Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Eur J Hum Genet ; 2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973392

RESUMEN

Autism spectrum disorder (ASD) is caused by combined genetic and environmental factors. Genetic heritability in ASD is estimated as 60-90%, and genetic investigations have revealed many monogenic factors. We analyzed 405 patients with ASD using family-based exome sequencing to detect disease-causing single-nucleotide variants (SNVs), small insertions and deletions (indels), and copy number variations (CNVs) for molecular diagnoses. All candidate variants were validated by Sanger sequencing or quantitative polymerase chain reaction and were evaluated using the American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines for molecular diagnosis. We identified 55 disease-causing SNVs/indels in 53 affected individuals and 13 disease-causing CNVs in 13 affected individuals, achieving a molecular diagnosis in 66 of 405 affected individuals (16.3%). Among the 55 disease-causing SNVs/indels, 51 occurred de novo, 2 were compound heterozygous (in one patient), and 2 were X-linked hemizygous variants inherited from unaffected mothers. The molecular diagnosis rate in females was significantly higher than that in males. We analyzed affected sibling cases of 24 quads and 2 quintets, but only one pair of siblings shared an identical pathogenic variant. Notably, there was a higher molecular diagnostic rate in simplex cases than in multiplex families. Our simulation indicated that the diagnostic yield is increasing by 0.63% (range 0-2.5%) per year. Based on our simple simulation, diagnostic yield is improving over time. Thus, periodical reevaluation of ES data should be strongly encouraged in undiagnosed ASD patients.

2.
Mol Brain ; 14(1): 59, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33757575

RESUMEN

Intracellular aggregates are a common pathological hallmark of neurodegenerative diseases such as polyglutamine (polyQ) diseases, amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and multiple system atrophy (MSA). Aggregates are mainly formed by aberrant disease-specific proteins and are accompanied by accumulation of other aggregate-interacting proteins. Although aggregate-interacting proteins have been considered to modulate the formation of aggregates and to be involved in molecular mechanisms of disease progression, the components of aggregate-interacting proteins remain unknown. In this study, we showed that small glutamine-rich tetratricopeptide repeat-containing protein alfa (SGTA) is an aggregate-interacting protein in neurodegenerative diseases. Immunohistochemistry showed that SGTA interacted with intracellular aggregates in Huntington disease (HD) cell models and neurons of HD model mice. We also revealed that SGTA colocalized with intracellular aggregates in postmortem brains of patients with polyQ diseases including spinocerebellar ataxia (SCA)1, SCA2, SCA3, and dentatorubral-pallidoluysian atrophy. In addition, SGTA colocalized with glial cytoplasmic inclusions in the brains of MSA patients, whereas no accumulation of SGTA was observed in neurons of PD and ALS patients. In vitro study showed that SGTA bound to polyQ aggregates through its C-terminal domain and SGTA overexpression reduced intracellular aggregates. These results suggest that SGTA may play a role in the formation of aggregates and may act as potential modifier of molecular pathological mechanisms of polyQ diseases and MSA.


Asunto(s)
Química Encefálica , Chaperonas Moleculares/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Péptidos/metabolismo , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Animales , Autopsia , Encéfalo/patología , Línea Celular Tumoral , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Cuerpos de Inclusión/química , Ratones , Ratones Transgénicos , Neuroblastoma , Enfermedades Neurodegenerativas/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes/metabolismo , Solubilidad , Fracciones Subcelulares/metabolismo , Transfección , alfa-Sinucleína/análisis
3.
Front Immunol ; 12: 625465, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33659007

RESUMEN

Here we report three cases of anti-myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) mimicking multiple sclerosis in which seropositivity for anti-MOG antibodies occurred during disease-modifying drug dimethyl fumarate (DMF) treatment. These patients developed relapses with anti-MOG antibody seroconversion after switching from fingolimod or steroid pulse therapy to DMF, which was associated with peripheral lymphocyte recovery. MOGAD is considered a humoral immune disease, and DMF reportedly enhances Th2-skewed humoral immune activity. Therefore, we suggest that DMF, but not fingolimod, may exacerbate humoral immune imbalance and enhance autoantibody production, leading to aggravation of MOGAD.


Asunto(s)
Autoanticuerpos/sangre , Dimetilfumarato/uso terapéutico , Inmunosupresores/uso terapéutico , Glicoproteína Mielina-Oligodendrócito/inmunología , Mielitis Transversa/tratamiento farmacológico , Neuritis Óptica/tratamiento farmacológico , Adulto , Anciano , Biomarcadores/sangre , Dimetilfumarato/efectos adversos , Sustitución de Medicamentos , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunosupresores/efectos adversos , Masculino , Mielitis Transversa/diagnóstico , Mielitis Transversa/inmunología , Neuritis Óptica/diagnóstico , Neuritis Óptica/inmunología , Recurrencia , Seroconversión , Esteroides/uso terapéutico , Resultado del Tratamiento
5.
Ann Neurol ; 86(6): 962-968, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31433517

RESUMEN

Leukoencephalopathies comprise a broad spectrum of disorders, but the genetic background of adult leukoencephalopathies has rarely been assessed. In this study, we analyzed 101 Japanese patients with genetically unresolved adult leukoencephalopathy using whole-exome sequencing and repeat-primed polymerase chain reaction for detecting GGC expansion in NOTCH2NLC. NOTCH2NLC was recently identified as the cause of neuronal intranuclear inclusion disease. We found 12 patients with GGC expansion in NOTCH2NLC as the most frequent cause of adult leukoencephalopathy followed by NOTCH3 variants in our cohort. Furthermore, we found 1 case with de novo GGC expansion, which might explain the underlying pathogenesis of sporadic cases. ANN NEUROL 2019;86:962-968.


Asunto(s)
Variación Genética/genética , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/genética , Receptor Notch2/genética , Expansión de Repetición de Trinucleótido/genética , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Front Neurol ; 10: 763, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379717

RESUMEN

Fingolimod acts as a functional antagonist of the sphingosine-1-phosphate receptor and is widely used for relapsing-remitting multiple sclerosis (MS). Here we report the first case of non-traumatic acute epidural hematoma in a relapsing-remitting MS patient treated with fingolimod. Fingolimod might increase the risk of hemorrhage by enhancing vasospasm and causing vascular disruption. Switching fingolimod to other disease-modifying drugs, including dimethyl fumarate, should be considered when non-traumatic hemorrhage is observed in MS patients.

7.
Brain Dev ; 41(4): 378-381, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30414707

RESUMEN

BACKGROUND: Cardio-facio-cutaneous syndrome (CFCS) is a rare genetic disorder characterized by cardiovascular anomalies, dysmorphic faces, ectodermal abnormalities and developmental delays. Mutations in BRAF and other RAS-MAPK pathway-associated genes are commonly identified in patients with CFCS. While this molecular pathway is known to be associated with neuro-inflammatory conditions, only one case with CFCS has been reported thus far to develop acute encephalopathy in childhood. CASE REPORT: A 3-year-old boy with dysmorphic features and mild psychomotor delay developed acute encephalopathy. After a 45-min long, generalized seizure, the magnetic resonance imaging revealed that the restricted diffusion signals spread to the bilateral subcortical white matters on day 1 of illness. Despite the 14 days of intensive care, the acute symptoms of encephalopathy left him intractable epilepsy and severe neurocognitive impairments. The whole-exome sequencing analysis identified a de novo heterozygous mutation of BRAF (NM_004333:p.Thr241Met) in this case. CONCLUSION: The present case suggests that the hyperactive condition of ERK signals might augment the development of acute encephalopathy and post-encephalopathic epilepsy in childhood.


Asunto(s)
Encefalopatías/etiología , Displasia Ectodérmica/fisiopatología , Insuficiencia de Crecimiento/fisiopatología , Cardiopatías Congénitas/fisiopatología , Proteínas Proto-Oncogénicas B-raf/genética , Anomalías Múltiples/genética , Encefalopatías/complicaciones , Encefalopatías/genética , Niño , Epilepsia Refractaria/complicaciones , Displasia Ectodérmica/complicaciones , Facies , Insuficiencia de Crecimiento/complicaciones , Cardiopatías Congénitas/complicaciones , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Mutación , Proteínas Proto-Oncogénicas B-raf/fisiología
8.
Eur J Med Genet ; 61(8): 451-454, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29510241

RESUMEN

Mutations in the X-linked gene CDKL5 cause early-onset epileptic encephalopathy and severe developmental delay. Because this disorder predominantly affects females, the full clinical spectrum of male patients remains elusive. We herein report a 16-year-old boy, who suffered from intractable seizures 20 days after birth. Serial electroencephalograms detected recurrent focal epileptiform discharges from age 4 months, which evolved to hypsarrhythmia later in infancy. Mass-spectrometric analyses revealed increase in urinary excretion of methylmalonic acid without perturbed concentrations of propionic acid, homocystein and methionine. Whole-exome sequencing identified a de novo, truncating mutation in CDKL5 (NM_003159.2:c.419dupA, p.Asn140Lysfs*8). Targeted sequencing excluded concomitant mutations in methylmalonic academia-associated genes. No methylmalonic acidemia has been reported in children with CDKL5 disorder. Extensive analyses on organic acid metabolism for males with CDKL5 mutations will gain more insight into their biochemical profiles in infancy.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Encefalopatías/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Errores Innatos del Metabolismo de los Aminoácidos/patología , Encefalopatías/patología , Humanos , Masculino , Metionina/orina , Ácido Metilmalónico/orina , Propionatos/orina , Síndrome
9.
J Hum Genet ; 63(4): 417-423, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29403087

RESUMEN

Autosomal recessive cerebellar ataxias (ARCAs) are clinically and genetically heterogeneous neurological disorders. Through whole-exome sequencing of Japanese ARCA patients, we identified three index patients from unrelated families who had biallelic mutations in ERCC4. ERCC4 mutations have been known to cause xeroderma pigmentosum complementation group F (XP-F), Cockayne syndrome, and Fanconi anemia phenotypes. All of the patients described here showed very slowly progressive cerebellar ataxia and cognitive decline with choreiform involuntary movement, with young adolescent or midlife onset. Brain MRI demonstrated atrophy that included the cerebellum and brainstem. Of note, cutaneous symptoms were very mild: there was normal to very mild pigmentation of exposed skin areas and/or an equivocal history of pathological sunburn. However, an unscheduled DNA synthesis assay of fibroblasts from the patient revealed impairment of nucleotide excision repair. A similar phenotype was very recently recognized through genetic analysis of Caucasian cerebellar ataxia patients. Our results confirm that biallelic ERCC4 mutations cause a cerebellar ataxia-dominant phenotype with mild cutaneous symptoms, possibly accounting for a high proportion of the genetic causes of ARCA in Japan, where XP-F is prevalent.


Asunto(s)
Ataxia Cerebelosa/diagnóstico , Ataxia Cerebelosa/genética , Proteínas de Unión al ADN/genética , Genes Dominantes , Mutación , Fenotipo , Adulto , Edad de Inicio , Anciano , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Encéfalo/anomalías , Encéfalo/diagnóstico por imagen , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Linaje
10.
J Neuroinflammation ; 15(1): 46, 2018 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-29454354

RESUMEN

BACKGROUND: Although inflammation in the central nervous system is responsible for multiple neurological diseases, the lack of appropriate biomarkers makes it difficult to evaluate inflammatory activities in these diseases. Therefore, a new biomarker reflecting neuroinflammation is required for accurate diagnosis, appropriate therapy, and comprehension of pathogenesis of these neurological disorders. We previously reported that the cerebrospinal fluid (CSF) concentration of lateral olfactory tract usher substance (LOTUS), which promotes axonal growth as a Nogo receptor 1 antagonist, negatively correlates with disease activity in multiple sclerosis, suggesting that variation in LOTUS reflects the inflammatory activities and is a useful biomarker to evaluate the disease activity. To extend this observation, we analyzed the variation of LOTUS in the CSF of patients with bacterial and viral meningitis, which are the most common neuroinflammatory diseases. METHODS: CSF samples were retrospectively obtained from patients with meningitis (n = 40), who were followed up by CSF study at least twice, and from healthy controls (n = 27). Patients were divided into bacterial (n = 14) and viral meningitis (n = 18) after exclusion of eight patients according to the criteria of this study. LOTUS concentrations, total protein levels, and CSF cell counts in the acute and recovery phases were analyzed chronologically. We also used lipopolysaccharide-injected mice as a model of neuroinflammation to evaluate LOTUS mRNA and protein expression in the brain. RESULTS: Regardless of whether meningitis was viral or bacterial, LOTUS concentrations in the CSF of patients in acute phase were lower than those of healthy controls. As the patients recovered from meningitis, LOTUS levels in the CSF returned to the normal range. Lipopolysaccharide-injected mice also exhibited reduced LOTUS mRNA and protein expression in the brain. CONCLUSIONS: CSF levels of LOTUS correlated inversely with disease activity in both bacterial and viral meningitis, as well as in multiple sclerosis, because neuroinflammation downregulated LOTUS expression. Our data strongly suggest that variation of CSF LOTUS is associated with neuroinflammation and is useful as a biomarker for a broader range of neuroinflammatory diseases.


Asunto(s)
Proteínas de Unión al Calcio/líquido cefalorraquídeo , Meningitis/líquido cefalorraquídeo , Meningitis/diagnóstico , Receptor Nogo 1/antagonistas & inhibidores , Receptor Nogo 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/líquido cefalorraquídeo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
11.
Cell Rep ; 22(3): 734-747, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29346770

RESUMEN

Recent studies have established important roles of de novo mutations (DNMs) in autism spectrum disorders (ASDs). Here, we analyze DNMs in 262 ASD probands of Japanese origin and confirm the "de novo paradigm" of ASDs across ethnicities. Based on this consistency, we combine the lists of damaging DNMs in our and published ASD cohorts (total number of trios, 4,244) and perform integrative bioinformatics analyses. Besides replicating the findings of previous studies, our analyses highlight ATP-binding genes and fetal cerebellar/striatal circuits. Analysis of individual genes identified 61 genes enriched for damaging DNMs, including ten genes for which our dataset now contributes to statistical significance. Screening of compounds altering the expression of genes hit by damaging DNMs reveals a global downregulating effect of valproic acid, a known risk factor for ASDs, whereas cardiac glycosides upregulate these genes. Collectively, our integrative approach provides deeper biological and potential medical insights into ASDs.


Asunto(s)
Trastorno del Espectro Autista/genética , Mutación/genética , Trastorno del Espectro Autista/patología , Predisposición Genética a la Enfermedad , Humanos
13.
Am J Pathol ; 188(2): 507-514, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29128563

RESUMEN

Mutations in the MATR3 gene have been identified as a cause of familial amyotrophic lateral sclerosis, but involvement of the matrin 3 (MATR3) protein in sporadic amyotrophic lateral sclerosis (SALS) pathology has not been fully assessed. We immunohistochemically analyzed MATR3 pathology in the spinal cords of SALS and control autopsy specimens. MATR3 immunostaining of the motor neuron nuclei revealed two distinct patterns: mild and strong staining. There were no differences in the ratio of mild versus strong nuclear staining between the SALS and control cases. MATR3-containing neuronal cytoplasmic inclusions (NCIs) were observed in 60% of SALS cases. Most motor neurons with MATR3-positive NCIs exhibited a mild nuclear staining pattern. Although 16.8% of NCIs positive for transactivating response region DNA-binding protein 43 (TDP-43) were estimated as double-labeled by MATR3, no MATR3-positive or TDP-43-negative NCIs were observed. Although a previous study found that MATR3-positive NCIs are present only in cases with C9orf72 hexanucleotide repeat expansion, ubiquitin-positive granular NCIs were not observed in the cerebellum, which have been reported as specific to C9orf72-related ALS. Six ALS cases were confirmed to be negative for the GGGGCC hexanucleotide. Our results reveal that MATR3 is a component of TDP-43-positive NCIs in motor neurons, even in SALS, and indicate the broader involvement of MATR3 in ALS pathology and the heterogeneity of TDP-43-positive NCIs.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Cuerpos de Inclusión/metabolismo , Neuronas Motoras/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/patología , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Cuerpos de Inclusión/patología , Vértebras Lumbares , Masculino , Persona de Mediana Edad , Neuronas Motoras/patología , Médula Espinal/metabolismo
14.
Brain Dev ; 39(5): 439-443, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27916449

RESUMEN

We report the case of a 19-year-old female patient who had progressive chorea associated with a GNAO1 mutation. Chorea was refractory to multiple anticonvulsants, and the patient suffered from tiapride-induced neuroleptic malignant syndrome. After identification of a GNAO1 missense mutation at the age of 18years, topiramate treatment was initiated and the frequency of chorea decreased dramatically. The efficacy of topiramate may have been related to the inhibitory modulation of voltage-activated Ca2+ channels. Given the side effects and complications associated with neuroleptics and deep brain stimulation, respectively, topiramate is recommended for the first-line management of severe chorea associated with a GNAO1 mutation.


Asunto(s)
Fructosa/análogos & derivados , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Trastornos del Movimiento/tratamiento farmacológico , Trastornos del Movimiento/genética , Mutación/genética , Bases de Datos Bibliográficas/estadística & datos numéricos , Femenino , Fructosa/uso terapéutico , Humanos , Imagen por Resonancia Magnética , Trastornos del Movimiento/diagnóstico por imagen , Fármacos Neuroprotectores/uso terapéutico , Farmacogenética , Topiramato , Resultado del Tratamiento , Adulto Joven
15.
Am J Hum Genet ; 99(4): 950-961, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27666374

RESUMEN

We describe four families with affected siblings showing unique clinical features: early-onset (before 1 year of age) progressive diffuse brain atrophy with regression, postnatal microcephaly, postnatal growth retardation, muscle weakness/atrophy, and respiratory failure. By whole-exome sequencing, we identified biallelic TBCD mutations in eight affected individuals from the four families. TBCD encodes TBCD (tubulin folding co-factor D), which is one of five tubulin-specific chaperones playing a pivotal role in microtubule assembly in all cells. A total of seven mutations were found: five missense mutations, one nonsense, and one splice site mutation resulting in a frameshift. In vitro cell experiments revealed the impaired binding between most mutant TBCD proteins and ARL2, TBCE, and ß-tubulin. The in vivo experiments using olfactory projection neurons in Drosophila melanogaster indicated that the TBCD mutations caused loss of function. The wide range of clinical severity seen in this neurodegenerative encephalopathy may result from the residual function of mutant TBCD proteins. Furthermore, the autopsied brain from one deceased individual showed characteristic neurodegenerative findings: cactus and somatic sprout formations in the residual Purkinje cells in the cerebellum, which are also seen in some diseases associated with mitochondrial impairment. Defects of microtubule formation caused by TBCD mutations may underlie the pathomechanism of this neurodegenerative encephalopathy.


Asunto(s)
Alelos , Encefalopatías/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Enfermedades Neurodegenerativas/genética , Adolescente , Edad de Inicio , Secuencia de Aminoácidos , Animales , Encefalopatías/patología , Encefalopatías/fisiopatología , Niño , Preescolar , Drosophila melanogaster/genética , Exoma , Femenino , Mutación del Sistema de Lectura/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Sitios de Empalme de ARN/genética , Tubulina (Proteína)/metabolismo , Adulto Joven
16.
BMC Neurol ; 16: 174, 2016 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-27634470

RESUMEN

BACKGROUND: Alternating hemiplegia of childhood (AHC) is a rare neurological disorder that manifests recurrent attacks of hemiplegia, oculogyric, and choreoathetotic involuntary movements. De novo mutations in ATP1A3 cause three types of neurological diseases: AHC; rapid-onset dystonia-Parkinsonism (RDP); and cerebellar ataxia, areflexia, pes cavus, optic atrophy, and sensorineural hearing loss (CAPOS) syndromes. It remains to be determined whether or not a rare mutation in ATP1A3 may cause atypical phenotypes. CASE PRESENTATION: A 7-year-old boy presented with recurrent symptoms of generalized paralysis since 1 year and 5 months of age. Hypotonia, dystonia, and choreoathetosis persisted with exacerbation under febrile conditions, but no cerebellar ataxia had ever evolved in 6 years. Whole-exome sequencing (WES) was performed to determine his genetic background, and mutations were validated by the Sanger method. Crude protein extracts were prepared from the cultured cells, and expression of the wild-type or mutant ATP1A3 proteins were analyzed by Western blotting. WES identified a de novo pathogenic mutation in ATP1A3 (c.2266C > T:p.R756C) for this patient. A literature overview of two reported cases with p.R756C and p.R756H mutations showed both overlapping and distinct phenotypes when compared with those of the present case. The expression of the mutant form (R756C) of ATP1A3 did not differ markedly from that of the wild-type and D801N proteins. CONCLUSIONS: This study confirmed that p.R756C mutation of ATP1A3 cause atypical forms of AHC-associated disorders. The wide spectra of neurological phenotypes in AHC are linked to as-yet-unknown deficits in the functions of mutant ATP1A3.


Asunto(s)
Atetosis/complicaciones , Atetosis/genética , Corea/genética , Hemiplejía/complicaciones , Hemiplejía/genética , Parálisis/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Niño , Corea/complicaciones , Humanos , Masculino , Mutación , Parálisis/complicaciones , Fenotipo
17.
Ann Hum Genet ; 80(4): 235-40, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27346735

RESUMEN

BACKGROUND: Early-onset epileptic encephalopathy (EOEE) is a heterogeneous group of neurodevelopmental disorders characterised by infantile-onset intractable epilepsy and unfavourable developmental outcomes. Hundreds of mutations have been reported to cause EOEE; however, little is known about the clinical features of individuals with rare variants. CASE REPORT AND METHODS: We present a 10-year-old boy with severe developmental delay. He started experiencing recurrent focal seizures at 2 months old. Serial electroencephalograms persistently detected epileptiform discharges from the left hemisphere. Whole-exome sequencing and array-comparative genome hybridization were performed to search for de novo variations. Two-week-old C57Bl/6 mice were used for immunofluorescence studies. RESULTS: This case had a paternally inherited, 0.2-Mb duplication at chromosome 22q11.22. The whole-exome sequencing identified a de novo truncating mutation of tripartite motif containing 8 (TRIM8) (NM_030912:c.1099_1100insG:p.C367fs), one of the epileptic encephalopathy-associated genes. We verified that the murine homologues of these genes are expressed in the postnatal mouse brain. CONCLUSION: This is the second case of EOEE caused by a de novo truncating mutation of TRIM8. Further studies are required to determine the functional roles of TRIM8 in the postnatal development of the human brain and its functional relationships with other EOEE-associated genes.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Tejido Nervioso/genética , Espasmos Infantiles/diagnóstico , Animales , Secuencia de Bases , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Niño , Codón sin Sentido , Análisis Mutacional de ADN , Discapacidades del Desarrollo/genética , Exoma , Expresión Génica , Humanos , Lactante , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Espasmos Infantiles/genética
18.
J Hum Genet ; 61(5): 381-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26818738

RESUMEN

The voltage-gated Kv10.1 potassium channel, also known as ether-a-go-go-related gene 1, encoded by KCNH1 (potassium voltage-gated channel, subfamily H (eag related), member 1) is predominantly expressed in the central nervous system. Recently, de novo missense KCNH1 mutations have been identified in six patients with Zimmermann-Laband syndrome and in four patients with Temple-Baraitser syndrome. These syndromes were historically considered distinct. Here we report three de novo missense KCNH1 mutations in four patients with syndromic developmental delay and epilepsy. Two novel KCNH1 mutations (p.R357Q and p.R357P), found in three patients, were located at the evolutionally highly conserved arginine in the channel voltage-sensor domain (S4). Another mutation (p.G496E) was found in the channel pore domain (S6) helix, which acts as a hinge in activation gating and mainly conducts non-inactivating outward potassium current. A previously reported p.G496R mutation was shown to produce no voltage-dependent outward current in CHO cells, suggesting that p.G496E may also disrupt the proper function of the Kv channel pore. Our report confirms that KCNH1 mutations are associated with syndromic neurodevelopmental disorder, and also support the functional importance of the S4 domain.


Asunto(s)
Discapacidades del Desarrollo/genética , Canales de Potasio Éter-A-Go-Go/genética , Hipotonía Muscular/genética , Mutación , Convulsiones/genética , Alelos , Sustitución de Aminoácidos , Niño , Discapacidades del Desarrollo/diagnóstico , Canales de Potasio Éter-A-Go-Go/química , Exoma , Facies , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Hipotonía Muscular/diagnóstico , Fenotipo , Dominios y Motivos de Interacción de Proteínas/genética , Convulsiones/diagnóstico , Síndrome
19.
J Hum Genet ; 61(5): 451-5, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26763878

RESUMEN

Three recessive mutations in the sodium leak channel, nonselective (NALCN) have been reported to cause intellectual disability and hypotonia. In addition, 14 de novo heterozygous mutations have been identified in 15 patients with arthrogryposis and neurodevelopmental impairment. Here, we report three patients with neurodevelopmental disease and hypotonia, harboring one recurrent (p.R1181Q) and two novel mutations (p.L312V and p.V1020F) occurring de novo in NALCN. Mutation p.L312 is located in the pore forming S6 region of domain I and p.V1020F in the S5 region of domain III. Mutation p.R1181Q is in a linker region. Mapping these three mutations to a model of NALCN showed p.Leu312 and p.Val1020 positioned in the hydrophobic core of the pore modules, indicating these two mutations may affect the gating function of NALCN. Although p.R1181Q is unlikely to affect the ion channel structure, previous studies have shown that an analogous mutation in Caenorhabditis elegans produced a phenotype with a coiling locomotion, suggesting that p.R1181Q could also affect NALCN function. Our three patients showed profound intellectual disability and growth delay, facial dysmorphologies and hypotonia. The present data support previous work suggesting heterozygous NALCN mutations lead to syndromic neurodevelopmental impairment.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Mutación Missense , Canales de Sodio/genética , Alelos , Preescolar , Discapacidades del Desarrollo/diagnóstico , Femenino , Genotipo , Heterocigoto , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Canales Iónicos , Masculino , Proteínas de la Membrana , Modelos Moleculares , Hipotonía Muscular/diagnóstico , Fenotipo , Conformación Proteica , Análisis de Secuencia de ADN , Canales de Sodio/química , Síndrome
20.
J Neurosurg ; 124(6): 1858-66, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26636390

RESUMEN

OBJECT The authors' goal in this paper is to provide the first clinical, radiological, and genetic studies of panventriculomegaly (PaVM) defined by a wide foramen of Magendie and large cisterna magna. METHODS Clinical and brain imaging data from 28 PaVM patients (including 10 patients from 5 families) were retrospectively studied. Five children were included. In adult patients, the age at onset was 56.0 ± 16.7 years. Tetraventricular dilation, aqueductal opening with flow void on T2-weighted images, and a wide foramen of Magendie and large cisterna magna (wide cerebrospinal fluid space at the fourth ventricle outlet) were essential MRI findings for PaVM diagnosis. 3D fast asymmetrical spin echo sequences were used for visualization of cistern membranes. Time-spatial labeling inversion pulse examination was performed to analyze cerebrospinal fluid movement. Copy number variations were determined using high-resolution microarray and were validated by quantitative polymerase chain reaction with breakpoint sequencing. RESULTS Adult patients showed gait disturbance, urinary dysfunction, and cognitive dysfunction. Five infant patients exhibited macrocranium. Patients were divided into 2 subcategories, those with or without downward bulging third ventricular floors and membranous structures in the prepontine cistern. Patients with bulging floors were successfully treated with endoscopic third ventriculostomy. Genetic analysis revealed a deletion in DNAH14 that encodes a dynein heavy chain protein associated with motile cilia function, and which co-segregated with patients in a family without a downward bulging third ventricular floor. CONCLUSIONS Panventriculomegaly with a wide foramen of Magendie and a large cisterna magna may belong to a subtype of congenital hydrocephalus with familial accumulation, younger age at onset, and symptoms of normal pressure hydrocephalus. In addition, a family with PaVM has a gene mutation associated with dysfunction of motile cilia.


Asunto(s)
Encéfalo/diagnóstico por imagen , Cisterna Magna/diagnóstico por imagen , Hidrocefalia/diagnóstico por imagen , Hidrocefalia/fisiopatología , Edad de Inicio , Encéfalo/patología , Líquido Cefalorraquídeo/fisiología , Variaciones en el Número de Copia de ADN , Dineínas/genética , Familia , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Genotipaje , Humanos , Hidrocefalia/genética , Hidrocefalia/cirugía , Imagenología Tridimensional , Imagen por Resonancia Magnética , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Linaje , Estudios Retrospectivos , Ventriculostomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...