Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Eur J Hum Genet ; 31(11): 1251-1260, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37644171

RESUMEN

Heterozygous, pathogenic CUX1 variants are associated with global developmental delay or intellectual disability. This study delineates the clinical presentation in an extended cohort and investigates the molecular mechanism underlying the disorder in a Cux1+/- mouse model. Through international collaboration, we assembled the phenotypic and molecular information for 34 individuals (23 unpublished individuals). We analyze brain CUX1 expression and susceptibility to epilepsy in Cux1+/- mice. We describe 34 individuals, from which 30 were unrelated, with 26 different null and four missense variants. The leading symptoms were mild to moderate delayed speech and motor development and borderline to moderate intellectual disability. Additional symptoms were muscular hypotonia, seizures, joint laxity, and abnormalities of the forehead. In Cux1+/- mice, we found delayed growth, histologically normal brains, and increased susceptibility to seizures. In Cux1+/- brains, the expression of Cux1 transcripts was half of WT animals. Expression of CUX1 proteins was reduced, although in early postnatal animals significantly more than in adults. In summary, disease-causing CUX1 variants result in a non-syndromic phenotype of developmental delay and intellectual disability. In some individuals, this phenotype ameliorates with age, resulting in a clinical catch-up and normal IQ in adulthood. The post-transcriptional balance of CUX1 expression in the heterozygous brain at late developmental stages appears important for this favorable clinical course.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Adulto , Animales , Humanos , Ratones , Heterocigoto , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Proteínas Represoras/genética , Convulsiones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
J Clin Invest ; 133(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36976648

RESUMEN

Neural differentiation, synaptic transmission, and action potential propagation depend on membrane sphingolipids, whose metabolism is tightly regulated. Mutations in the ceramide transporter CERT (CERT1), which is involved in sphingolipid biosynthesis, are associated with intellectual disability, but the pathogenic mechanism remains obscure. Here, we characterize 31 individuals with de novo missense variants in CERT1. Several variants fall into a previously uncharacterized dimeric helical domain that enables CERT homeostatic inactivation, without which sphingolipid production goes unchecked. The clinical severity reflects the degree to which CERT autoregulation is disrupted, and inhibiting CERT pharmacologically corrects morphological and motor abnormalities in a Drosophila model of the disease, which we call ceramide transporter (CerTra) syndrome. These findings uncover a central role for CERT autoregulation in the control of sphingolipid biosynthetic flux, provide unexpected insight into the structural organization of CERT, and suggest a possible therapeutic approach for patients with CerTra syndrome.


Asunto(s)
Ceramidas , Esfingolípidos , Humanos , Ceramidas/metabolismo , Homeostasis , Mutación , Esfingolípidos/genética , Esfingolípidos/metabolismo
4.
Clin Genet ; 102(3): 191-200, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35699227

RESUMEN

The study describes all patients in Denmark with vascular Ehlers-Danlos syndrome (vEDS). Carriers of pathogenic or likely pathogenic COL3A1 variants were retrospectively identified through registries and specialized clinics. Medical records were reviewed for vascular- or organ ruptures and invasive procedures performed. Identified families were divided by variant type (null, splice, and missense) and familial phenotypes (severe or attenuated). Families in which at least one carrier has suffered a major event before the age of 30 were classified as severe, whereas families in which at least three carriers had reached the age of 40 without a major event were classified as attenuated. Eighty-seven persons (59 still alive) from 25 families were included with a mean observation time of 44 years. Sixty-seven percent of patients could be subclassified in a familial phenotype. Thirty-one major events were observed. Eleven complications in 172 invasive procedures were recorded. No fatal complications to elective surgery were observed. The type of COL3A1 variant did not reliably predict phenotype, but a pattern of intrafamilial consistency emerged with some families showing an attenuated form of vEDS. Elective medical procedures appear to be safer than previously thought, although data only allow for conclusions regarding individuals from families with the attenuated form of vEDS.


Asunto(s)
Colágeno Tipo III , Síndrome de Ehlers-Danlos , Colágeno Tipo III/genética , Dinamarca/epidemiología , Síndrome de Ehlers-Danlos/genética , Procedimientos Quirúrgicos Electivos , Humanos , Estudios Retrospectivos
5.
Sci Rep ; 12(1): 902, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35042901

RESUMEN

Shank proteins are major scaffolds of the postsynaptic density of excitatory synapses. Mutations in SHANK genes are associated with autism and intellectual disability. The effects of missense mutations on Shank3 function, and therefore the pathomechanisms are unclear. Several missense mutations in SHANK3 affect the N-terminal region, consisting of the Shank/ProSAP N-terminal (SPN) domain and a set of Ankyrin (Ank) repeats. Here we identify a novel SHANK3 missense mutation (p.L270M) in the Ankyrin repeats in patients with an ADHD-like phenotype. We functionally analysed this and a series of other mutations, using biochemical and biophysical techniques. We observe two major effects: (1) a loss of binding to δ-catenin (e.g. in the p.L270M variant), and (2) interference with the intramolecular interaction between N-terminal SPN domain and the Ank repeats. This also interferes with binding to the α-subunit of the calcium-/calmodulin dependent kinase II (αCaMKII), and appears to be associated with a more severe neurodevelopmental pathology.


Asunto(s)
Sinapsis
6.
Clin Genet ; 101(2): 208-213, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34708403

RESUMEN

The YTH domain family member 3 gene (YTHDF3) encodes a reader of the abundant N6-methyladenosine (m6 A) modification of eukaryotic mRNA, which plays an essential role in regulating mRNA stability and is necessary to achieve normal development of the central nervous system in animal models. YTHDF3 has not previously been implicated in Mendelian disease despite a high probability of loss of function intolerance and statistical evidence of enrichment for gene-disruptive de novo variants in large-scale studies of individuals with intellectual disability and/or developmental delay. We report four individuals with deletion of 8q12.3, deletion size 1.38-2.60 Mb, encompassing YTHDF3, three of them were de novo, and in one case, the inheritance was unknown. Common features of the individuals (age range, 4-22 years) were developmental delay and/or intellectual disability. Two individuals underwent squint surgery. We suggest that haploinsufficiency of YTHDF3 causes a neurodevelopmental disorder with developmental delay and intellectual disability of variable degree.


Asunto(s)
Alelos , Deleción Cromosómica , Cromosomas Humanos Par 8 , Predisposición Genética a la Enfermedad , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Proteínas de Unión al ARN/genética , Adolescente , Niño , Femenino , Estudios de Asociación Genética , Humanos , Pérdida de Heterocigocidad , Masculino , Fenotipo , Adulto Joven
7.
Eur J Med Genet ; 64(9): 104280, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34229113

RESUMEN

Xia-Gibbs syndrome (XGS) is a neurodevelopmental disorder characterized by intellectual disability, developmental delay, seizures, hypotonia, obstructive sleep apnoea and mild facial dysmorphism. Heterozygosity for loss-of-function variants in AHDC1, encoding the AT-hook DNA binding motif containing protein 1, were discovered in 2014 as the likely genetic cause of Xia-Gibbs syndrome. We present five patients with Xia-Gibbs syndrome caused by previously unreported variants in AHDC1. Two of the patients share a frameshift variant: c.2849del (p.(Pro950Argfs*192)) in AHDC1. Despite sharing this variant, the two patients show remarkable phenotypic differences underscoring the clinical heterogeneity of Xia-Gibbs syndrome. In addition, we present a case of Xia-Gibbs syndrome caused by mosaicism for an AHDC1 variant.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Deformidades del Pie/genética , Hipotonía Muscular/genética , Fenotipo , Adolescente , Adulto , Anomalías Craneofaciales/patología , Discapacidades del Desarrollo/patología , Femenino , Deformidades del Pie/patología , Mutación del Sistema de Lectura , Humanos , Masculino , Hipotonía Muscular/patología , Síndrome , Adulto Joven
8.
Clin Genet ; 100(5): 607-614, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34296759

RESUMEN

Early infantile epileptic encephalopathy 38 (EIEE38, MIM #617020) is caused by biallelic variants in ARV1, encoding a transmembrane protein of the endoplasmic reticulum with a pivotal role in glycosylphosphatidylinositol (GPI) biosynthesis. We ascertained seven new patients from six unrelated families harboring biallelic variants in ARV1, including five novel variants. Affected individuals showed psychomotor delay, hypotonia, early onset refractory seizures followed by regression and specific neuroimaging features. Flow cytometric analysis on patient fibroblasts showed a decrease in GPI-anchored proteins on the cell surface, supporting a lower residual activity of the mutant ARV1 as compared to the wildtype. A rescue assay through the transduction of lentivirus expressing wild type ARV1 cDNA effectively rescued these alterations. This study expands the clinical and molecular spectrum of the ARV1-related encephalopathy, confirming the essential role of ARV1 in GPI biosynthesis and brain function.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/deficiencia , Fenotipo , Espasmos Infantiles/diagnóstico , Espasmos Infantiles/genética , Alelos , Sustitución de Aminoácidos , Encéfalo/anomalías , Proteínas Portadoras/genética , Análisis Mutacional de ADN , Facies , Femenino , Proteínas Ligadas a GPI/biosíntesis , Estudios de Asociación Genética/métodos , Glicosilfosfatidilinositoles/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/genética , Mutación , Linaje , Embarazo , Diagnóstico Prenatal/métodos , Espasmos Infantiles/metabolismo
9.
Am J Med Genet A ; 185(12): 3740-3753, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34331327

RESUMEN

Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome is caused by de novo loss-of-function variants in the SON gene (MIM #617140). This multisystemic disorder is characterized by intellectual disability, seizures, abnormal brain imaging, variable dysmorphic features, and various congenital anomalies. The wide application and increasing accessibility of whole exome sequencing (WES) has helped to identify new cases of ZTTK syndrome over the last few years. To date, there have been approximately 45 cases reported in the literature. Here, we describe 15 additional individuals with variants in the SON gene, including those with missense variants bringing the total number of known cases to 60. We have reviewed the clinical and molecular data of these new cases and all previously reported cases to further delineate the most common as well as emerging clinical findings related to this syndrome. Furthermore, we aim to delineate any genotype-phenotype correlations specifically for a recurring pathogenic four base pair deletion (c.5753_5756del) along with discussing the impact of missense variants seen in the SON gene.


Asunto(s)
Anomalías Congénitas/genética , Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Antígenos de Histocompatibilidad Menor/genética , Convulsiones/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/patología , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Mutación Missense/genética , Fenotipo , Convulsiones/diagnóstico , Convulsiones/patología , Secuenciación del Exoma
10.
Genet Med ; 23(6): 1028-1040, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33658631

RESUMEN

PURPOSE: We describe a novel neurobehavioral phenotype of autism spectrum disorder (ASD), intellectual disability, and/or attention-deficit/hyperactivity disorder (ADHD) associated with de novo or inherited deleterious variants in members of the RFX family of genes. RFX genes are evolutionarily conserved transcription factors that act as master regulators of central nervous system development and ciliogenesis. METHODS: We assembled a cohort of 38 individuals (from 33 unrelated families) with de novo variants in RFX3, RFX4, and RFX7. We describe their common clinical phenotypes and present bioinformatic analyses of expression patterns and downstream targets of these genes as they relate to other neurodevelopmental risk genes. RESULTS: These individuals share neurobehavioral features including ASD, intellectual disability, and/or ADHD; other frequent features include hypersensitivity to sensory stimuli and sleep problems. RFX3, RFX4, and RFX7 are strongly expressed in developing and adult human brain, and X-box binding motifs as well as RFX ChIP-seq peaks are enriched in the cis-regulatory regions of known ASD risk genes. CONCLUSION: These results establish a likely role of deleterious variation in RFX3, RFX4, and RFX7 in cases of monogenic intellectual disability, ADHD and ASD, and position these genes as potentially critical transcriptional regulators of neurobiological pathways associated with neurodevelopmental disease pathogenesis.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Trastorno Autístico , Discapacidad Intelectual , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Humanos , Discapacidad Intelectual/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética
11.
Am J Hum Genet ; 107(5): 963-976, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33157009

RESUMEN

NCKAP1/NAP1 regulates neuronal cytoskeletal dynamics and is essential for neuronal differentiation in the developing brain. Deleterious variants in NCKAP1 have been identified in individuals with autism spectrum disorder (ASD) and intellectual disability; however, its clinical significance remains unclear. To determine its significance, we assemble genotype and phenotype data for 21 affected individuals from 20 unrelated families with predicted deleterious variants in NCKAP1. This includes 16 individuals with de novo (n = 8), transmitted (n = 6), or inheritance unknown (n = 2) truncating variants, two individuals with structural variants, and three with potentially disruptive de novo missense variants. We report a de novo and ultra-rare deleterious variant burden of NCKAP1 in individuals with neurodevelopmental disorders which needs further replication. ASD or autistic features, language and motor delay, and variable expression of intellectual or learning disability are common clinical features. Among inherited cases, there is evidence of deleterious variants segregating with neuropsychiatric disorders. Based on available human brain transcriptomic data, we show that NCKAP1 is broadly and highly expressed in both prenatal and postnatal periods and demostrate enriched expression in excitatory neurons and radial glias but depleted expression in inhibitory neurons. Mouse in utero electroporation experiments reveal that Nckap1 loss of function promotes neuronal migration during early cortical development. Combined, these data support a role for disruptive NCKAP1 variants in neurodevelopmental delay/autism, possibly by interfering with neuronal migration early in cortical development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Trastorno del Espectro Autista/genética , Discapacidad Intelectual/genética , Discapacidades para el Aprendizaje/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adolescente , Animales , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Niño , Femenino , Expresión Génica , Genotipo , Células HEK293 , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Discapacidades para el Aprendizaje/diagnóstico , Discapacidades para el Aprendizaje/patología , Masculino , Ratones , Ratones Noqueados , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Linaje , Fenotipo , Embarazo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transcriptoma , Adulto Joven
12.
Am J Hum Genet ; 106(5): 623-631, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32275884

RESUMEN

Nucleoporins (NUPs) are an essential component of the nuclear-pore complex, which regulates nucleocytoplasmic transport of macromolecules. Pathogenic variants in NUP genes have been linked to several inherited human diseases, including a number with progressive neurological degeneration. We present six affected individuals with bi-allelic truncating variants in NUP188 and strikingly similar phenotypes and clinical courses, representing a recognizable genetic syndrome; the individuals are from four unrelated families. Key clinical features include congenital cataracts, hypotonia, prenatal-onset ventriculomegaly, white-matter abnormalities, hypoplastic corpus callosum, congenital heart defects, and central hypoventilation. Characteristic dysmorphic features include small palpebral fissures, a wide nasal bridge and nose, micrognathia, and digital anomalies. All affected individuals died as a result of respiratory failure, and five of them died within the first year of life. Nuclear import of proteins was decreased in affected individuals' fibroblasts, supporting a possible disease mechanism. CRISPR-mediated knockout of NUP188 in Drosophila revealed motor deficits and seizure susceptibility, partially recapitulating the neurological phenotype seen in affected individuals. Removal of NUP188 also resulted in aberrant dendrite tiling, suggesting a potential role of NUP188 in dendritic development. Two of the NUP188 pathogenic variants are enriched in the Ashkenazi Jewish population in gnomAD, a finding we confirmed with a separate targeted population screen of an international sampling of 3,225 healthy Ashkenazi Jewish individuals. Taken together, our results implicate bi-allelic loss-of-function NUP188 variants in a recessive syndrome characterized by a distinct neurologic, ophthalmologic, and facial phenotype.


Asunto(s)
Alelos , Encéfalo/anomalías , Proteínas de Drosophila/genética , Anomalías del Ojo/genética , Cardiopatías Congénitas/genética , Mutación con Pérdida de Función/genética , Proteínas de Complejo Poro Nuclear/genética , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Preescolar , Dendritas/metabolismo , Dendritas/patología , Drosophila melanogaster , Anomalías del Ojo/mortalidad , Femenino , Fibroblastos , Genes Recesivos , Cardiopatías Congénitas/mortalidad , Humanos , Lactante , Recién Nacido , Judíos/genética , Masculino , Proteínas de Complejo Poro Nuclear/deficiencia , Convulsiones/metabolismo , Síndrome , beta Carioferinas/metabolismo
13.
Clin Genet ; 97(6): 927-932, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32170730

RESUMEN

Two 1p36 contiguous gene deletion syndromes are known so far: the terminal 1p36 deletion syndrome and a 1p36 deletion syndrome with a critical region located more proximal at 1p36.23-1p36.22. We present even more proximally located overlapping deletions from seven individuals, with the smallest region of overlap comprising 1 Mb at 1p36.13-1p36.12 (chr1:19077793-20081292 (GRCh37/hg19)) defining a new contiguous gene deletion syndrome. The characteristic features of this new syndrome are learning disability or mild intellectual disability, speech delay, behavioral abnormalities, and ptosis. The genes UBR4 and CAPZB are considered the most likely candidate genes for the features of this new syndrome.


Asunto(s)
Blefaroptosis/genética , Proteínas de Unión a Calmodulina/genética , Proteína CapZ/genética , Trastornos de los Cromosomas/genética , Discapacidades para el Aprendizaje/genética , Ubiquitina-Proteína Ligasas/genética , Blefaroptosis/patología , Deleción Cromosómica , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 1/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Femenino , Estudios de Asociación Genética , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidades para el Aprendizaje/patología , Masculino , Fenotipo
14.
Clin Genet ; 97(5): 779-784, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32067224

RESUMEN

There is growing evidence that TP63 is associated with isolated as well as syndromic premature ovarian insufficiency (POI). We report two adolescent sisters diagnosed with undetectable ovaries, uterine hypoplasia, and mammary gland hypoplasia. A novel paternally inherited nonsense variant in TP63 [NM_003722.4 c.1927C > T,p.(Arg643*)] in exon 14 was identified by exome sequencing. One of the syndromes linked to TP63 is limb mammary syndrome (LMS), an autosomal dominant inherited disorder characterized by ectrodactyly, hypoplasia of mammary-gland and nipple, lacrimal duct stenosis, nail dysplasia, dental anomalies, cleft palate and/or cleft lip and absence of skin and hair defects. The TP63 variant segregated with symptoms of LMS in the family, however, no affected individual had limb defects. The phenotype reported here represents a novel syndromic phenotype associated with TP63. Reported cases with TP63 associated POI are reviewed.


Asunto(s)
Mama/anomalías , Predisposición Genética a la Enfermedad , Deformidades Congénitas de las Extremidades/genética , Insuficiencia Ovárica Primaria/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Mama/patología , Femenino , Genotipo , Humanos , Deformidades Congénitas de las Extremidades/patología , Persona de Mediana Edad , Mutación/genética , Linaje , Insuficiencia Ovárica Primaria/patología , Secuenciación del Exoma , Adulto Joven
15.
Brain ; 143(1): 94-111, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31855247

RESUMEN

Cerebral choline metabolism is crucial for normal brain function, and its homoeostasis depends on carrier-mediated transport. Here, we report on four individuals from three families with neurodegenerative disease and homozygous frameshift mutations (Asp517Metfs*19, Ser126Metfs*8, and Lys90Metfs*18) in the SLC44A1 gene encoding choline transporter-like protein 1. Clinical features included progressive ataxia, tremor, cognitive decline, dysphagia, optic atrophy, dysarthria, as well as urinary and bowel incontinence. Brain MRI demonstrated cerebellar atrophy and leukoencephalopathy. Moreover, low signal intensity in globus pallidus with hyperintensive streaking and low signal intensity in substantia nigra were seen in two individuals. The Asp517Metfs*19 and Ser126Metfs*8 fibroblasts were structurally and functionally indistinguishable. The most prominent ultrastructural changes of the mutant fibroblasts were reduced presence of free ribosomes, the appearance of elongated endoplasmic reticulum and strikingly increased number of mitochondria and small vesicles. When chronically treated with choline, those characteristics disappeared and mutant ultrastructure resembled healthy control cells. Functional analysis revealed diminished choline transport yet the membrane phosphatidylcholine content remained unchanged. As part of the mechanism to preserve choline and phosphatidylcholine, choline transporter deficiency was implicated in impaired membrane homeostasis of other phospholipids. Choline treatments could restore the membrane lipids, repair cellular organelles and protect mutant cells from acute iron overload. In conclusion, we describe a novel childhood-onset neurometabolic disease caused by choline transporter deficiency with autosomal recessive inheritance.


Asunto(s)
Antígenos CD/genética , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Proteínas de Transporte de Catión Orgánico/genética , Adolescente , Ataxia/genética , Ataxia/fisiopatología , Atrofia , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Colina/farmacología , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/ultraestructura , Trastornos de Deglución/genética , Trastornos de Deglución/fisiopatología , Disartria/genética , Disartria/fisiopatología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/ultraestructura , Incontinencia Fecal/genética , Incontinencia Fecal/fisiopatología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Mutación del Sistema de Lectura , Globo Pálido/diagnóstico por imagen , Trastornos Heredodegenerativos del Sistema Nervioso/diagnóstico por imagen , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Trastornos Heredodegenerativos del Sistema Nervioso/fisiopatología , Homocigoto , Humanos , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/genética , Leucoencefalopatías/fisiopatología , Imagen por Resonancia Magnética , Masculino , Microscopía Electrónica , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Nootrópicos/farmacología , Atrofia Óptica/genética , Atrofia Óptica/fisiopatología , Linaje , Ribosomas/efectos de los fármacos , Ribosomas/ultraestructura , Sustancia Negra/diagnóstico por imagen , Síndrome , Temblor/genética , Temblor/fisiopatología , Incontinencia Urinaria/genética , Incontinencia Urinaria/fisiopatología
16.
Am J Hum Genet ; 105(3): 640-657, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31402090

RESUMEN

The identification of genetic variants implicated in human developmental disorders has been revolutionized by second-generation sequencing combined with international pooling of cases. Here, we describe seven individuals who have diverse yet overlapping developmental anomalies, and who all have de novo missense FBXW11 variants identified by whole exome or whole genome sequencing and not reported in the gnomAD database. Their phenotypes include striking neurodevelopmental, digital, jaw, and eye anomalies, and in one individual, features resembling Noonan syndrome, a condition caused by dysregulated RAS signaling. FBXW11 encodes an F-box protein, part of the Skp1-cullin-F-box (SCF) ubiquitin ligase complex, involved in ubiquitination and proteasomal degradation and thus fundamental to many protein regulatory processes. FBXW11 targets include ß-catenin and GLI transcription factors, key mediators of Wnt and Hh signaling, respectively, critical to digital, neurological, and eye development. Structural analyses indicate affected residues cluster at the surface of the loops of the substrate-binding domain of FBXW11, and the variants are predicted to destabilize the protein and/or its interactions. In situ hybridization studies on human and zebrafish embryonic tissues demonstrate FBXW11 is expressed in the developing eye, brain, mandibular processes, and limb buds or pectoral fins. Knockdown of the zebrafish FBXW11 orthologs fbxw11a and fbxw11b resulted in embryos with smaller, misshapen, and underdeveloped eyes and abnormal jaw and pectoral fin development. Our findings support the role of FBXW11 in multiple developmental processes, including those involving the brain, eye, digits, and jaw.


Asunto(s)
Encéfalo/anomalías , Anomalías del Ojo/genética , Dedos/anomalías , Mutación Missense , Fenotipo , Ubiquitina-Proteína Ligasas/genética , Proteínas con Repetición de beta-Transducina/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino
19.
Clin Genet ; 95(3): 403-408, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30417326

RESUMEN

Rett syndrome is rarely suspected in males because of the X-linked dominant inheritance. In the literature, only six male patients have been reported with methyl-CpG-binding protein 2 (MECP2) mosaicism. Next-generation sequencing (NGS) methods have enabled better detection of somatic mosaicism compared to conventional Sanger sequencing; however, mosaics can still be difficult to detect. We present clinical and molecular findings in two males mosaic for a pathogenic MECP2 variant. Both have been reexamined using deep sequencing of DNA isolated from four different cell tissues (blood, muscle, fibroblasts and oral mucosa). Deep sequencing of the different tissues revealed that the variants were present in all tissues. In one patient, the molecular diagnosis could only be established by reexamination after a normal whole exome sequencing, and the other case is an example of reverse genetic diagnostics. Rett syndrome should be considered in males with neurodevelopmental delay and stereotypical hand movements. Subsequent to clinical diagnosis males should be investigated with NGS-based technologies of MECP2 with high read depth and a low threshold for variant calls. If the initial analysis on full blood derived DNA fails to confirm the suspicion, we recommend repeating the analysis on another tissue, preferentially fibroblasts to increase the diagnostic yield.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Mosaicismo , Mutación , Fenotipo , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Alelos , Biopsia , Niño , Facies , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Masculino
20.
Am J Hum Genet ; 103(5): 666-678, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343943

RESUMEN

Developmental and epileptic encephalopathies (DEEs) are severe neurodevelopmental disorders often beginning in infancy or early childhood that are characterized by intractable seizures, abundant epileptiform activity on EEG, and developmental impairment or regression. CACNA1E is highly expressed in the central nervous system and encodes the α1-subunit of the voltage-gated CaV2.3 channel, which conducts high voltage-activated R-type calcium currents that initiate synaptic transmission. Using next-generation sequencing techniques, we identified de novo CACNA1E variants in 30 individuals with DEE, characterized by refractory infantile-onset seizures, severe hypotonia, and profound developmental impairment, often with congenital contractures, macrocephaly, hyperkinetic movement disorders, and early death. Most of the 14, partially recurring, variants cluster within the cytoplasmic ends of all four S6 segments, which form the presumed CaV2.3 channel activation gate. Functional analysis of several S6 variants revealed consistent gain-of-function effects comprising facilitated voltage-dependent activation and slowed inactivation. Another variant located in the domain II S4-S5 linker results in facilitated activation and increased current density. Five participants achieved seizure freedom on the anti-epileptic drug topiramate, which blocks R-type calcium channels. We establish pathogenic variants in CACNA1E as a cause of DEEs and suggest facilitated R-type calcium currents as a disease mechanism for human epilepsy and developmental disorders.


Asunto(s)
Canales de Calcio Tipo R/genética , Proteínas de Transporte de Catión/genética , Contractura/genética , Discinesias/genética , Epilepsia/genética , Variación Genética/genética , Megalencefalia/genética , Espasmos Infantiles/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Trastornos del Neurodesarrollo/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA