Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 57(20): 2381-2396.e13, 2022 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-36228617

RESUMEN

Kinesins are canonical molecular motors but can also function as modulators of intracellular signaling. KIF26A, an unconventional kinesin that lacks motor activity, inhibits growth-factor-receptor-bound protein 2 (GRB2)- and focal adhesion kinase (FAK)-dependent signal transduction, but its functions in the brain have not been characterized. We report a patient cohort with biallelic loss-of-function variants in KIF26A, exhibiting a spectrum of congenital brain malformations. In the developing brain, KIF26A is preferentially expressed during early- and mid-gestation in excitatory neurons. Combining mice and human iPSC-derived organoid models, we discovered that loss of KIF26A causes excitatory neuron-specific defects in radial migration, localization, dendritic and axonal growth, and apoptosis, offering a convincing explanation of the disease etiology in patients. Single-cell RNA sequencing in KIF26A knockout organoids revealed transcriptional changes in MAPK, MYC, and E2F pathways. Our findings illustrate the pathogenesis of KIF26A loss-of-function variants and identify the surprising versatility of this non-motor kinesin.


Asunto(s)
Cinesinas , Neuronas , Humanos , Animales , Ratones , Cinesinas/genética , Neuronas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Apoptosis , Encéfalo/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161264

RESUMEN

Osmotic equilibrium and membrane potential in animal cells depend on concentration gradients of sodium (Na+) and potassium (K+) ions across the plasma membrane, a function catalyzed by the Na+,K+-ATPase α-subunit. Here, we describe ATP1A3 variants encoding dysfunctional α3-subunits in children affected by polymicrogyria, a developmental malformation of the cerebral cortex characterized by abnormal folding and laminar organization. To gain cell-biological insights into the spatiotemporal dynamics of prenatal ATP1A3 expression, we built an ATP1A3 transcriptional atlas of fetal cortical development using mRNA in situ hybridization and transcriptomic profiling of ∼125,000 individual cells with single-cell RNA sequencing (Drop-seq) from 11 areas of the midgestational human neocortex. We found that fetal expression of ATP1A3 is most abundant to a subset of excitatory neurons carrying transcriptional signatures of the developing subplate, yet also maintains expression in nonneuronal cell populations. Moving forward a year in human development, we profiled ∼52,000 nuclei from four areas of an infant neocortex and show that ATP1A3 expression persists throughout early postnatal development, most predominantly in inhibitory neurons, including parvalbumin interneurons in the frontal cortex. Finally, we discovered the heteromeric Na+,K+-ATPase pump complex may form nonredundant cell-type-specific α-ß isoform combinations, including α3-ß1 in excitatory neurons and α3-ß2 in inhibitory neurons. Together, the developmental malformation phenotype of affected individuals and single-cell ATP1A3 expression patterns point to a key role for α3 in human cortex development, as well as a cell-type basis for pre- and postnatal ATP1A3-associated diseases.


Asunto(s)
Encéfalo/embriología , Encéfalo/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adulto , Encéfalo/anomalías , Encéfalo/diagnóstico por imagen , Niño , Femenino , Feto/embriología , Regulación del Desarrollo de la Expresión Génica , Humanos , Lactante , Recién Nacido , Interneuronas/metabolismo , Imagen por Resonancia Magnética , Masculino , Mutación/genética , Neocórtex/embriología , Neocórtex/enzimología , Neuronas/metabolismo , Parvalbúminas/metabolismo , Fenotipo , Polimicrogiria/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de la Célula Individual , ATPasa Intercambiadora de Sodio-Potasio/genética
3.
Genet Med ; 23(6): 1158-1162, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33531666

RESUMEN

PURPOSE: The endoplasmic reticulum membrane complex (EMC) is a highly conserved, multifunctional 10-protein complex related to membrane protein biology. In seven families, we identified 13 individuals with highly overlapping phenotypes who harbor a single identical homozygous frameshift variant in EMC10. METHODS: Using exome, genome, and Sanger sequencing, a recurrent frameshift EMC10 variant was identified in affected individuals in an international cohort of consanguineous families. Multiple families were independently identified and connected via Matchmaker Exchange and internal databases. We assessed the effect of the frameshift variant on EMC10 RNA and protein expression and evaluated EMC10 expression in normal human brain tissue using immunohistochemistry. RESULTS: A homozygous variant EMC10 c.287delG (Refseq NM_206538.3, p.Gly96Alafs*9) segregated with affected individuals in each family, who exhibited a phenotypic spectrum of intellectual disability (ID) and global developmental delay (GDD), variable seizures and variable dysmorphic features (elongated face, curly hair, cubitus valgus, and arachnodactyly). The variant arose on two founder haplotypes and results in significantly reduced EMC10 RNA expression and an unstable truncated EMC10 protein. CONCLUSION: We propose that a homozygous loss-of-function variant in EMC10 causes a novel syndromic neurodevelopmental phenotype. Remarkably, the recurrent variant is likely the result of a hypermutable site and arose on distinct founder haplotypes.


Asunto(s)
Discapacidades del Desarrollo , Discapacidad Intelectual , Niño , Discapacidades del Desarrollo/genética , Mutación del Sistema de Lectura , Homocigoto , Humanos , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Linaje , Fenotipo , Convulsiones/genética
4.
Sci Rep ; 10(1): 14045, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32820185

RESUMEN

More than 98% of the human genome is made up of non-coding DNA, but techniques to ascertain its contribution to human disease have lagged far behind our understanding of protein coding variations. Autism spectrum disorder (ASD) has been mostly associated with coding variations via de novo single nucleotide variants (SNVs), recessive/homozygous SNVs, or de novo copy number variants (CNVs); however, most ASD cases continue to lack a genetic diagnosis. We analyzed 187 consanguineous ASD families for biallelic CNVs. Recessive deletions were significantly enriched in affected individuals relative to their unaffected siblings (17% versus 4%, p < 0.001). Only a small subset of biallelic deletions were predicted to result in coding exon disruption. In contrast, biallelic deletions in individuals with ASD were enriched for overlap with regulatory regions, with 23/28 CNVs disrupting histone peaks in ENCODE (p < 0.009). Overlap with regulatory regions was further demonstrated by comparisons to the 127-epigenome dataset released by the Roadmap Epigenomics project, with enrichment for enhancers found in primary brain tissue and neuronal progenitor cells. Our results suggest a novel noncoding mechanism of ASD, describe a powerful method to identify important noncoding regions in the human genome, and emphasize the potential significance of gene activation and regulation in cognitive and social function.


Asunto(s)
Trastorno del Espectro Autista/genética , Epigénesis Genética , Eliminación de Gen , Homocigoto , Variaciones en el Número de Copia de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino
5.
Genet Med ; 22(6): 1040-1050, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32103185

RESUMEN

PURPOSE: The exocyst complex is a conserved protein complex that mediates fusion of intracellular vesicles to the plasma membrane and is implicated in processes including cell polarity, cell migration, ciliogenesis, cytokinesis, autophagy, and fusion of secretory vesicles. The essential role of these genes in human genetic disorders, however, is unknown. METHODS: We performed homozygosity mapping and exome sequencing of consanguineous families with recessively inherited brain development disorders. We modeled an EXOC7 splice variant in vitro and examined EXOC7 messenger RNA (mRNA) expression in developing mouse and human cortex. We modeled exoc7 loss-of-function in a zebrafish knockout. RESULTS: We report variants in exocyst complex members, EXOC7 and EXOC8, in a novel disorder of cerebral cortex development. In EXOC7, we identified four independent partial loss-of-function (LOF) variants in a recessively inherited disorder characterized by brain atrophy, seizures, and developmental delay, and in severe cases, microcephaly and infantile death. In EXOC8, we found a homozygous truncating variant in a family with a similar clinical disorder. We modeled exoc7 deficiency in zebrafish and found the absence of exoc7 causes microcephaly. CONCLUSION: Our results highlight the essential role of the exocyst pathway in normal cortical development and how its perturbation causes complex brain disorders.


Asunto(s)
Encefalopatías , Microcefalia , Animales , Proliferación Celular/genética , Homocigoto , Humanos , Ratones , Microcefalia/genética , Pez Cebra/genética
6.
Neuron ; 106(2): 246-255.e6, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32097629

RESUMEN

Genes mutated in human neuronal migration disorders encode tubulin proteins and a variety of tubulin-binding and -regulating proteins, but it is very poorly understood how these proteins function together to coordinate migration. Additionally, the way in which regional differences in neocortical migration are controlled is completely unknown. Here we describe a new syndrome with remarkably region-specific effects on neuronal migration in the posterior cortex, reflecting de novo variants in CEP85L. We show that CEP85L is required cell autonomously in vivo and in vitro for migration, that it localizes to the maternal centriole, and that it forms a complex with many other proteins required for migration, including CDK5, LIS1, NDE1, KIF2A, and DYNC1H1. Loss of CEP85L disrupts CDK5 localization and activation, leading to centrosome disorganization and disrupted microtubule cytoskeleton organization. Together, our findings suggest that CEP85L highlights a complex that controls CDK5 activity to promote neuronal migration.


Asunto(s)
Movimiento Celular , Quinasa 5 Dependiente de la Ciclina/genética , Proteínas del Citoesqueleto/genética , Lisencefalia/genética , Lisencefalia/patología , Neocórtex/patología , Neuronas/patología , Proteínas de Fusión Oncogénica/genética , Centriolos/genética , Niño , Preescolar , Femenino , Humanos , Masculino , Microtúbulos/genética , Microtúbulos/ultraestructura , Proteínas del Tejido Nervioso/fisiología , Adulto Joven
7.
Am J Med Genet B Neuropsychiatr Genet ; 177(8): 736-745, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30421579

RESUMEN

Protein homeostasis is tightly regulated by the ubiquitin proteasome pathway. Disruption of this pathway gives rise to a host of neurological disorders. Through whole exome sequencing (WES) in families with neurodevelopmental disorders, we identified mutations in PSMD12, a core component of the proteasome, underlying a neurodevelopmental disorder with intellectual disability (ID) and features of autism spectrum disorder (ASD). We performed WES on six affected siblings from a multiplex family with ID and autistic features, the affected father, and two unaffected mothers, and a trio from a simplex family with one affected child with ID and periventricular nodular heterotopia. We identified an inherited heterozygous nonsense mutation in PSMD12 (NM_002816: c.367C>T: p.R123X) in the multiplex family and a de novo nonsense mutation in the same gene (NM_002816: c.601C>T: p.R201X) in the simplex family. PSMD12 encodes a non-ATPase regulatory subunit of the 26S proteasome. We confirm the association of PSMD12 with ID, present the first cases of inherited PSMD12 mutation, and demonstrate the heterogeneity of phenotypes associated with PSMD12 mutations.


Asunto(s)
Discapacidad Intelectual/genética , Complejo de la Endopetidasa Proteasomal/genética , Adolescente , Adulto , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Niño , Preescolar , Familia , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia/genética , Humanos , Masculino , Mutación , Trastornos del Neurodesarrollo/genética , Linaje , Complejo de la Endopetidasa Proteasomal/metabolismo , Hermanos , Secuenciación del Exoma
8.
Neuron ; 99(5): 905-913.e7, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30146301

RESUMEN

Channelopathies are disorders caused by abnormal ion channel function in differentiated excitable tissues. We discovered a unique neurodevelopmental channelopathy resulting from pathogenic variants in SCN3A, a gene encoding the voltage-gated sodium channel NaV1.3. Pathogenic NaV1.3 channels showed altered biophysical properties including increased persistent current. Remarkably, affected individuals showed disrupted folding (polymicrogyria) of the perisylvian cortex of the brain but did not typically exhibit epilepsy; they presented with prominent speech and oral motor dysfunction, implicating SCN3A in prenatal development of human cortical language areas. The development of this disorder parallels SCN3A expression, which we observed to be highest early in fetal cortical development in progenitor cells of the outer subventricular zone and cortical plate neurons and decreased postnatally, when SCN1A (NaV1.1) expression increased. Disrupted cerebral cortical folding and neuronal migration were recapitulated in ferrets expressing the mutant channel, underscoring the unexpected role of SCN3A in progenitor cells and migrating neurons.


Asunto(s)
Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/crecimiento & desarrollo , Desarrollo del Lenguaje , Canal de Sodio Activado por Voltaje NAV1.3/genética , Canales de Sodio/genética , Adolescente , Adulto , Animales , Movimiento Celular/fisiología , Células Cultivadas , Corteza Cerebral/patología , Niño , Preescolar , Femenino , Hurones , Células HEK293 , Humanos , Lactante , Masculino , Megalencefalia/diagnóstico por imagen , Megalencefalia/genética , Megalencefalia/patología , Persona de Mediana Edad , Linaje , Polimicrogiria/diagnóstico por imagen , Polimicrogiria/genética , Polimicrogiria/patología
9.
Genome Res ; 27(8): 1323-1335, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28630177

RESUMEN

While next-generation sequencing has accelerated the discovery of human disease genes, progress has been largely limited to the "low hanging fruit" of mutations with obvious exonic coding or canonical splice site impact. In contrast, the lack of high-throughput, unbiased approaches for functional assessment of most noncoding variants has bottlenecked gene discovery. We report the integration of transcriptome sequencing (RNA-seq), which surveys all mRNAs to reveal functional impacts of variants at the transcription level, into the gene discovery framework for a unique human disease, microcephaly-micromelia syndrome (MMS). MMS is an autosomal recessive condition described thus far in only a single First Nations population and causes intrauterine growth restriction, severe microcephaly, craniofacial anomalies, skeletal dysplasia, and neonatal lethality. Linkage analysis of affected families, including a very large pedigree, identified a single locus on Chromosome 21 linked to the disease (LOD > 9). Comprehensive genome sequencing did not reveal any pathogenic coding or canonical splicing mutations within the linkage region but identified several nonconserved noncoding variants. RNA-seq analysis detected aberrant splicing in DONSON due to one of these noncoding variants, showing a causative role for DONSON disruption in MMS. We show that DONSON is expressed in progenitor cells of embryonic human brain and other proliferating tissues, is co-expressed with components of the DNA replication machinery, and that Donson is essential for early embryonic development in mice as well, suggesting an essential conserved role for DONSON in the cell cycle. Our results demonstrate the utility of integrating transcriptomics into the study of human genetic disease when DNA sequencing alone is not sufficient to reveal the underlying pathogenic mutation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN , Microcefalia/genética , Microcefalia/patología , Mutación , Proteínas Nucleares/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Transcriptoma , Animales , Mapeo Cromosómico , Femenino , Ligamiento Genético , Inestabilidad Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Noqueados , Microcefalia/etiología , Osteocondrodisplasias/etiología , Linaje , Embarazo , Empalme del ARN , Análisis de Secuencia de ARN , Secuenciación Completa del Genoma
10.
Hum Mutat ; 38(10): 1348-1354, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28493438

RESUMEN

Aminoacyl-transfer RNA (tRNA) synthetases ligate amino acids to specific tRNAs and are essential for protein synthesis. Although alanyl-tRNA synthetase (AARS) is a synthetase implicated in a wide range of neurological disorders from Charcot-Marie-Tooth disease to infantile epileptic encephalopathy, there have been limited data on their pathogenesis. Here, we report loss-of-function mutations in AARS in two siblings with progressive microcephaly with hypomyelination, intractable epilepsy, and spasticity. Whole-exome sequencing identified that the affected individuals were compound heterozygous for mutations in AARS gene, c.2067dupC (p.Tyr690Leufs*3) and c.2738G>A (p.Gly913Asp). A lymphoblastoid cell line developed from one of the affected individuals showed a strong reduction in AARS abundance. The mutations decrease aminoacylation efficiency by 70%-90%. The p.Tyr690Leufs*3 mutation also abolished editing activity required for hydrolyzing misacylated tRNAs, thereby increasing errors during aminoacylation. Our study has extended potential mechanisms underlying AARS-related disorders to include destabilization of the protein, aminoacylation dysfunction, and defective editing activity.


Asunto(s)
Alanina-ARNt Ligasa/genética , Síndrome de Lennox-Gastaut/genética , Microcefalia/genética , Espasmos Infantiles/genética , Paraplejía Espástica Hereditaria/genética , Secuencia de Aminoácidos/genética , Aminoacilación/genética , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Preescolar , Electroencefalografía , Femenino , Humanos , Lactante , Síndrome de Lennox-Gastaut/complicaciones , Síndrome de Lennox-Gastaut/diagnóstico , Síndrome de Lennox-Gastaut/patología , Microcefalia/diagnóstico por imagen , Microcefalia/patología , Mutación/genética , Biosíntesis de Proteínas/genética , Hermanos , Espasmos Infantiles/complicaciones , Espasmos Infantiles/diagnóstico por imagen , Espasmos Infantiles/patología , Paraplejía Espástica Hereditaria/complicaciones , Paraplejía Espástica Hereditaria/patología , Secuenciación del Exoma
11.
Am J Hum Genet ; 100(3): 537-545, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28190459

RESUMEN

Congenital muscular dystrophies display a wide phenotypic and genetic heterogeneity. The combination of clinical, biochemical, and molecular genetic findings must be considered to obtain the precise diagnosis and provide appropriate genetic counselling. Here we report five individuals from four families presenting with variable clinical features including muscular dystrophy with a reduction in dystroglycan glycosylation, short stature, intellectual disability, and cataracts, overlapping both the dystroglycanopathies and Marinesco-Sjögren syndrome. Whole-exome sequencing revealed homozygous missense and compound heterozygous mutations in INPP5K in the affected members of each family. INPP5K encodes the inositol polyphosphate-5-phosphatase K, also known as SKIP (skeletal muscle and kidney enriched inositol phosphatase), which is highly expressed in the brain and muscle. INPP5K localizes to both the endoplasmic reticulum and to actin ruffles in the cytoplasm. It has been shown to regulate myoblast differentiation and has also been implicated in protein processing through its interaction with the ER chaperone HSPA5/BiP. We show that morpholino-mediated inpp5k loss of function in the zebrafish results in shortened body axis, microphthalmia with disorganized lens, microcephaly, reduced touch-evoked motility, and highly disorganized myofibers. Altogether these data demonstrate that mutations in INPP5K cause a congenital muscular dystrophy syndrome with short stature, cataracts, and intellectual disability.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Monoéster Fosfórico Hidrolasas/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Niño , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Estudio de Asociación del Genoma Completo , Glicosilación , Trastornos del Crecimiento/genética , Humanos , Discapacidad Intelectual/genética , Masculino , Microcefalia/genética , Músculo Esquelético/metabolismo , Mutación , Linaje , Adulto Joven , Pez Cebra/genética
12.
Proc Natl Acad Sci U S A ; 113(38): E5598-607, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27601654

RESUMEN

Mutations that cause neurological phenotypes are highly informative with regard to mechanisms governing human brain function and disease. We report autosomal recessive mutations in the enzyme glutamate pyruvate transaminase 2 (GPT2) in large kindreds initially ascertained for intellectual and developmental disability (IDD). GPT2 [also known as alanine transaminase 2 (ALT2)] is one of two related transaminases that catalyze the reversible addition of an amino group from glutamate to pyruvate, yielding alanine and α-ketoglutarate. In addition to IDD, all affected individuals show postnatal microcephaly and ∼80% of those followed over time show progressive motor symptoms, a spastic paraplegia. Homozygous nonsense p.Arg404* and missense p.Pro272Leu mutations are shown biochemically to be loss of function. The GPT2 gene demonstrates increasing expression in brain in the early postnatal period, and GPT2 protein localizes to mitochondria. Akin to the human phenotype, Gpt2-null mice exhibit reduced brain growth. Through metabolomics and direct isotope tracing experiments, we find a number of metabolic abnormalities associated with loss of Gpt2. These include defects in amino acid metabolism such as low alanine levels and elevated essential amino acids. Also, we find defects in anaplerosis, the metabolic process involved in replenishing TCA cycle intermediates. Finally, mutant brains demonstrate misregulated metabolites in pathways implicated in neuroprotective mechanisms previously associated with neurodegenerative disorders. Overall, our data reveal an important role for the GPT2 enzyme in mitochondrial metabolism with relevance to developmental as well as potentially to neurodegenerative mechanisms.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Mitocondrias/enzimología , Enfermedades del Sistema Nervioso/genética , Transaminasas/genética , Secuencia de Aminoácidos/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Ciclo del Ácido Cítrico/genética , Homocigoto , Humanos , Ácidos Cetoglutáricos/metabolismo , Ratones , Mitocondrias/patología , Mutación Missense , Enfermedades del Sistema Nervioso/patología , Fenotipo , Ácido Pirúvico/metabolismo , Transaminasas/metabolismo
13.
Am J Hum Genet ; 96(5): 709-19, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25865492

RESUMEN

Despite recent advances in understanding the genetic bases of microcephaly, a large number of cases of microcephaly remain unexplained, suggesting that many microcephaly syndromes and associated genes have yet to be identified. Here, we report mutations in PYCR2, which encodes an enzyme in the proline biosynthesis pathway, as the cause of a unique syndrome characterized by postnatal microcephaly, hypomyelination, and reduced cerebral white-matter volume. Linkage mapping and whole-exome sequencing identified homozygous mutations (c.355C>T [p.Arg119Cys] and c.751C>T [p.Arg251Cys]) in PYCR2 in the affected individuals of two consanguineous families. A lymphoblastoid cell line from one affected individual showed a strong reduction in the amount of PYCR2. When mutant cDNAs were transfected into HEK293FT cells, both variant proteins retained normal mitochondrial localization but had lower amounts than the wild-type protein, suggesting that the variant proteins were less stable. A PYCR2-deficient HEK293FT cell line generated by genome editing with the clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system showed that PYCR2 loss of function led to decreased mitochondrial membrane potential and increased susceptibility to apoptosis under oxidative stress. Morpholino-based knockdown of a zebrafish PYCR2 ortholog, pycr1b, recapitulated the human microcephaly phenotype, which was rescued by wild-type human PYCR2 mRNA, but not by mutant mRNAs, further supporting the pathogenicity of the identified variants. Hypomyelination and the absence of lax, wrinkly skin distinguishes this condition from that caused by previously reported mutations in the gene encoding PYCR2's isozyme, PYCR1, suggesting a unique and indispensable role for PYCR2 in the human CNS during development.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/deficiencia , Antiportadores/deficiencia , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Microcefalia/genética , Enfermedades Mitocondriales/genética , Trastornos Psicomotores/genética , Pirrolina Carboxilato Reductasas/genética , Sistemas de Transporte de Aminoácidos Acídicos/genética , Antiportadores/genética , Femenino , Genotipo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Homocigoto , Humanos , Masculino , Microcefalia/patología , Enfermedades Mitocondriales/patología , Mutación , Fenotipo , Trastornos Psicomotores/patología , delta-1-Pirrolina-5-Carboxilato Reductasa
14.
Neurology ; 84(17): 1745-50, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25832664

RESUMEN

OBJECTIVE: To identify the genetic cause of pontocerebellar hypoplasia type III (PCH3). METHODS: We studied the original reported pedigree of PCH3 and performed genetic analysis including genome-wide single nucleotide polymorphism genotyping, linkage analysis, whole-exome sequencing, and Sanger sequencing. Human fetal brain RNA sequencing data were then analyzed for the identified candidate gene. RESULTS: The affected individuals presented with severe global developmental delay and seizures starting in the first year of life. Brain MRI of an affected individual showed diffuse atrophy of the cerebrum, cerebellum, and brainstem. Genome-wide single nucleotide polymorphism analysis confirmed the linkage to chromosome 7q we previously reported, and showed no other genomic areas of linkage. Whole-exome sequencing of 2 affected individuals identified a shared homozygous, nonsense variant in the PCLO (piccolo) gene. This variant segregated with the disease phenotype in the pedigree was rare in the population and was predicted to eliminate the PDZ and C2 domains in the C-terminus of the protein. RNA sequencing data of human fetal brain showed that PCLO was moderately expressed in the developing cerebral cortex. CONCLUSIONS: Here, we show that a homozygous, nonsense PCLO mutation underlies the autosomal recessive neurodegenerative disorder, PCH3. PCLO is a component of the presynaptic cytoskeletal matrix, and is thought to be involved in regulation of presynaptic proteins and synaptic vesicles. Our findings suggest that PCLO is crucial for the development and survival of a wide range of neuronal types in the human brain.


Asunto(s)
Codón sin Sentido/genética , Proteínas del Citoesqueleto/genética , Neuropéptidos/genética , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/patología , Enfermedades Cerebelosas/fisiopatología , Niño , Consanguinidad , Exoma , Ligamiento Genético , Humanos , Omán , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ARN
15.
Nature ; 515(7526): 209-15, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25363760

RESUMEN

The genetic architecture of autism spectrum disorder involves the interplay of common and rare variants and their impact on hundreds of genes. Using exome sequencing, here we show that analysis of rare coding variation in 3,871 autism cases and 9,937 ancestry-matched or parental controls implicates 22 autosomal genes at a false discovery rate (FDR) < 0.05, plus a set of 107 autosomal genes strongly enriched for those likely to affect risk (FDR < 0.30). These 107 genes, which show unusual evolutionary constraint against mutations, incur de novo loss-of-function mutations in over 5% of autistic subjects. Many of the genes implicated encode proteins for synaptic formation, transcriptional regulation and chromatin-remodelling pathways. These include voltage-gated ion channels regulating the propagation of action potentials, pacemaking and excitability-transcription coupling, as well as histone-modifying enzymes and chromatin remodellers-most prominently those that mediate post-translational lysine methylation/demethylation modifications of histones.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Cromatina/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Sinapsis/metabolismo , Transcripción Genética/genética , Secuencia de Aminoácidos , Trastornos Generalizados del Desarrollo Infantil/patología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Exoma/genética , Femenino , Mutación de Línea Germinal/genética , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense/genética , Red Nerviosa/metabolismo , Oportunidad Relativa
16.
Cell Rep ; 8(3): 647-55, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25066123

RESUMEN

Autism spectrum disorder (ASD) and intellectual disability (ID) are often comorbid, but the extent to which they share common genetic causes remains controversial. Here, we present two autosomal-recessive "founder" mutations in the CC2D1A gene causing fully penetrant cognitive phenotypes, including mild-to-severe ID, ASD, as well as seizures, suggesting shared developmental mechanisms. CC2D1A regulates multiple intracellular signaling pathways, and we found its strongest effect to be on the transcription factor nuclear factor κB (NF-κB). Cc2d1a gain and loss of function both increase activation of NF-κB, revealing a critical role of Cc2d1a in homeostatic control of intracellular signaling. Cc2d1a knockdown in neurons reduces dendritic complexity and increases NF-κB activity, and the effects of Cc2d1a depletion can be rescued by inhibiting NF-κB activity. Homeostatic regulation of neuronal signaling pathways provides a mechanism whereby common founder mutations could manifest diverse symptoms in different patients.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Proteínas de Unión al ADN/metabolismo , Discapacidad Intelectual/genética , FN-kappa B/metabolismo , Neuronas/metabolismo , Convulsiones/genética , Animales , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Proteínas de Unión al ADN/genética , Homeostasis , Humanos , Discapacidad Intelectual/metabolismo , Ratones , Mutación , Neuronas/citología , Linaje , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Convulsiones/metabolismo , Transducción de Señal
17.
Am J Hum Genet ; 94(4): 547-58, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24656866

RESUMEN

Progressive microcephaly is a heterogeneous condition with causes including mutations in genes encoding regulators of neuronal survival. Here, we report the identification of mutations in QARS (encoding glutaminyl-tRNA synthetase [QARS]) as the causative variants in two unrelated families affected by progressive microcephaly, severe seizures in infancy, atrophy of the cerebral cortex and cerebellar vermis, and mild atrophy of the cerebellar hemispheres. Whole-exome sequencing of individuals from each family independently identified compound-heterozygous mutations in QARS as the only candidate causative variants. QARS was highly expressed in the developing fetal human cerebral cortex in many cell types. The four QARS mutations altered highly conserved amino acids, and the aminoacylation activity of QARS was significantly impaired in mutant cell lines. Variants p.Gly45Val and p.Tyr57His were located in the N-terminal domain required for QARS interaction with proteins in the multisynthetase complex and potentially with glutamine tRNA, and recombinant QARS proteins bearing either substitution showed an over 10-fold reduction in aminoacylation activity. Conversely, variants p.Arg403Trp and p.Arg515Trp, each occurring in a different family, were located in the catalytic core and completely disrupted QARS aminoacylation activity in vitro. Furthermore, p.Arg403Trp and p.Arg515Trp rendered QARS less soluble, and p.Arg403Trp disrupted QARS-RARS (arginyl-tRNA synthetase 1) interaction. In zebrafish, homozygous qars loss of function caused decreased brain and eye size and extensive cell death in the brain. Our results highlight the importance of QARS during brain development and that epilepsy due to impairment of QARS activity is unusually severe in comparison to other aminoacyl-tRNA synthetase disorders.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Encefalopatías/genética , Predisposición Genética a la Enfermedad , Microcefalia/genética , Mutación , Convulsiones/genética , Aminoacilación , Animales , Preescolar , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Microcefalia/patología , Linaje , Pez Cebra
18.
Hum Mol Genet ; 23(13): 3456-66, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24501276

RESUMEN

Whereas many genes associated with intellectual disability (ID) encode synaptic proteins, transcriptional defects leading to ID are less well understood. We studied a large, consanguineous pedigree of Arab origin with seven members affected with ID and mild dysmorphic features. Homozygosity mapping and linkage analysis identified a candidate region on chromosome 17 with a maximum multipoint logarithm of odds score of 6.01. Targeted high-throughput sequencing of the exons in the candidate region identified a homozygous 4-bp deletion (c.169_172delCACT) in the METTL23 (methyltransferase like 23) gene, which is predicted to result in a frameshift and premature truncation (p.His57Valfs*11). Overexpressed METTL23 protein localized to both nucleus and cytoplasm, and physically interacted with GABPA (GA-binding protein transcription factor, alpha subunit). GABP, of which GABPA is a component, is known to regulate the expression of genes such as THPO (thrombopoietin) and ATP5B (ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide) and is implicated in a wide variety of important cellular functions. Overexpression of METTL23 resulted in increased transcriptional activity at the THPO promoter, whereas knockdown of METTL23 with siRNA resulted in decreased expression of ATP5B, thus revealing the importance of METTL23 as a regulator of GABPA function. The METTL23 mutation highlights a new transcriptional pathway underlying human intellectual function.


Asunto(s)
Metilasas de Modificación del ADN/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Metilasas de Modificación del ADN/genética , Femenino , Factor de Transcripción de la Proteína de Unión a GA/genética , Genotipo , Humanos , Inmunoprecipitación , Masculino , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Polimorfismo de Nucleótido Simple/genética , Unión Proteica , ARN Interferente Pequeño , Trombopoyetina/genética , Trombopoyetina/metabolismo , Técnicas del Sistema de Dos Híbridos
19.
Neurology ; 81(16): 1378-86, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24078737

RESUMEN

OBJECTIVE: To identify the genetic cause of a syndrome causing cerebellar ataxia and eye movement abnormalities. METHODS: We identified 2 families with cerebellar ataxia, eye movement abnormalities, and global developmental delay. We performed genetic analyses including single nucleotide polymorphism genotyping, linkage analysis, array comparative genomic hybridization, quantitative PCR, and Sanger sequencing. We obtained eye movement recordings of mutant mice deficient for the ortholog of the identified candidate gene, and performed immunohistochemistry using human and mouse brain specimens. RESULTS: All affected individuals had ataxia, eye movement abnormalities, most notably tonic upgaze, and delayed speech and cognitive development. Homozygosity mapping identified the disease locus on chromosome 4q. Within this region, a homozygous deletion of GRID2 exon 4 in the index family and compound heterozygous deletions involving GRID2 exon 2 in the second family were identified. Grid2-deficient mice showed larger spontaneous and random eye movements compared to wild-type mice. In developing mouse and human cerebella, GRID2 localized to the Purkinje cell dendritic spines. Brain MRI in 2 affected children showed progressive cerebellar atrophy, which was more severe than that of Grid2-deficient mice. CONCLUSIONS: Biallelic deletions of GRID2 lead to a syndrome of cerebellar ataxia and tonic upgaze in humans. The phenotypic resemblance and similarity in protein expression pattern between humans and mice suggest a conserved role for GRID2 in the synapse organization between parallel fibers and Purkinje cells. However, the progressive and severe cerebellar atrophy seen in the affected individuals could indicate an evolutionarily unique role for GRID2 in the human cerebellum.


Asunto(s)
Ataxia Cerebelosa/genética , Trastornos de la Motilidad Ocular/genética , Receptores de Glutamato/genética , Adolescente , Animales , Niño , Preescolar , Exones/genética , Femenino , Genes Recesivos/genética , Humanos , Masculino , Ratones , Eliminación de Secuencia/genética , Síndrome
20.
Neuron ; 77(2): 259-73, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23352163

RESUMEN

Despite significant heritability of autism spectrum disorders (ASDs), their extreme genetic heterogeneity has proven challenging for gene discovery. Studies of primarily simplex families have implicated de novo copy number changes and point mutations, but are not optimally designed to identify inherited risk alleles. We apply whole-exome sequencing (WES) to ASD families enriched for inherited causes due to consanguinity and find familial ASD associated with biallelic mutations in disease genes (AMT, PEX7, SYNE1, VPS13B, PAH, and POMGNT1). At least some of these genes show biallelic mutations in nonconsanguineous families as well. These mutations are often only partially disabling or present atypically, with patients lacking diagnostic features of the Mendelian disorders with which these genes are classically associated. Our study shows the utility of WES for identifying specific genetic conditions not clinically suspected and the importance of partial loss of gene function in ASDs.


Asunto(s)
Trastorno Autístico/diagnóstico , Trastorno Autístico/genética , Exoma/genética , Estudio de Asociación del Genoma Completo/métodos , Adolescente , Animales , Células Cultivadas , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Linaje , Ratas , Análisis de Secuencia de ADN/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...