Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
Am J Hum Genet ; 105(5): 1048-1056, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31668703

RESUMEN

NTNG2 encodes netrin-G2, a membrane-anchored protein implicated in the molecular organization of neuronal circuitry and synaptic organization and diversification in vertebrates. In this study, through a combination of exome sequencing and autozygosity mapping, we have identified 16 individuals (from seven unrelated families) with ultra-rare homozygous missense variants in NTNG2; these individuals present with shared features of a neurodevelopmental disorder consisting of global developmental delay, severe to profound intellectual disability, muscle weakness and abnormal tone, autistic features, behavioral abnormalities, and variable dysmorphisms. The variants disrupt highly conserved residues across the protein. Functional experiments, including in silico analysis of the protein structure, in vitro assessment of cell surface expression, and in vitro knockdown, revealed potential mechanisms of pathogenicity of the variants, including loss of protein function and decreased neurite outgrowth. Our data indicate that appropriate expression of NTNG2 plays an important role in neurotypical development.


Asunto(s)
Proteínas Ligadas a GPI/genética , Mutación Missense/genética , Netrinas/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Adulto , Niño , Preescolar , Exoma/genética , Femenino , Homocigoto , Humanos , Discapacidad Intelectual/genética , Masculino , Linaje , Secuenciación del Exoma/métodos , Adulto Joven
4.
Genet Med ; 21(3): 545-552, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30214071

RESUMEN

PURPOSE: Congenital microcephaly (CM) is an important birth defect with long term neurological sequelae. We aimed to perform detailed phenotypic and genomic analysis of patients with Mendelian forms of CM. METHODS: Clinical phenotyping, targeted or exome sequencing, and autozygome analysis. RESULTS: We describe 150 patients (104 families) with 56 Mendelian forms of CM. Our data show little overlap with the genetic causes of postnatal microcephaly. We also show that a broad definition of primary microcephaly -as an autosomal recessive form of nonsyndromic CM with severe postnatal deceleration of occipitofrontal circumference-is highly sensitive but has a limited specificity. In addition, we expand the overlap between primary microcephaly and microcephalic primordial dwarfism both clinically (short stature in >52% of patients with primary microcephaly) and molecularly (e.g., we report the first instance of CEP135-related microcephalic primordial dwarfism). We expand the allelic and locus heterogeneity of CM by reporting 37 novel likely disease-causing variants in 27 disease genes, confirming the candidacy of ANKLE2, YARS, FRMD4A, and THG1L, and proposing the candidacy of BPTF, MAP1B, CCNH, and PPFIBP1. CONCLUSION: Our study refines the phenotype of CM, expands its genetics heterogeneity, and informs the workup of children born with this developmental brain defect.


Asunto(s)
Microcefalia/genética , Microcefalia/fisiopatología , Adulto , Niño , Preescolar , Enanismo/genética , Femenino , Genómica/métodos , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Mutación/genética , Linaje , Fenotipo , Secuenciación del Exoma/métodos
5.
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
6.
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
7.
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
8.
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
9.
Eur J Med Genet ; 60(5): 245-249, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28254648

RESUMEN

Arthrogryposis multiplex congenital, the occurrence of multiple joint contractures at birth, can in some cases be accompanied by insufficient myelination of peripheral nerves, muscular hypotonia, reduced tendon reflexes, and respiratory insufficiency. Recently mutations in the CASPR/CNTN1 complex have been associated with similar severe phenotypes and CNTNAP1 gene mutations, causing loss of the CASPR protein, were shown to cause severe, prenatal onset arthrogryposis multiplex congenita in four unrelated families. Here we report a consanguineous Arab family from Qatar with three children having an early lethal form of arthrogryposis multiplex congenita and a novel frameshift mutation in CNTNAP1. We further expand the existing CNTNAP1-associated phenotype to include profound cerebral and cerebellar atrophy.


Asunto(s)
Artrogriposis/genética , Encéfalo/patología , Moléculas de Adhesión Celular Neuronal/genética , Mutación del Sistema de Lectura , Consanguinidad , Femenino , Humanos , Recién Nacido , Masculino , Linaje
10.
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
11.
Nature ; 539(7628): 242-247, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27830782

RESUMEN

Sensory stimuli drive the maturation and function of the mammalian nervous system in part through the activation of gene expression networks that regulate synapse development and plasticity. These networks have primarily been studied in mice, and it is not known whether there are species- or clade-specific activity-regulated genes that control features of brain development and function. Here we use transcriptional profiling of human fetal brain cultures to identify an activity-dependent secreted factor, Osteocrin (OSTN), that is induced by membrane depolarization of human but not mouse neurons. We find that OSTN has been repurposed in primates through the evolutionary acquisition of DNA regulatory elements that bind the activity-regulated transcription factor MEF2. In addition, we demonstrate that OSTN is expressed in primate neocortex and restricts activity-dependent dendritic growth in human neurons. These findings suggest that, in response to sensory input, OSTN regulates features of neuronal structure and function that are unique to primates.


Asunto(s)
Evolución Molecular , Proteínas Musculares/metabolismo , Neocórtex/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Animales , Secuencia de Bases , Huesos/metabolismo , Dendritas/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Humanos , Factores de Transcripción MEF2/metabolismo , Macaca mulatta , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/genética , Músculos/metabolismo , Neocórtex/citología , Neuronas/citología , Especificidad de Órganos , Especificidad de la Especie , Factores de Transcripción/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.
Elife ; 42015 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-26297806

RESUMEN

Primary microcephaly (MCPH) associated proteins CDK5RAP2, CEP152, WDR62 and CEP63 colocalize at the centrosome. We found that they interact to promote centriole duplication and form a hierarchy in which each is required to localize another to the centrosome, with CDK5RAP2 at the apex, and CEP152, WDR62 and CEP63 at sequentially lower positions. MCPH proteins interact with distinct centriolar satellite proteins; CDK5RAP2 interacts with SPAG5 and CEP72, CEP152 with CEP131, WDR62 with MOONRAKER, and CEP63 with CEP90 and CCDC14. These satellite proteins localize their cognate MCPH interactors to centrosomes and also promote centriole duplication. Consistent with a role for satellites in microcephaly, homozygous mutations in one satellite gene, CEP90, may cause MCPH. The satellite proteins, with the exception of CCDC14, and MCPH proteins promote centriole duplication by recruiting CDK2 to the centrosome. Thus, centriolar satellites build a MCPH complex critical for human neurodevelopment that promotes CDK2 centrosomal localization and centriole duplication.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Línea Celular , Humanos , Multimerización de Proteína
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.
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
16.
Neuron ; 84(6): 1240-57, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25521379

RESUMEN

Katanin is a microtubule-severing complex whose catalytic activities are well characterized, but whose in vivo functions are incompletely understood. Human mutations in KATNB1, which encodes the noncatalytic regulatory p80 subunit of katanin, cause severe microlissencephaly. Loss of Katnb1 in mice confirms essential roles in neurogenesis and cell survival, while loss of zebrafish katnb1 reveals specific roles for katnin p80 in early and late developmental stages. Surprisingly, Katnb1 null mutant mouse embryos display hallmarks of aberrant Sonic hedgehog signaling, including holoprosencephaly. KATNB1-deficient human cells show defective proliferation and spindle structure, while Katnb1 null fibroblasts also demonstrate a remarkable excess of centrioles, with supernumerary cilia but deficient Hedgehog signaling. Our results reveal unexpected functions for KATNB1 in regulating overall centriole, mother centriole, and cilia number, and as an essential gene for normal Hedgehog signaling during neocortical development.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Centriolos/fisiología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Cilios/fisiología , Adenosina Trifosfatasas/genética , Animales , Estudios de Casos y Controles , Proliferación Celular/genética , Proliferación Celular/fisiología , Centriolos/genética , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Cilios/genética , Embrión de Mamíferos , Desarrollo Embrionario/genética , Fibroblastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Katanina , Ratones , Microcefalia/genética , Mutación , Linaje , Empalme del ARN/genética , Población Blanca/genética , Pez Cebra
17.
N Engl J Med ; 371(8): 733-43, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25140959

RESUMEN

BACKGROUND: Although there is increasing recognition of the role of somatic mutations in genetic disorders, the prevalence of somatic mutations in neurodevelopmental disease and the optimal techniques to detect somatic mosaicism have not been systematically evaluated. METHODS: Using a customized panel of known and candidate genes associated with brain malformations, we applied targeted high-coverage sequencing (depth, ≥200×) to leukocyte-derived DNA samples from 158 persons with brain malformations, including the double-cortex syndrome (subcortical band heterotopia, 30 persons), polymicrogyria with megalencephaly (20), periventricular nodular heterotopia (61), and pachygyria (47). We validated candidate mutations with the use of Sanger sequencing and, for variants present at unequal read depths, subcloning followed by colony sequencing. RESULTS: Validated, causal mutations were found in 27 persons (17%; range, 10 to 30% for each phenotype). Mutations were somatic in 8 of the 27 (30%), predominantly in persons with the double-cortex syndrome (in whom we found mutations in DCX and LIS1), persons with periventricular nodular heterotopia (FLNA), and persons with pachygyria (TUBB2B). Of the somatic mutations we detected, 5 (63%) were undetectable with the use of traditional Sanger sequencing but were validated through subcloning and subsequent sequencing of the subcloned DNA. We found potentially causal mutations in the candidate genes DYNC1H1, KIF5C, and other kinesin genes in persons with pachygyria. CONCLUSIONS: Targeted sequencing was found to be useful for detecting somatic mutations in patients with brain malformations. High-coverage sequencing panels provide an important complement to whole-exome and whole-genome sequencing in the evaluation of somatic mutations in neuropsychiatric disease. (Funded by the National Institute of Neurological Disorders and Stroke and others.).


Asunto(s)
Corteza Cerebral/anomalías , Análisis Mutacional de ADN/métodos , Malformaciones del Desarrollo Cortical/genética , Mutación , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/genética , Humanos , Lisencefalia/genética , Imagen por Resonancia Magnética , Malformaciones del Desarrollo Cortical/patología , Heterotopia Nodular Periventricular/genética
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...