , Idoso de 80 Anos ou mais , Animais , Gânglios da Base/patologia , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Lactente , Ferro/metabolismo , Distúrbios do Metabolismo do Ferro/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Distrofias Neuroaxonais/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Substância Branca/metabolismo , Substância Branca/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA