Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Prostaglandins Other Lipid Mediat ; 172: 106836, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38599513

RESUMO

Dravet syndrome is an intractable epilepsy with a high seizure burden that is resistant to current anti-seizure medications. There is evidence that neuroinflammation plays a role in epilepsy and seizures, however few studies have specifically examined neuroinflammation in Dravet syndrome under conditions of a higher seizure burden. Here we used an established genetic mouse model of Dravet syndrome (Scn1a+/- mice), to examine whether a higher seizure burden impacts the number and morphology of microglia in the hippocampus. Moreover, we examined whether a high seizure burden influences classical inflammatory mediators in this brain region. Scn1a+/- mice with a high seizure burden induced by thermal priming displayed a localised reduction in microglial cell density in the granule cell layer and subgranular zone of the dentate gyrus, regions important to postnatal neurogenesis. However, microglial cell number and morphology remained unchanged in other hippocampal subfields. The high seizure burden in Scn1a+/- mice did not affect hippocampal mRNA expression of classical inflammatory mediators such as interleukin 1ß and tumour necrosis factor α, but increased cyclooxygenase 2 (COX-2) expression. We then quantified hippocampal levels of prostanoids that arise from COX-2 mediated metabolism of fatty acids and found that Scn1a+/- mice with a high seizure burden displayed increased hippocampal concentrations of numerous prostaglandins, notably PGF2α, PGE2, PGD2, and 6-K-PGF1A, compared to Scn1a+/- mice with a low seizure burden. In conclusion, a high seizure burden increased hippocampal concentrations of various prostaglandin mediators in a mouse model of Dravet syndrome. Future studies could interrogate the prostaglandin pathways to further better understand their role in the pathophysiology of Dravet syndrome.


Assuntos
Modelos Animais de Doenças , Epilepsias Mioclônicas , Hipocampo , Canal de Sódio Disparado por Voltagem NAV1.1 , Prostaglandinas , Convulsões , Animais , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Camundongos , Hipocampo/metabolismo , Hipocampo/patologia , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.1/metabolismo , Convulsões/metabolismo , Convulsões/genética , Convulsões/patologia , Prostaglandinas/metabolismo , Masculino , Microglia/metabolismo , Microglia/patologia
2.
Epilepsia ; 64(8): e170-e176, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37114479

RESUMO

IRF2BPL has recently been described as a novel cause of neurodevelopmental disorders with multisystemic regression, epilepsy, cerebellar symptoms, dysphagia, dystonia, and pyramidal signs. We describe a novel IRF2BPL phenotype consistent with progressive myoclonus epilepsy (PME) in three novel subjects and review the features of the 31 subjects with IRF2BPL-related disorders previously reported. Our three probands, aged 28-40 years, harbored de novo nonsense variants in IRF2BPL (c.370C > T, p.[Gln124*] and c.364C > T; p.[Gln122*], respectively). From late childhood/adolescence, they presented with severe myoclonus epilepsy, stimulus-sensitive myoclonus, and progressive cognitive, speech, and cerebellar impairment, consistent with a typical PME syndrome. The skin biopsy revealed massive intracellular glycogen inclusions in one proband, suggesting a similar pathogenic pathway to other storage disorders. Whereas the two older probands were severely affected, the younger proband had a milder PME phenotype, partially overlapping with some of the previously reported IRF2BPL cases, suggesting that some of them might be unrecognized PME. Interestingly, all three patients harbored protein-truncating variants clustered in a proximal, highly conserved gene region around the "coiled-coil" domain. Our data show that PME can be an additional phenotype within the spectrum of IRF2BPL-related disorders and suggest IRF2BPL as a novel causative gene for PME.


Assuntos
Epilepsias Mioclônicas , Epilepsia , Epilepsias Mioclônicas Progressivas , Mioclonia , Humanos , Criança , Mutação , Epilepsias Mioclônicas Progressivas/genética , Epilepsias Mioclônicas/patologia , Família , Proteínas de Transporte/genética , Proteínas Nucleares/genética
3.
Neuromolecular Med ; 25(2): 145-162, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36153432

RESUMO

The attribution of seizure freedom is yet to be achieved for patients suffering from refractory epilepsy, e.g. Dravet Syndrome (DS). The confined ability of mono-chemical entity-based antiseizure drugs (ASDs) to act directly at genomic level is one of the factors, combined with undetermined seizure triggers lead to recurrent seizure (RS) in DS, abominably affecting the sub-genomic architecture of neural cells. Thus, the RS and ASD appear to be responsible for the spectrum of exorbitant clinical pathology. The RS distresses the 5-HT-serotonin pathway, hypomethylates genes of CNS, and modulates the microRNA (miRNA)/long non-coding RNA (lncRNA), eventually leading to frozen molecular alterations. These changes shall be reverted by compatible epigenetic regulators (EGR) like, miRNA and lncRNA from Breast milk (BML) and Bacopa monnieri (BMI). The absence of studious seizure in SCN1A mutation-positive babies for the first 6 months raises the possibility that the consequences of mutation in SCN1A are subsidized by EGRs from BML. EGR-dependent-modifier gene effect is likely imposed by the other members of the SCN family. Therefore, we advocate that miRNA/lncRNA from BML and bacosides/miRNA from BMI buffer the effect of SCN1A mutation by sustainably maintaining modifier gene effect in the aberrant neurons. The presence of miRNA-155-5p, -30b-5p, and -30c-5p family in BML and miR857, miR168, miR156, and miR158 in BMI target at regulating SCN family and CLCN5 as visualized by Cystoscope. Thus, we envisage that the possible effects of EGR might include (a) upregulating the haploinsufficient SCN1A strand, (b) down-regulating seizure-elevated miRNA, (c) suppressing the seizure-induced methyltransferases, and (d) enhancing the GluN2A subunit of NMDA receptor to improve cognition. The potential of these EGRs from BML and BML is to further experimentally strengthen, long-haul step forward in molecular therapeutics.


Assuntos
Epilepsia Resistente a Medicamentos , Epilepsias Mioclônicas , MicroRNAs , RNA Longo não Codificante , Lactente , Feminino , Humanos , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Epilepsia Resistente a Medicamentos/genética , RNA Longo não Codificante/genética , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Convulsões , Mutação , MicroRNAs/genética , Epigênese Genética
4.
Turk Patoloji Derg ; 37(3): 264-265, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33432562

RESUMO

Lafora disease is a severe form of progressive myoclonic epilepsy with autosomal recessive inheritance diagnosed by inclusion body in biopsy. A 26-year-old woman was admitted due to complaints of frequent twitches and fainting. The 0.5x0.3x0.3 cm axillary skin punch biopsy was subjected to routine histopathological evaluation. Cytoplasmic PAS-positive inclusion bodies were observed at the basal side of the eccrine and apocrine glands. The diagnosis of Lafora disease can also be made by the observation of the polyglycosan cytoplasmic inclusion bodies in the brain, liver and skeletal muscle biopsies. Although we need more work to understand the etiopathogenesis of Lafora disease, we would like to draw attention to the importance of skin biopsy in the differential diagnosis of young patients with clinically refractory epilepsy, myoclonus, and cognitive decline.


Assuntos
Epilepsias Mioclônicas/diagnóstico , Doença de Lafora/diagnóstico , Pele/patologia , Adulto , Biópsia , Epilepsias Mioclônicas/patologia , Feminino , Humanos , Doença de Lafora/genética , Microscopia Eletrônica
5.
Dev Med Child Neurol ; 62(11): 1331-1335, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32538476

RESUMO

Variants in the gene SCN1A are a common genetic cause for a wide range of epilepsy phenotypes ranging from febrile seizures to Dravet syndrome. Focal onset seizures and structural lesions can be present in these patients and the question arises whether epilepsy surgery should be considered. We report eight patients (mean age 13y 11mo [SD 8y 1mo], range 3-26y; four females, four males) with SCN1A variants, who underwent epilepsy surgery. Outcomes were variable and seemed to be directly related to the patient's anatomo-electroclinical epilepsy phenotype. Patients with Dravet syndrome had unfavourable outcomes, whilst patients with focal epilepsy, proven to arise from a single structural lesion, had good results. We conclude that the value of epilepsy surgery in patients with an SCN1A variant rests on two issues: understanding whether the variant is pathogenic and the patient's anatomo-electroclinical phenotype. Careful evaluation of epilepsy phenotype integrated with understanding the significance of genetic variants is essential in determining a patient's suitability for epilepsy surgery. Patients with focal onset epilepsy may benefit from epilepsy surgery, whereas those with Dravet syndrome do not. WHAT THIS PAPER ADDS: Patients should not automatically be excluded from epilepsy surgery evaluation if they carry an SCN1A variant. Patients with focal epilepsy may benefit from epilepsy surgery; those with Dravet syndrome do not.


Assuntos
Epilepsias Mioclônicas/cirurgia , Epilepsias Parciais/cirurgia , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Adolescente , Adulto , Criança , Pré-Escolar , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Epilepsias Mioclônicas/fisiopatologia , Epilepsias Parciais/genética , Epilepsias Parciais/patologia , Epilepsias Parciais/fisiopatologia , Feminino , Humanos , Masculino , Avaliação de Resultados em Cuidados de Saúde , Adulto Jovem
6.
Clin Genet ; 97(4): 556-566, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31957011

RESUMO

NGLY1 encodes the enzyme N-glycanase that is involved in the degradation of glycoproteins as part of the endoplasmatic reticulum-associated degradation pathway. Variants in this gene have been described to cause a multisystem disease characterized by neuromotor impairment, neuropathy, intellectual disability, and dysmorphic features. Here, we describe four patients with pathogenic variants in NGLY1. As the clinical features and laboratory results of the patients suggested a multisystem mitochondrial disease, a muscle biopsy had been performed. Biochemical analysis in muscle showed a strongly reduced ATP production rate in all patients, while individual OXPHOS enzyme activities varied from normal to reduced. No causative variants in any mitochondrial disease genes were found using mtDNA analysis and whole exome sequencing. In all four patients, variants in NGLY1 were identified, including two unreported variants (c.849T>G (p.(Cys283Trp)) and c.1067A>G (p.(Glu356Gly)). Western blot analysis of N-glycanase in muscle and fibroblasts showed a complete absence of N-glycanase. One patient showed a decreased basal and maximal oxygen consumption rates in fibroblasts. Mitochondrial morphofunction fibroblast analysis showed patient specific differences when compared to control cell lines. In conclusion, variants in NGLY1 affect mitochondrial energy metabolism which in turn might contribute to the clinical disease course.


Assuntos
Epilepsias Mioclônicas/genética , Deficiência Intelectual/genética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética , Polineuropatias/genética , Criança , Pré-Escolar , Defeitos Congênitos da Glicosilação/diagnóstico por imagem , Defeitos Congênitos da Glicosilação/genética , Defeitos Congênitos da Glicosilação/metabolismo , Defeitos Congênitos da Glicosilação/patologia , Epilepsias Mioclônicas/diagnóstico por imagem , Epilepsias Mioclônicas/patologia , Feminino , Humanos , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/patologia , Masculino , Mitocôndrias/genética , Mitocôndrias/patologia , Mutação/genética , Polineuropatias/diagnóstico por imagem , Polineuropatias/patologia
7.
J Clin Invest ; 129(6): 2571-2577, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30946030

RESUMO

Increased urinary oxalate excretion (hyperoxaluria) promotes the formation of calcium oxalate crystals. Monogenic diseases due to hepatic enzymes deficiency result in chronic hyperoxaluria, promoting end-stage renal disease in children and young adults. Ethylene glycol poisoning also results in hyperoxaluria promoting acute renal failure and frequently death. Stiripentol is an antiepileptic drug used to treat children affected by Dravet syndrome, possibly by inhibiting neuronal lactate dehydrogenase 5 isoenzyme. As this isoenzyme is also the last step of hepatic oxalate production, we hypothesized that Stiripentol would potentially reduce hepatic oxalate production and urine oxalate excretion. In vitro, Stiripentol decreased in a dose-dependent manner the synthesis of oxalate by hepatocytes. In vivo, Stiripentol oral administration reduced significantly urine oxalate excretion in rats. Stiripentol protected kidneys against calcium oxalate crystal deposits in acute ethylene glycol intoxication and chronic calcium oxalate nephropathy models. In both models, Stiripentol improved significantly renal function. Patients affected by Dravet syndrome and treated with Stiripentol had a lower urine oxalate excretion than control patients. A young girl affected by severe type I hyperoxaluria received Stiripentol for several weeks: urine oxalate excretion decreased by two-thirds. Stiripentol is a promising potential therapy against genetic hyperoxaluria and ethylene glycol poisoning.


Assuntos
Dioxolanos/farmacologia , Etilenoglicol/intoxicação , Hiperoxalúria Primária/prevenção & controle , Nefrolitíase/prevenção & controle , Animais , Oxalato de Cálcio/metabolismo , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Feminino , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Hiperoxalúria Primária/metabolismo , Hiperoxalúria Primária/patologia , Rim/metabolismo , Rim/patologia , Masculino , Nefrolitíase/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Eur J Med Genet ; 62(12): 103591, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30500434

RESUMO

BACKGROUND: Mutations in the KCTD7 gene have been associated with progressive myoclonus epilepsy and, in a single patient, with the so-called "Neuronal Ceroid Lipofuscinosis 14" (characterised by myoclonic seizures, cognitive regression, optic atrophy leading to visual loss, and progressive cortical and cerebellar atrophy). CLINICAL REPORTS: We describe two new patients carrying two novel pathogenic mutations in the KCTD7 gene. Patient 1 (NM_153033.4: c.[533C > T], NP_694578: p.[(Ala178Val)]) was a 17-year-old girl who presented with early-onset epilepsy resembling epilepsia partialis continua (responsive to intravenous corticosteroids and immunoglobulins), and later developed myoclonic seizures and atypical absences, photosensitivity to very low frequencies and progressive seizures-related neurocognitive and motor deterioration. Patient 2 (NM_153033.4: c.[172G>A], NP_694578: p.[(Gly58Arg)]) presented with early neurological regression, myoclonic seizures and lysosomal storage material which was consistent with a neuronal ceroid lipofuscinosis (NCL) at skin biopsy. Both patients had non epileptic myoclonus. CONCLUSIONS: The two reported patients carrying novel pathogenic variants in KCTD7 gene presented with a remarkable phenotypic heterogeneity including: a) progressive myoclonus epilepsy without NCL-type lysosomal storages; b) progressive myoclonus epilepsy with lysosomal storages resembling NCL pattern (NCL14); c) progressive myoclonus epilepsy with epilepsia partialis continua.


Assuntos
Epilepsias Mioclônicas/genética , Doenças por Armazenamento dos Lisossomos/genética , Lipofuscinoses Ceroides Neuronais/genética , Fenótipo , Canais de Potássio/genética , Adolescente , Criança , Eletroencefalografia , Epilepsias Mioclônicas/diagnóstico por imagem , Epilepsias Mioclônicas/patologia , Feminino , Humanos , Doenças por Armazenamento dos Lisossomos/diagnóstico por imagem , Doenças por Armazenamento dos Lisossomos/patologia , Lipofuscinoses Ceroides Neuronais/diagnóstico por imagem , Lipofuscinoses Ceroides Neuronais/patologia , Pele/metabolismo , Pele/patologia , Síndrome
9.
Methods Mol Biol ; 1826: 109-121, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30194596

RESUMO

Our current knowledge about the cellular mechanisms underlying serpin-related disorders, the serpinopathies, is predominantly based on studies in cell culture models of disease, particularly for alpha-1 antitrypsin (AAT, SERPINA1) deficiency causing emphysema and the familial encephalopathy with neuroserpin (NS, SERPINI1) inclusion bodies (FENIB). FENIB, a neurodegenerative dementia, is caused by polymerization of NS (Miranda and Lomas, Cell Mol Life Sci 63:709-722, 2006; Roussel BD et al., Epileptic Disor 18:103-110, 2016), while AAT deficiency presents as a result of several divergent mutations in the AAT gene that cause lack of protein synthesis or complete intracellular degradation (null variants) or polymer formation (polymerogenic variants) (Lomas et al., J Hepatol 65:413-424, 2016; Greene et al., Nat Rev Dis Primers 2:16051, 2016; Ferrarotti et al. Orphanet J Rare D 9:172, 2014). Both diseases have been extensively modeled in cell culture systems by expressing mutant variants in a variety of ways. Here we describe the methodologies we follow in our cell model systems used to examine serpin disorders.


Assuntos
Enfisema , Epilepsias Mioclônicas , Transtornos Heredodegenerativos do Sistema Nervoso , Modelos Biológicos , Mutação , Neuropeptídeos , Serpinas , alfa 1-Antitripsina , Animais , Células COS , Chlorocebus aethiops , Enfisema/genética , Enfisema/metabolismo , Enfisema/patologia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Células HEK293 , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Humanos , Camundongos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Células PC12 , Ratos , Serpinas/genética , Serpinas/metabolismo , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo , Neuroserpina
10.
Nat Genet ; 50(4): 581-590, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29507423

RESUMO

Epilepsy is a common neurological disorder, and mutations in genes encoding ion channels or neurotransmitter receptors are frequent causes of monogenic forms of epilepsy. Here we show that abnormal expansions of TTTCA and TTTTA repeats in intron 4 of SAMD12 cause benign adult familial myoclonic epilepsy (BAFME). Single-molecule, real-time sequencing of BAC clones and nanopore sequencing of genomic DNA identified two repeat configurations in SAMD12. Intriguingly, in two families with a clinical diagnosis of BAFME in which no repeat expansions in SAMD12 were observed, we identified similar expansions of TTTCA and TTTTA repeats in introns of TNRC6A and RAPGEF2, indicating that expansions of the same repeat motifs are involved in the pathogenesis of BAFME regardless of the genes in which the expanded repeats are located. This discovery that expansions of noncoding repeats lead to neuronal dysfunction responsible for myoclonic tremor and epilepsy extends the understanding of diseases with such repeat expansion.


Assuntos
Expansão das Repetições de DNA , Epilepsias Mioclônicas/genética , Repetições de Microssatélites , Proteínas do Tecido Nervoso/genética , Motivo Estéril alfa/genética , Adulto , Idade de Início , Autoantígenos/genética , Sequência de Bases , Epilepsias Mioclônicas/etiologia , Epilepsias Mioclônicas/patologia , Feminino , Instabilidade Genômica , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Íntrons , Masculino , Linhagem , Células de Purkinje/patologia , Proteínas de Ligação a RNA/genética , Análise de Sequência de DNA
11.
Proc Natl Acad Sci U S A ; 114(10): 2669-2674, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28223533

RESUMO

The clinical and electroencephalographic features of a canine generalized myoclonic epilepsy with photosensitivity and onset in young Rhodesian Ridgeback dogs (6 wk to 18 mo) are described. A fully penetrant recessive 4-bp deletion was identified in the DIRAS family GTPase 1 (DIRAS1) gene with an altered expression pattern of DIRAS1 protein in the affected brain. This neuronal DIRAS1 gene with a proposed role in cholinergic transmission provides not only a candidate for human myoclonic epilepsy but also insights into the disease etiology, while establishing a spontaneous model for future intervention studies and functional characterization.


Assuntos
Epilepsias Mioclônicas/genética , GTP Fosfo-Hidrolases/genética , Deleção de Genes , Transtornos de Fotossensibilidade/genética , Proteínas Supressoras de Tumor/genética , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Cães , Epilepsias Mioclônicas/patologia , Humanos , Transtornos de Fotossensibilidade/patologia
12.
Nucleic Acids Res ; 45(10): e76, 2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28132024

RESUMO

Genomic mosaicism arising from postzygotic mutations has long been associated with cancer and more recently with non-cancer diseases. It has also been detected in healthy individuals including healthy parents of children affected with genetic disorders, highlighting its critical role in the origin of genetic mutations. However, most existing software for the genome-wide identification of single-nucleotide mosaicisms (SNMs) requires a paired control tissue obtained from the same individual which is often unavailable for non-cancer individuals and sometimes missing in cancer studies. Here, we present MosaicHunter (http://mosaichunter.cbi.pku.edu.cn), a bioinformatics tool that can identify SNMs in whole-genome and whole-exome sequencing data of unpaired samples without matched controls using Bayesian genotypers. We evaluate the accuracy of MosaicHunter on both simulated and real data and demonstrate that it has improved performance compared with other somatic mutation callers. We further demonstrate that incorporating sequencing data of the parents can be an effective approach to significantly improve the accuracy of detecting SNMs in an individual when a matched control sample is unavailable. Finally, MosaicHunter also has a paired mode that can take advantage of matched control samples when available, making it a useful tool for detecting SNMs in both non-cancer and cancer studies.


Assuntos
Algoritmos , Neoplasias da Mama/genética , Epilepsias Mioclônicas/genética , Mosaicismo , Polimorfismo de Nucleotídeo Único , Adulto , Teorema de Bayes , Benchmarking , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Criança , Conjuntos de Dados como Assunto , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Exoma , Feminino , Genótipo , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Padrões de Herança , Masculino , Análise de Sequência de DNA , Software
13.
J Child Neurol ; 31(9): 1127-37, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27095821

RESUMO

Mutations in mitochondrial aminoacyl-tRNA synthetases are an increasingly recognized cause of human diseases, often arising in individuals with compound heterozygous mutations and presenting with system-specific phenotypes, frequently neurologic. FARS2 encodes mitochondrial phenylalanyl transfer ribonucleic acid (RNA) synthetase (mtPheRS), perturbations of which have been reported in 6 cases of an infantile, lethal disease with refractory epilepsy and progressive myoclonus. Here the authors report the case of juvenile onset refractory epilepsy and progressive myoclonus with compound heterozygous FARS2 mutations. The authors describe the clinical course over 6 years of care at their institution and diagnostic studies including electroencephalogram (EEG), brain magnetic resonance imaging (MRI), serum and cerebrospinal fluid analyses, skeletal muscle biopsy histology, and autopsy gross and histologic findings, which include features shared with Alpers-Huttenlocher syndrome, Leigh syndrome, and a previously published case of FARS2 mutation associated infantile onset disease. The authors also present structure-guided analysis of the relevant mutations based on published mitochondrial phenylalanyl transfer RNA synthetase and related protein crystal structures as well as biochemical analysis of the corresponding recombinant mutant proteins.


Assuntos
Epilepsia Resistente a Medicamentos/genética , Epilepsias Mioclônicas/genética , Heterozigoto , Proteínas Mitocondriais/genética , Mutação , Fenilalanina-tRNA Ligase/genética , Adolescente , Epilepsia Resistente a Medicamentos/diagnóstico por imagem , Epilepsia Resistente a Medicamentos/patologia , Epilepsia Resistente a Medicamentos/fisiopatologia , Epilepsias Mioclônicas/diagnóstico por imagem , Epilepsias Mioclônicas/patologia , Epilepsias Mioclônicas/fisiopatologia , Evolução Fatal , Feminino , Humanos , Fenótipo
15.
Am J Med Genet A ; 167A(10): 2231-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26079862

RESUMO

De novo mutations (DNM) in SYNGAP1, encoding Ras/Rap GTPase-activating protein SynGAP, have been reported in individuals with nonsyndromic intellectual disability (ID). We identified 10 previously unreported individuals with SYNGAP1 DNM; seven via the Deciphering Developmental Disorders (DDD) Study, one through clinical analysis for copy number variation and the remaining two (monozygotic twins) via a research multi-gene panel analysis. Seven of the nine heterozygous mutations are likely to result in loss-of-function (3 nonsense; 3 frameshift; 1 whole gene deletion). The remaining two mutations, one of which affected the monozygotic twins, were missense variants. Each individual carrying a DNM in SYNGAP1 had moderate-to-severe ID and 7/10 had epilepsy; typically myoclonic seizures, absences or drop attacks. 8/10 had hypotonia, 5/10 had significant constipation, 7/10 had wide-based/unsteady gait, 3/10 had strabismus, and 2/10 had significant hip dysplasia. A proportion of the affected individuals had a similar, myopathic facial appearance, with broad nasal bridge, relatively long nose and full lower lip vermilion. A distinctive behavioral phenotype was also observed with aggressive/challenging behavior and significant sleep problems being common. 7/10 individuals had MR imaging of the brain each of which was reported as normal. The clinical features of the individuals reported here show significant overlap with those associated with 6p21.3 microdeletions, confirming that haploinsufficiency for SYNGAP1 is responsible for both disorders. © 2015 Wiley Periodicals, Inc.


Assuntos
Heterozigoto , Deficiência Intelectual/genética , Mutação , Proteínas Ativadoras de ras GTPase/genética , Adolescente , Criança , Pré-Escolar , Constipação Intestinal/diagnóstico , Constipação Intestinal/genética , Constipação Intestinal/patologia , Análise Mutacional de DNA , Epilepsias Mioclônicas/diagnóstico , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Feminino , Transtornos Neurológicos da Marcha/diagnóstico , Transtornos Neurológicos da Marcha/genética , Transtornos Neurológicos da Marcha/patologia , Expressão Gênica , Haploinsuficiência , Luxação do Quadril/diagnóstico , Luxação do Quadril/genética , Luxação do Quadril/patologia , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/patologia , Masculino , Hipotonia Muscular/diagnóstico , Hipotonia Muscular/genética , Hipotonia Muscular/patologia , Fenótipo , Estrabismo/diagnóstico , Estrabismo/genética , Estrabismo/patologia , Gêmeos Monozigóticos
16.
Cell Res ; 24(11): 1311-27, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25312340

RESUMO

Postzygotic single-nucleotide mutations (pSNMs) have been studied in cancer and a few other overgrowth human disorders at whole-genome scale and found to play critical roles. However, in clinically unremarkable individuals, pSNMs have never been identified at whole-genome scale largely due to technical difficulties and lack of matched control tissue samples, and thus the genome-wide characteristics of pSNMs remain unknown. We developed a new Bayesian-based mosaic genotyper and a series of effective error filters, using which we were able to identify 17 SNM sites from ~80× whole-genome sequencing of peripheral blood DNAs from three clinically unremarkable adults. The pSNMs were thoroughly validated using pyrosequencing, Sanger sequencing of individual cloned fragments, and multiplex ligation-dependent probe amplification. The mutant allele fraction ranged from 5%-31%. We found that C→T and C→A were the predominant types of postzygotic mutations, similar to the somatic mutation profile in tumor tissues. Simulation data showed that the overall mutation rate was an order of magnitude lower than that in cancer. We detected varied allele fractions of the pSNMs among multiple samples obtained from the same individuals, including blood, saliva, hair follicle, buccal mucosa, urine, and semen samples, indicating that pSNMs could affect multiple sources of somatic cells as well as germ cells. Two of the adults have children who were diagnosed with Dravet syndrome. We identified two non-synonymous pSNMs in SCN1A, a causal gene for Dravet syndrome, from these two unrelated adults and found that the mutant alleles were transmitted to their children, highlighting the clinical importance of detecting pSNMs in genetic counseling.


Assuntos
Genoma Humano , Polimorfismo de Nucleotídeo Único , Zigoto/metabolismo , Acetil-CoA Carboxilase/genética , Adulto , Alelos , Teorema de Bayes , Pré-Escolar , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Feminino , Aconselhamento Genético , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mosaicismo , Linhagem , Análise de Sequência de DNA
17.
Neurosciences (Riyadh) ; 19(4): 312-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25274592

RESUMO

Folinic acid-responsive seizures (FARS) are a rare treatable cause of neonatal epilepsy. They have characteristic peaks on CSF monoamine metabolite analysis, and have mutations in the ALDH7A1 gene, characteristically found in pyridoxine-dependent epilepsy. There are case reports of patients presenting with seizures at a later age, and with folate deficiency due to different mechanisms with variable response to folinic acid supplementation. Here, we report 2 siblings who presented with global developmental delay and intractable seizures who responded clinically to folinic acid therapy. Their work-up included metabolic and genetic testing. The DNA sequencing was carried out for the ALDH7A1 gene, and the folate receptor 1 (FOLR1) gene. They had very low 5-methyltetrahydrofolate (5-MTHF) in CSF with no systemic folate deficiency and no characteristic peaks on neurotransmitter metabolite chromatogram. A novel mutation in the FOLR1 gene was found. The mutation in this gene is shown to affect CSF folate transport leading to cerebral folate deficiency. The response to treatment with folinic acid was dramatic with improvement in social interaction, mobility, and complete seizure control. We should consider the possibility of this treatable condition in appropriate clinical circumstances early, as diagnosis with favorable outcome depends on the specialized tests.


Assuntos
Encefalopatias Metabólicas Congênitas/tratamento farmacológico , Epilepsias Mioclônicas/tratamento farmacológico , Receptor 1 de Folato/genética , Deficiência de Ácido Fólico/tratamento farmacológico , Leucovorina/uso terapêutico , Mutação de Sentido Incorreto , Mutação Puntual , Atrofia , Encéfalo/patologia , Encefalopatias Metabólicas Congênitas/líquido cefalorraquidiano , Encefalopatias Metabólicas Congênitas/diagnóstico , Encefalopatias Metabólicas Congênitas/genética , Encefalopatias Metabólicas Congênitas/patologia , Transtornos Globais do Desenvolvimento Infantil/genética , Pré-Escolar , Consanguinidade , Deficiências do Desenvolvimento/genética , Diagnóstico Precoce , Eletroencefalografia , Epilepsias Mioclônicas/líquido cefalorraquidiano , Epilepsias Mioclônicas/diagnóstico , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Feminino , Receptor 1 de Folato/deficiência , Deficiência de Ácido Fólico/líquido cefalorraquidiano , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Piridoxina/uso terapêutico , Irmãos , Tetra-Hidrofolatos/líquido cefalorraquidiano
18.
Eur J Paediatr Neurol ; 17(6): 625-30, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23816342

RESUMO

BACKGROUND: Primary coenzyme Q10 (CoQ10) deficiencies are heterogeneous autosomal recessive disorders. CoQ2 mutations have been identified only rarely in patients. All affected individuals presented with nephrotic syndrome in the first year of life. METHODS: An infant is studied with myoclonic seizures and hypertrophic cardiomyopathy in the first months of life and developed a nephrotic syndrome in a later stage. RESULTS: At three weeks of age, the index patient developed myoclonic seizures. In addition, he had hypertrophic cardiomyopathy and increased CSF lactate. A skeletal muscle biopsy performed at two months of age disclosed normal activities of the oxidative phosphorylation complexes. The child was supplemented with CoQ10 (5 mg/kg/day). At the age of four months, brain MR images showed bilateral increased signal intensities in putamen and cerebral cortex. After that age, he developed massive proteinuria. The daily dose of CoQ10 was increased to 30 mg/kg. Renal biopsy showed focal segmental glomerulosclerosis. Biochemical analyses of a kidney biopsy sample revealed a severely decreased activity of succinate cytochrome c reductase [complex II + III] suggesting ubiquinone depletion. Incorporation of labelled precursors necessary for CoQ10 synthesis was significantly decreased in cultured skin fibroblasts. His condition deteriorated and he died at the age of five months. A novel homozygous mutation c.326G > A (p.Ser109Asn) was found in COQ2. CONCLUSIONS: In contrast to previously reported patients with CoQ2 the proband presented with early myoclonic epilepsy, hypertrophic cardiomyopathy and only in a later stage developed a nephrotic syndrome. The phenotype of this patient enlarges the phenotypical spectrum of the multisystem infantile variant.


Assuntos
Alquil e Aril Transferases/genética , Ataxia/genética , Cardiomiopatia Hipertrófica/genética , Epilepsias Mioclônicas/genética , Doenças Mitocondriais/genética , Debilidade Muscular/genética , Mutação/genética , Síndrome Nefrótica/genética , Ubiquinona/deficiência , Ataxia/complicações , Ataxia/patologia , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/patologia , Imagem de Difusão por Ressonância Magnética , Eletroencefalografia , Epilepsias Mioclônicas/complicações , Epilepsias Mioclônicas/patologia , Testes Genéticos , Humanos , Lactente , Rim/patologia , Rim/ultraestrutura , Espectroscopia de Ressonância Magnética , Masculino , Microscopia Eletrônica de Transmissão , Doenças Mitocondriais/complicações , Doenças Mitocondriais/patologia , Debilidade Muscular/complicações , Debilidade Muscular/patologia , Músculo Esquelético/patologia , Síndrome Nefrótica/etiologia , Síndrome Nefrótica/patologia , Ubiquinona/genética
19.
J Biol Chem ; 287(50): 42001-8, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23086956

RESUMO

Mutations in SCN1A, encoding the voltage-gated sodium channel Na(V)1.1, are the most common cause of severe myoclonic epilepsy of infancy (SMEI) or Dravet syndrome. SMEI is most often associated with premature truncations of Na(V)1.1 that cause loss of function, but nontruncating mutations also occur. We hypothesized that some nontruncating mutations might impair trafficking of Na(V)1.1 to the plasma membrane. Here we demonstrated that seven nontruncating missense or in-frame deletion mutations (L986F, delF1289, R1648C, F1661S, G1674R, and G1979E) exhibited reduced cell surface expression relative to wild type (WT) Na(V)1.1 consistent with impaired trafficking. We tested whether two commonly prescribed antiepileptic drugs (phenytoin, lamotrigine), as well as the cystic fibrosis transmembrane conductance regulator (CFTR) trafficking corrector VRT-325, could rescue cell surface and functional expression of two representative Na(V)1.1 mutants (R1648C, G1674R). Treatment of cells with phenytoin increased cell surface expression of WT-Na(V)1.1 and both mutant channels, whereas lamotrigine only increased surface expression of R1648C. VRT-325 did not alter surface expression of WT-Na(V)1.1 or mutant channels. Although phenytoin increased surface expression of G1674R, channel function was not restored, suggesting that this mutation also causes an intrinsic loss of function. Both phenytoin and lamotrigine increased functional expression of R1648C, but lamotrigine also increased persistent sodium current evoked by this mutation. Our findings indicate that certain nontruncating SCN1A mutations associated with SMEI have impaired cell surface expression and that some alleles may be amenable to pharmacological rescue of this defect. However, rescue of dysfunctional Na(V)1.1 channels to the plasma membrane could contribute to exacerbating rather than ameliorating the disease.


Assuntos
Sequência de Aminoácidos , Membrana Celular/metabolismo , Epilepsias Mioclônicas , Regulação da Expressão Gênica/genética , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.1 , Deleção de Sequência , Alelos , Substituição de Aminoácidos , Anticonvulsivantes/farmacologia , Membrana Celular/genética , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/metabolismo , Epilepsias Mioclônicas/patologia , Células HEK293 , Humanos , Lamotrigina , Canal de Sódio Disparado por Voltagem NAV1.1/biossíntese , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Fenitoína/farmacologia , Piperazinas/farmacologia , Quinazolinas/farmacologia , Triazinas/farmacologia
20.
Methods Enzymol ; 501: 421-66, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22078544

RESUMO

The serpinopathies result from point mutations in members of the serine protease inhibitor or serpin superfamily. They are characterized by the formation of ordered polymers that are retained within the cell of synthesis. This causes disease by a "toxic gain of function" from the accumulated protein and a "loss of function" as a result of the deficiency of inhibitors that control important proteolytic cascades. The serpinopathies are exemplified by the Z (Glu342Lys) mutant of α1-antitrypsin that results in the retention of ordered polymers within the endoplasmic reticulum of hepatocytes. These polymers form the intracellular inclusions that are associated with neonatal hepatitis, cirrhosis, and hepatocellular carcinoma. A second example results from mutations in the neurone-specific serpin-neuroserpin to form ordered polymers that are retained as inclusions within subcortical neurones as Collins' bodies. These inclusions underlie the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. There are different pathways to polymer formation in vitro but not all form polymers that are relevant in vivo. It is therefore essential that protein-based structural studies are interpreted in the context of human samples and cell and animal models of disease. We describe here the biochemical techniques, monoclonal antibodies, cell biology, animal models, and stem cell technology that are useful to characterize the serpin polymers that form in vivo.


Assuntos
Biofísica/métodos , Epilepsias Mioclônicas/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Processamento de Imagem Assistida por Computador/métodos , Pulmão/metabolismo , Neuropeptídeos/metabolismo , Mutação Puntual , Serpinas/metabolismo , alfa 1-Antitripsina/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Neuropeptídeos/química , Neuropeptídeos/genética , Neutrófilos/citologia , Neutrófilos/metabolismo , Fragmentos de Peptídeos , Polimerização , Ligação Proteica , Conformação Proteica , Proteólise , Serpinas/química , Serpinas/genética , Transfecção , alfa 1-Antitripsina/química , alfa 1-Antitripsina/genética , Neuroserpina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA