Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Epilepsia ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953796

RESUMEN

OBJECTIVE: DYNC1H1 variants are involved on a disease spectrum from neuromuscular disorders to neurodevelopmental disorders. DYNC1H1-related epilepsy has been reported in small cohorts. We dissect the electroclinical features of 34 patients harboring de novo DYNC1H1 pathogenic variants, identify subphenotypes on the DYNC1H1-related epilepsy spectrum, and compare the genotype-phenotype correlations observed in our cohort with the literature. METHODS: Patients harboring de novo DYNC1H1 pathogenic variants were recruited through international collaborations. Clinical data were retrospectively collected. Latent class analysis was performed to identify subphenotypes. Multivariable binary logistic regression analysis was applied to investigate the association with DYNC1H1 protein domains. RESULTS: DYNC1H1-related epilepsy presented with infantile epileptic spasms syndrome (IESS) in 17 subjects (50%), and in 25% of these individuals the epileptic phenotype evolved into Lennox-Gastaut syndrome (LGS). In 12 patients (35%), focal onset epilepsy was defined. In two patients, the epileptic phenotype consisted of generalized myoclonic epilepsy, with a progressive phenotype in one individual harboring a frameshift variant. In approximately 60% of our cohort, seizures were drug-resistant. Malformations of cortical development were noticed in 79% of our patients, mostly on the lissencephaly-pachygyria spectrum, particularly with posterior predominance in a half of them. Midline and infratentorial abnormalities were additionally reported in 45% and 27% of subjects. We have identified three main classes of subphenotypes on the DYNC1H1-related epilepsy spectrum. SIGNIFICANCE: We propose a classification in which pathogenic de novo DYNC1H1 variants feature drug-resistant IESS in half of cases with potential evolution to LGS (Class 1), developmental and epileptic encephalopathy other than IESS and LGS (Class 2), or less severe focal or genetic generalized epilepsy including a progressive phenotype (Class 3). We observed an association between stalk domain variants and Class 1 phenotypes. The variants p.Arg309His and p.Arg1962His were common and associated with Class 1 subphenotype in our cohort. These findings may aid genetic counseling of patients with DYNC1H1-related epilepsy.

3.
Genet Med ; 24(11): 2351-2366, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36083290

RESUMEN

PURPOSE: Germline loss-of-function variants in CTNNB1 cause neurodevelopmental disorder with spastic diplegia and visual defects (NEDSDV; OMIM 615075) and are the most frequent, recurrent monogenic cause of cerebral palsy (CP). We investigated the range of clinical phenotypes owing to disruptions of CTNNB1 to determine the association between NEDSDV and CP. METHODS: Genetic information from 404 individuals with collectively 392 pathogenic CTNNB1 variants were ascertained for the study. From these, detailed phenotypes for 52 previously unpublished individuals were collected and combined with 68 previously published individuals with comparable clinical information. The functional effects of selected CTNNB1 missense variants were assessed using TOPFlash assay. RESULTS: The phenotypes associated with pathogenic CTNNB1 variants were similar. A diagnosis of CP was not significantly associated with any set of traits that defined a specific phenotypic subgroup, indicating that CP is not additional to NEDSDV. Two CTNNB1 missense variants were dominant negative regulators of WNT signaling, highlighting the utility of the TOPFlash assay to functionally assess variants. CONCLUSION: NEDSDV is a clinically homogeneous disorder irrespective of initial clinical diagnoses, including CP, or entry points for genetic testing.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Fenotipo , Trastornos del Neurodesarrollo/genética , Vía de Señalización Wnt/genética , Discapacidad Intelectual/genética , Genómica , beta Catenina/genética
4.
Am J Med Genet A ; 188(9): 2617-2626, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35670295

RESUMEN

Pathogenic variants in CASK, an X-linked gene that plays a role in brain development and synaptic function, are the cause of both microcephaly with pontine and cerebellar hypoplasia (MICPCH), and X-linked intellectual disability (XLID) with or without nystagmus. MICPCH is caused by loss of function variants in CASK, typically affects females, and is associated with moderate-to-severe intellectual disability (ID). Additional findings, present in about one-third of individuals, include feeding difficulties, ophthalmologic issues, hypertonicity, epilepsy, and sensorineural hearing loss. Only a few affected males with MICPCH phenotype have been reported and most have had profound developmental disability and intractable epilepsy. The XLID phenotype is typically caused by missense variants and most often manifests in males; carrier females are mildly affected or unaffected. Nystagmus is often present. In total, over 175 patients have been reported in the literature. We now report an additional 11 patients with pathogenic variants in CASK that expand these phenotypes and reported genotype-phenotype correlations.


Asunto(s)
Discapacidad Intelectual , Cerebelo/anomalías , Discapacidades del Desarrollo , Femenino , Estudios de Asociación Genética , Guanilato-Quinasas/genética , Humanos , Discapacidad Intelectual/genética , Masculino , Discapacidad Intelectual Ligada al Cromosoma X , Microcefalia , Mutación , Malformaciones del Sistema Nervioso , Fenotipo
5.
Am J Hum Genet ; 102(6): 1158-1168, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861105

RESUMEN

ßIV spectrin links ankyrinG (AnkG) and clustered ion channels at axon initial segments (AISs) and nodes of Ranvier to the axonal cytoskeleton. Here, we report bi-allelic pathogenic SPTBN4 variants (three homozygous and two compound heterozygous) that cause a severe neurological syndrome that includes congenital hypotonia, intellectual disability, and motor axonal and auditory neuropathy. We introduced these variants into ßIV spectrin, expressed these in neurons, and found that 5/7 were loss-of-function variants disrupting AIS localization or abolishing phosphoinositide binding. Nerve biopsies from an individual with a loss-of-function variant had reduced nodal Na+ channels and no nodal KCNQ2 K+ channels. Modeling the disease in mice revealed that although ankyrinR (AnkR) and ßI spectrin can cluster Na+ channels and partially compensate for the loss of AnkG and ßIV spectrin at nodes of Ranvier, AnkR and ßI spectrin cannot cluster KCNQ2- and KCNQ3-subunit-containing K+ channels. Our findings define a class of spectrinopathies and reveal the molecular pathologies causing nervous-system dysfunction.


Asunto(s)
Axones/patología , Discapacidad Intelectual/genética , Enfermedad de la Neurona Motora/genética , Hipotonía Muscular/congénito , Hipotonía Muscular/genética , Proteínas del Tejido Nervioso/genética , Espectrina/genética , Alelos , Animales , Axones/metabolismo , Células COS , Niño , Preescolar , Chlorocebus aethiops , Femenino , Células HEK293 , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/fisiopatología , Lípidos , Masculino , Ratones Noqueados , Enfermedad de la Neurona Motora/complicaciones , Enfermedad de la Neurona Motora/fisiopatología , Hipotonía Muscular/complicaciones , Hipotonía Muscular/fisiopatología , Proteínas Mutantes/metabolismo , Mutación/genética , Ratas Sprague-Dawley
6.
Am J Hum Genet ; 102(1): 44-57, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29276004

RESUMEN

Although the role of typical Rho GTPases and other Rho-linked proteins in synaptic plasticity and cognitive function and dysfunction is widely acknowledged, the role of atypical Rho GTPases (such as RHOBTB2) in neurodevelopment has barely been characterized. We have now identified de novo missense variants clustering in the BTB-domain-encoding region of RHOBTB2 in ten individuals with a similar phenotype, including early-onset epilepsy, severe intellectual disability, postnatal microcephaly, and movement disorders. Three of the variants were recurrent. Upon transfection of HEK293 cells, we found that mutant RHOBTB2 was more abundant than the wild-type, most likely because of impaired degradation in the proteasome. Similarly, elevated amounts of the Drosophila ortholog RhoBTB in vivo were associated with seizure susceptibility and severe locomotor defects. Knockdown of RhoBTB in the Drosophila dendritic arborization neurons resulted in a decreased number of dendrites, thus suggesting a role of RhoBTB in dendritic development. We have established missense variants in the BTB-domain-encoding region of RHOBTB2 as causative for a developmental and epileptic encephalopathy and have elucidated the role of atypical Rho GTPase RhoBTB in Drosophila neurological function and possibly dendrite development.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Epilepsia/genética , Proteínas de Unión al GTP/genética , Mutación Missense/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Secuencia de Aminoácidos , Animales , Conducta Animal , Niño , Preescolar , Dendritas/metabolismo , Femenino , Proteínas de Unión al GTP/química , Dosificación de Gen , Células HEK293 , Humanos , Masculino , Fenotipo , Sinapsis/patología , Proteínas Supresoras de Tumor/química
7.
Dev Neurosci ; 43(3-4): 222-229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34350863

RESUMEN

Mitochondria, the organelles classically seen as the powerhouse of the cell, are increasingly associated with a wide variety of neurodevelopmental disorders. Although individually rare, a myriad of pediatric neurogenetic disorders have been identified in the last few years, thanks to advances in clinical genetic sequencing and data analysis. As this exponential growth continues, mitochondrial dysfunction is increasingly implicated in childhood neurodevelopmental disorders, with clinical presentations ranging from syndromic autism, intellectual disability, and epileptic encephalopathies to childhood onset neurodegeneration. Here we review recent evidence demonstrating mitochondrial involvement in neurodevelopmental disorders, identify emerging mechanistic trends, and reconsider the long-standing question of the role of mitochondria in light of new evidence: causation versus mere association.


Asunto(s)
Trastorno Autístico , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Discapacidad Intelectual/genética , Mitocondrias , Trastornos del Neurodesarrollo/genética
8.
Genet Med ; 23(10): 1922-1932, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34163037

RESUMEN

PURPOSE: CACNA1C encodes the alpha-1-subunit of a voltage-dependent L-type calcium channel expressed in human heart and brain. Heterozygous variants in CACNA1C have previously been reported in association with Timothy syndrome and long QT syndrome. Several case reports have suggested that CACNA1C variation may also be associated with a primarily neurological phenotype. METHODS: We describe 25 individuals from 22 families with heterozygous variants in CACNA1C, who present with predominantly neurological manifestations. RESULTS: Fourteen individuals have de novo, nontruncating variants and present variably with developmental delays, intellectual disability, autism, hypotonia, ataxia, and epilepsy. Functional studies of a subgroup of missense variants via patch clamp experiments demonstrated differential effects on channel function in vitro, including loss of function (p.Leu1408Val), neutral effect (p.Leu614Arg), and gain of function (p.Leu657Phe, p.Leu614Pro). The remaining 11 individuals from eight families have truncating variants in CACNA1C. The majority of these individuals have expressive language deficits, and half have autism. CONCLUSION: We expand the phenotype associated with CACNA1C variants to include neurodevelopmental abnormalities and epilepsy, in the absence of classic features of Timothy syndrome or long QT syndrome.


Asunto(s)
Trastorno Autístico , Canales de Calcio Tipo L , Síndrome de QT Prolongado , Sindactilia , Trastorno Autístico/genética , Canales de Calcio Tipo L/genética , Humanos , Fenotipo
9.
Clin Genet ; 100(2): 187-200, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33955014

RESUMEN

Mutations affecting the transcriptional regulator Ankyrin Repeat Domain 11 (ANKRD11) are mainly associated with the multisystem developmental disorder known as KBG syndrome, but have also been identified in individuals with Cornelia de Lange syndrome (CdLS) and other developmental disorders caused by variants affecting different chromatin regulators. The extensive functional overlap of these proteins results in shared phenotypical features, which complicate the assessment of the clinical diagnosis. Additionally, re-evaluation of individuals at a later age occasionally reveals that the initial phenotype has evolved toward clinical features more reminiscent of a developmental disorder different from the one that was initially diagnosed. For this reason, variants in ANKRD11 can be ascribed to a broader class of disorders that fall within the category of the so-called chromatinopathies. In this work, we report on the clinical characterization of 23 individuals with variants in ANKRD11. The subjects present primarily with developmental delay, intellectual disability and dysmorphic features, and all but two received an initial clinical diagnosis of either KBG syndrome or CdLS. The number and the severity of the clinical signs are overlapping but variable and result in a broad spectrum of phenotypes, which could be partially accounted for by the presence of additional molecular diagnoses and distinct pathogenic mechanisms.


Asunto(s)
Anomalías Múltiples/etiología , Enfermedades del Desarrollo Óseo/etiología , Discapacidad Intelectual/etiología , Proteínas Represoras/genética , Anomalías Dentarias/etiología , Anomalías Múltiples/genética , Adolescente , Enfermedades del Desarrollo Óseo/genética , Niño , Preescolar , Cara/anomalías , Facies , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Mutación , Linaje , Anomalías Dentarias/genética , Adulto Joven
10.
Am J Med Genet A ; 185(6): 1700-1711, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33751773

RESUMEN

Over the past decade, pathogenic variants in all members of the ASXL family of genes, ASXL1, ASXL2, and ASXL3, have been found to lead to clinically distinct but overlapping syndromes. Bohring-Opitz syndrome (BOPS) was first described as a clinical syndrome and later found to be associated with pathogenic variants in ASXL1. This syndrome is characterized by developmental delay, microcephaly, characteristic facies, hypotonia, and feeding difficulties. Subsequently, pathogenic variants in ASXL2 were found to lead to Shashi-Pena syndrome (SHAPNS) and in ASXL3 to lead to Bainbridge-Ropers syndrome (BRPS). While SHAPNS and BRPS share many core features with BOPS, there also seem to be emerging clear differences. Here, we present five cases of BOPS, one case of SHAPNS, and four cases of BRPS. By adding our cohort to the limited number of previously published patients, we review the overlapping features of ASXL-related diseases that bind them together, while focusing on the characteristics that make each neurodevelopmental syndrome unique. This will assist in diagnosis of these overlapping conditions and allow clinicians to more comprehensively counsel affected families.


Asunto(s)
Craneosinostosis/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Preescolar , Craneosinostosis/patología , Discapacidades del Desarrollo/epidemiología , Discapacidades del Desarrollo/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Microcefalia , Hipotonía Muscular/epidemiología , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Mutación , Fenotipo , Adulto Joven
11.
Epilepsia ; 62(7): 1617-1628, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34075580

RESUMEN

OBJECTIVE: Improvement in epilepsy care requires standardized methods to assess disease severity. We report the results of implementing common data elements (CDEs) to document epilepsy history data in the electronic medical record (EMR) after 12 months of clinical use in outpatient encounters. METHODS: Data regarding seizure frequency were collected during routine clinical encounters using a CDE-based form within our EMR. We extracted CDE data from the EMR and developed measurements for seizure severity and seizure improvement scores. Seizure burden and improvement was evaluated by patient demographic and encounter variables for in-person and telemedicine encounters. RESULTS: We assessed a total of 1696 encounters in 1038 individuals with childhood epilepsies between September 6, 2019 and September 11, 2020 contributed by 32 distinct providers. Childhood absence epilepsy (n = 121), Lennox-Gastaut syndrome (n = 86), and Dravet syndrome (n = 42) were the most common epilepsy syndromes. Overall, 43% (737/1696) of individuals had at least monthly seizures, 17% (296/1696) had a least daily seizures, and 18% (311/1696) were seizure-free for >12 months. Quantification of absolute seizure burden and changes in seizure burden over time differed between epilepsy syndromes, including high and persistent seizure burden in patients with Lennox-Gastaut syndrome. Individuals seen via telemedicine or in-person encounters had comparable seizure frequencies. Individuals identifying as Hispanic/Latino, particularly from postal codes with lower median household incomes, were more likely to have ongoing seizures that worsened over time. SIGNIFICANCE: Standardized documentation of clinical data in childhood epilepsies through CDE can be implemented in routine clinical care at scale and enables assessment of disease burden, including characterization of seizure burden over time. Our data provide insights into heterogeneous patterns of seizure control in common pediatric epilepsy syndromes and will inform future initiatives focusing on patient-centered outcomes in childhood epilepsies, including the impact of telemedicine and health care disparities.


Asunto(s)
Costo de Enfermedad , Registros Electrónicos de Salud , Epilepsia/economía , Adolescente , Anticonvulsivantes/uso terapéutico , Niño , Preescolar , Elementos de Datos Comunes , Epilepsias Mioclónicas/epidemiología , Epilepsia Tipo Ausencia/epidemiología , Femenino , Hispánicos o Latinos , Humanos , Síndrome de Lennox-Gastaut/epidemiología , Masculino , Convulsiones/epidemiología , Factores Socioeconómicos , Telemedicina , Resultado del Tratamiento
12.
Pediatr Radiol ; 50(10): 1448-1475, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32642802

RESUMEN

This article is the second of a two-part series on intracranial calcification in childhood. In Part 1, the authors discussed the main differences between physiological and pathological intracranial calcification. They also outlined histological intracranial calcification characteristics and how these can be detected across different neuroimaging modalities. Part 1 emphasized the importance of age at presentation and intracranial calcification location and proposed a comprehensive neuroimaging approach toward the differential diagnosis of the causes of intracranial calcification. Pathological intracranial calcification can be divided into infectious, congenital, endocrine/metabolic, vascular, and neoplastic. In Part 2, the chief focus is on discussing endocrine/metabolic, vascular, and neoplastic intracranial calcification etiologies of intracranial calcification. Endocrine/metabolic diseases causing intracranial calcification are mainly from parathyroid and thyroid dysfunction and inborn errors of metabolism, such as mitochondrial disorders (MELAS, or mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes; Kearns-Sayre; and Cockayne syndromes), interferonopathies (Aicardi-Goutières syndrome), and lysosomal disorders (Krabbe disease). Specific noninfectious causes of intracranial calcification that mimic TORCH (toxoplasmosis, other [syphilis, varicella-zoster, parvovirus B19], rubella, cytomegalovirus, and herpes) infections are known as pseudo-TORCH. Cavernous malformations, arteriovenous malformations, arteriovenous fistulas, and chronic venous hypertension are also known causes of intracranial calcification. Other vascular-related causes of intracranial calcification include early atherosclerosis presentation (children with risk factors such as hyperhomocysteinemia, familial hypercholesterolemia, and others), healed hematoma, radiotherapy treatment, old infarct, and disorders of the microvasculature such as COL4A1- and COL4A2-related diseases. Intracranial calcification is also seen in several pediatric brain tumors. Clinical and familial information such as age at presentation, maternal exposure to teratogens including viruses, and association with chromosomal abnormalities, pathogenic genes, and postnatal infections facilitates narrowing the differential diagnosis of the multiple causes of intracranial calcification.


Asunto(s)
Encefalopatías/diagnóstico por imagen , Encefalopatías/etiología , Calcinosis/diagnóstico por imagen , Calcinosis/etiología , Neuroimagen/métodos , Niño , Preescolar , Humanos , Lactante , Recién Nacido
13.
Pediatr Radiol ; 50(10): 1424-1447, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32734340

RESUMEN

This article is the first of a two-part series on intracranial calcification in childhood. Intracranial calcification can be either physiological or pathological. Physiological intracranial calcification is not an expected neuroimaging finding in the neonatal or infantile period but occurs, as children grow older, in the pineal gland, habenula, choroid plexus and occasionally the dura mater. Pathological intracranial calcification can be broadly divided into infectious, congenital, endocrine/metabolic, vascular and neoplastic. The main goals in Part 1 are to discuss the chief differences between physiological and pathological intracranial calcification, to discuss the histological characteristics of intracranial calcification and how intracranial calcification can be detected across neuroimaging modalities, to emphasize the importance of age at presentation and intracranial calcification location, and to propose a comprehensive neuroimaging approach toward the differential diagnosis of the causes of intracranial calcification. Finally, in Part 1 the authors discuss the most common causes of infectious intracranial calcification, especially in the neonatal period, and congenital causes of intracranial calcification. Various neuroimaging modalities have distinct utilities and sensitivities in the depiction of intracranial calcification. Age at presentation, intracranial calcification location, and associated neuroimaging findings are useful information to help narrow the differential diagnosis of intracranial calcification. Intracranial calcification can occur in isolation or in association with other neuroimaging features. Intracranial calcification in congenital infections has been associated with clastic changes, hydrocephalus, chorioretinitis, white matter abnormalities, skull changes and malformations of cortical development. Infections are common causes of intracranial calcification, especially neonatal TORCH (toxoplasmosis, other [syphilis, varicella-zoster, parvovirus B19], rubella, cytomegalovirus and herpes) infections.


Asunto(s)
Encefalopatías/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Calcificación Fisiológica , Calcinosis/diagnóstico por imagen , Neuroimagen/métodos , Niño , Preescolar , Humanos , Lactante , Recién Nacido
14.
Neurogenetics ; 20(3): 129-143, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31041561

RESUMEN

We previously reported a pathogenic de novo p.R342W mutation in the transcriptional corepressor CTBP1 in four independent patients with neurodevelopmental disabilities [1]. Here, we report the clinical phenotypes of seven additional individuals with the same recurrent de novo CTBP1 mutation. Within this cohort, we identified consistent CtBP1-related phenotypes of intellectual disability, ataxia, hypotonia, and tooth enamel defects present in most patients. The R342W mutation in CtBP1 is located within a region implicated in a high affinity-binding cleft for CtBP-interacting proteins. Unbiased proteomic analysis demonstrated reduced interaction of several chromatin-modifying factors with the CtBP1 W342 mutant. Genome-wide transcriptome analysis in human glioblastoma cell lines expressing -CtBP1 R342 (wt) or W342 mutation revealed changes in the expression profiles of genes controlling multiple cellular processes. Patient-derived dermal fibroblasts were found to be more sensitive to apoptosis during acute glucose deprivation compared to controls. Glucose deprivation strongly activated the BH3-only pro-apoptotic gene NOXA, suggesting a link between enhanced cell death and NOXA expression in patient fibroblasts. Our results suggest that context-dependent relief of transcriptional repression of the CtBP1 mutant W342 allele may contribute to deregulation of apoptosis in target tissues of patients leading to neurodevelopmental phenotypes.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Proteínas de Unión al ADN/genética , Mutación Missense , Adolescente , Oxidorreductasas de Alcohol/metabolismo , Alelos , Apoptosis , Ataxia/complicaciones , Ataxia/genética , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Niño , Preescolar , Cromatina/química , Proteínas de Unión al ADN/metabolismo , Femenino , Fibroblastos/metabolismo , Glioblastoma/genética , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Masculino , Hipotonía Muscular/complicaciones , Hipotonía Muscular/genética , Fenotipo , Unión Proteica , Proteómica , Anomalías Dentarias/complicaciones , Anomalías Dentarias/genética , Adulto Joven
15.
Am J Hum Genet ; 98(4): 772-81, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27040692

RESUMEN

Infantile encephalopathies are a group of clinically and biologically heterogeneous disorders for which the genetic basis remains largely unknown. Here, we report a syndromic neonatal encephalopathy characterized by profound developmental disability, severe hypotonia, seizures, diminished respiratory drive requiring mechanical ventilation, brain atrophy, dysgenesis of the corpus callosum, cerebellar vermis hypoplasia, and facial dysmorphism. Biallelic inactivating mutations in TBCK (TBC1-domain-containing kinase) were independently identified by whole-exome sequencing as the cause of this condition in four unrelated families. Matching these families was facilitated by the sharing of phenotypic profiles and WES data in a recently released web-based tool (Geno2MP) that links phenotypic information to rare variants in families with Mendelian traits. TBCK is a putative GTPase-activating protein (GAP) for small GTPases of the Rab family and has been shown to control cell growth and proliferation, actin-cytoskeleton dynamics, and mTOR signaling. Two of the three mutations (c.376C>T [p.Arg126(∗)] and c.1363A>T [p.Lys455(∗)]) are predicted to truncate the protein, and loss of the major TBCK isoform was confirmed in primary fibroblasts from one affected individual. The third mutation, c.1532G>A (p.Arg511His), alters a conserved residue within the TBC1 domain. Structural analysis implicated Arg511 as a required residue for Rab-GAP function, and in silico homology modeling predicted impaired GAP function in the corresponding mutant. These results suggest that loss of Rab-GAP activity is the underlying mechanism of disease. In contrast to other disorders caused by dysregulated mTOR signaling associated with focal or global brain overgrowth, impaired TBCK function results in progressive loss of brain volume.


Asunto(s)
Encefalopatías/genética , Proteínas Activadoras de GTPasa/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Alelos , Secuencia de Aminoácidos , Encefalopatías/diagnóstico , Niño , Preescolar , Cuerpo Calloso/patología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Conformación Proteica , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
16.
Am J Hum Genet ; 99(4): 802-816, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27616483

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) are ligand-gated cation channels that mediate excitatory synaptic transmission. Genetic mutations in multiple NMDAR subunits cause various childhood epilepsy syndromes. Here, we report a de novo recurrent heterozygous missense mutation-c.1999G>A (p.Val667Ile)-in a NMDAR gene previously unrecognized to harbor disease-causing mutations, GRIN2D, identified by exome and candidate panel sequencing in two unrelated children with epileptic encephalopathy. The resulting GluN2D p.Val667Ile exchange occurs in the M3 transmembrane domain involved in channel gating. This gain-of-function mutation increases glutamate and glycine potency by 2-fold, increases channel open probability by 6-fold, and reduces receptor sensitivity to endogenous negative modulators such as extracellular protons. Moreover, this mutation prolongs the deactivation time course after glutamate removal, which controls the synaptic time course. Transfection of cultured neurons with human GRIN2D cDNA harboring c.1999G>A leads to dendritic swelling and neuronal cell death, suggestive of excitotoxicity mediated by NMDAR over-activation. Because both individuals' seizures had proven refractory to conventional antiepileptic medications, the sensitivity of mutant NMDARs to FDA-approved NMDAR antagonists was evaluated. Based on these results, oral memantine was administered to both children, with resulting mild to moderate improvement in seizure burden and development. The older proband subsequently developed refractory status epilepticus, with dramatic electroclinical improvement upon treatment with ketamine and magnesium. Overall, these results suggest that NMDAR antagonists can be useful as adjuvant epilepsy therapy in individuals with GRIN2D gain-of-function mutations. This work further demonstrates the value of functionally evaluating a mutation, enabling mechanistic understanding and therapeutic modeling to realize precision medicine for epilepsy.


Asunto(s)
Genes Dominantes/genética , Mutación , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/genética , Secuencia de Aminoácidos , Secuencia de Bases , Muerte Celular , Niño , Análisis Mutacional de ADN , Dendritas/patología , Electroencefalografía , Exoma/genética , Femenino , Ácido Glutámico/metabolismo , Humanos , Lactante , Recién Nacido , Ketamina/uso terapéutico , Magnesio/uso terapéutico , Memantina/administración & dosificación , Memantina/uso terapéutico , Modelos Moleculares , Medicina de Precisión , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/genética , Convulsiones/metabolismo , Espasmos Infantiles/metabolismo
17.
Ann Neurol ; 83(1): 153-165, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29283439

RESUMEN

OBJECTIVE: Autosomal-recessive mutations in TBCK cause intellectual disability of variable severity. Although the physiological function of TBCK remains unclear, loss-of-function mutations are associated with inhibition of mechanistic target of rapamycin complex 1 (mTORC1) signaling. Given that mTORC1 signaling is known to regulate autophagy, we hypothesized that TBCK-encephalopathy patients with a neurodegenerative course have defects in autophagic-lysosomal dysfunction. METHODS: Children (n = 8) of Puerto Rican (Boricua) descent affected with homozygous TBCK p.R126X mutations underwent extensive neurological phenotyping and neurophysiological studies. We quantified autophagosome content in TBCK-/- patient-derived fibroblasts by immunostaining and assayed autophagic markers by western assay. Free sialylated oligosaccharide profiles were assayed in patient's urine and fibroblasts. RESULTS: The neurological phenotype of children with TBCK p.R126X mutations, which we call TBCK-encephaloneuronopathy (TBCKE), include congenital hypotonia, progressive motor neuronopathy, leukoencephalopathy, and epilepsy. Systemic features include coarse facies, dyslipidemia, and osteoporosis. TBCK-/- fibroblasts in vitro exhibit increased numbers of LC3+ autophagosomes and increased autophagic flux by immunoblots. Free oligosaccharide profiles in fibroblasts and urine of TBCKE patients differ from control fibroblasts and are ameliorated by treatment with the mTORC1 activator leucine. INTERPRETATION: TBCKE is a clinically distinguishable syndrome with progressive central and peripheral nervous system dysfunction, consistently observed in patients with the p.R126X mutation. We provide evidence that inappropriate autophagy in the absence of cellular stressors may play a role in this disorder, and that mTORC1 activation may ameliorate the autophagic-lysosomal system dysfunction. Free oligosaccharide profiles could serve as a novel biomarker for this disorder as well as a tool to evaluate potential therapeutic interventions. Ann Neurol 2018;83:153-165.


Asunto(s)
Autofagia/genética , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Biomarcadores/análisis , Niño , Exoma/genética , Fibroblastos , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Humanos , Discapacidad Intelectual , Leucina/uso terapéutico , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/agonistas , Diana Mecanicista del Complejo 1 de la Rapamicina/biosíntesis , Oligosacáridos/análisis , Fagosomas/patología , Fenotipo , Puerto Rico
18.
Hum Genet ; 137(5): 375-388, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29740699

RESUMEN

Many genetic causes of developmental delay and/or intellectual disability (DD/ID) are extremely rare, and robust discovery of these requires both large-scale DNA sequencing and data sharing. Here we describe a GeneMatcher collaboration which led to a cohort of 13 affected individuals harboring protein-altering variants, 11 of which are de novo, in MED13; the only inherited variant was transmitted to an affected child from an affected mother. All patients had intellectual disability and/or developmental delays, including speech delays or disorders. Other features that were reported in two or more patients include autism spectrum disorder, attention deficit hyperactivity disorder, optic nerve abnormalities, Duane anomaly, hypotonia, mild congenital heart abnormalities, and dysmorphisms. Six affected individuals had mutations that are predicted to truncate the MED13 protein, six had missense mutations, and one had an in-frame-deletion of one amino acid. Out of the seven non-truncating mutations, six clustered in two specific locations of the MED13 protein: an N-terminal and C-terminal region. The four N-terminal clustering mutations affect two adjacent amino acids that are known to be involved in MED13 ubiquitination and degradation, p.Thr326 and p.Pro327. MED13 is a component of the CDK8-kinase module that can reversibly bind Mediator, a multi-protein complex that is required for Polymerase II transcription initiation. Mutations in several other genes encoding subunits of Mediator have been previously shown to associate with DD/ID, including MED13L, a paralog of MED13. Thus, our findings add MED13 to the group of CDK8-kinase module-associated disease genes.


Asunto(s)
Secuencia de Aminoácidos , Complejo Mediador/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Eliminación de Secuencia , Adulto , Niño , Preescolar , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Femenino , Humanos , Masculino , Complejo Mediador/metabolismo , Trastornos del Neurodesarrollo/metabolismo , Iniciación de la Transcripción Genética , Ubiquitinación/genética , Reino Unido
19.
Am J Hum Genet ; 97(3): 457-64, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26299366

RESUMEN

Using whole-exome sequencing, we have identified in ten families 14 individuals with microcephaly, developmental delay, intellectual disability, hypotonia, spasticity, seizures, sensorineural hearing loss, cortical visual impairment, and rare autosomal-recessive predicted pathogenic variants in spermatogenesis-associated protein 5 (SPATA5). SPATA5 encodes a ubiquitously expressed member of the ATPase associated with diverse activities (AAA) protein family and is involved in mitochondrial morphogenesis during early spermatogenesis. It might also play a role in post-translational modification during cell differentiation in neuronal development. Mutations in SPATA5 might affect brain development and function, resulting in microcephaly, developmental delay, and intellectual disability.


Asunto(s)
Anomalías Múltiples/genética , Pérdida Auditiva/genética , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Convulsiones/genética , ATPasas Asociadas con Actividades Celulares Diversas , Anomalías Múltiples/patología , Secuencia de Aminoácidos , Secuencia de Bases , Exoma/genética , Femenino , Frecuencia de los Genes , Genes Recesivos , Humanos , Masculino , Datos de Secuencia Molecular , Mutación/genética , Alineación de Secuencia , Análisis de Secuencia de ADN
20.
Am J Med Genet A ; 176(12): 2710-2719, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30450772

RESUMEN

SOX2 is a transcription factor that is essential for maintenance of pluripotency and has several conserved roles in early embryonic development. Heterozygous loss-of-function variants in SOX2 are identified in approximately 40% of all cases of bilateral anophthalmia/micropthalmia (A/M). Increasingly SOX2 mutation-positive patients without major eye findings, but with a range of other developmental disorders including autism, mild to moderate intellectual disability with or without structural brain changes, esophageal atresia, urogenital anomalies, and endocrinopathy are being reported, suggesting that the clinical phenotype associated with SOX2 loss is much broader than previously appreciated. In this report we describe six new cases, four of which carry novel pathogenic SOX2 variants. Four cases presented with bilateral anophthalmia in addition to extraocular involvement. Another individual presented with only unilateral anophthalmia. One individual did not have any eye findings but presented with a suprasellar teratoma in infancy and was found to have the recurrent c.70del20 mutation in SOX2 (c.70_89del, p.Asn24Argfs*65). This is this first time this tumor type has been reported in the context of a de novo SOX2 mutation. Notably, individuals with hypothalamic hamartomas and slow-growing hypothalamo-pituitary tumors have been reported previously, but it is still unclear how SOX2 loss contributes to their formation.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Fenotipo , Factores de Transcripción SOXB1/genética , Biopsia , Encéfalo/anomalías , Encéfalo/diagnóstico por imagen , Preescolar , Consanguinidad , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Facies , Femenino , Humanos , Imagenología Tridimensional , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Análisis de Secuencia de ADN , Cráneo/anomalías , Cráneo/diagnóstico por imagen , Teratoma/diagnóstico , Teratoma/genética , Tomografía Computarizada por Rayos X , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA