Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 110(6): 963-978, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37196654

RESUMEN

De novo variants are a leading cause of neurodevelopmental disorders (NDDs), but because every monogenic NDD is different and usually extremely rare, it remains a major challenge to understand the complete phenotype and genotype spectrum of any morbid gene. According to OMIM, heterozygous variants in KDM6B cause "neurodevelopmental disorder with coarse facies and mild distal skeletal abnormalities." Here, by examining the molecular and clinical spectrum of 85 reported individuals with mostly de novo (likely) pathogenic KDM6B variants, we demonstrate that this description is inaccurate and potentially misleading. Cognitive deficits are seen consistently in all individuals, but the overall phenotype is highly variable. Notably, coarse facies and distal skeletal anomalies, as defined by OMIM, are rare in this expanded cohort while other features are unexpectedly common (e.g., hypotonia, psychosis, etc.). Using 3D protein structure analysis and an innovative dual Drosophila gain-of-function assay, we demonstrated a disruptive effect of 11 missense/in-frame indels located in or near the enzymatic JmJC or Zn-containing domain of KDM6B. Consistent with the role of KDM6B in human cognition, we demonstrated a role for the Drosophila KDM6B ortholog in memory and behavior. Taken together, we accurately define the broad clinical spectrum of the KDM6B-related NDD, introduce an innovative functional testing paradigm for the assessment of KDM6B variants, and demonstrate a conserved role for KDM6B in cognition and behavior. Our study demonstrates the critical importance of international collaboration, sharing of clinical data, and rigorous functional analysis of genetic variants to ensure correct disease diagnosis for rare disorders.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Animales , Facies , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Drosophila , Discapacidad Intelectual/patología , Histona Demetilasas con Dominio de Jumonji/genética
2.
Brain ; 147(4): 1436-1456, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37951597

RESUMEN

The acyl-CoA-binding domain-containing protein 6 (ACBD6) is ubiquitously expressed, plays a role in the acylation of lipids and proteins and regulates the N-myristoylation of proteins via N-myristoyltransferase enzymes (NMTs). However, its precise function in cells is still unclear, as is the consequence of ACBD6 defects on human pathophysiology. Using exome sequencing and extensive international data sharing efforts, we identified 45 affected individuals from 28 unrelated families (consanguinity 93%) with bi-allelic pathogenic, predominantly loss-of-function (18/20) variants in ACBD6. We generated zebrafish and Xenopus tropicalis acbd6 knockouts by CRISPR/Cas9 and characterized the role of ACBD6 on protein N-myristoylation with myristic acid alkyne (YnMyr) chemical proteomics in the model organisms and human cells, with the latter also being subjected further to ACBD6 peroxisomal localization studies. The affected individuals (23 males and 22 females), aged 1-50 years, typically present with a complex and progressive disease involving moderate-to-severe global developmental delay/intellectual disability (100%) with significant expressive language impairment (98%), movement disorders (97%), facial dysmorphism (95%) and mild cerebellar ataxia (85%) associated with gait impairment (94%), limb spasticity/hypertonia (76%), oculomotor (71%) and behavioural abnormalities (65%), overweight (59%), microcephaly (39%) and epilepsy (33%). The most conspicuous and common movement disorder was dystonia (94%), frequently leading to early-onset progressive postural deformities (97%), limb dystonia (55%) and cervical dystonia (31%). A jerky tremor in the upper limbs (63%), a mild head tremor (59%), parkinsonism/hypokinesia developing with advancing age (32%) and simple motor and vocal tics were among other frequent movement disorders. Midline brain malformations including corpus callosum abnormalities (70%), hypoplasia/agenesis of the anterior commissure (66%), short midbrain and small inferior cerebellar vermis (38% each) as well as hypertrophy of the clava (24%) were common neuroimaging findings. Acbd6-deficient zebrafish and Xenopus models effectively recapitulated many clinical phenotypes reported in patients including movement disorders, progressive neuromotor impairment, seizures, microcephaly, craniofacial dysmorphism and midbrain defects accompanied by developmental delay with increased mortality over time. Unlike ACBD5, ACBD6 did not show a peroxisomal localization and ACBD6-deficiency was not associated with altered peroxisomal parameters in patient fibroblasts. Significant differences in YnMyr-labelling were observed for 68 co- and 18 post-translationally N-myristoylated proteins in patient-derived fibroblasts. N-myristoylation was similarly affected in acbd6-deficient zebrafish and X. tropicalis models, including Fus, Marcks and Chchd-related proteins implicated in neurological diseases. The present study provides evidence that bi-allelic pathogenic variants in ACBD6 lead to a distinct neurodevelopmental syndrome accompanied by complex and progressive cognitive and movement disorders.


Asunto(s)
Discapacidad Intelectual , Microcefalia , Trastornos del Movimiento , Malformaciones del Sistema Nervioso , Trastornos del Neurodesarrollo , Animales , Femenino , Humanos , Masculino , Transportadoras de Casetes de Unión a ATP , Discapacidad Intelectual/genética , Trastornos del Movimiento/genética , Malformaciones del Sistema Nervioso/genética , Trastornos del Neurodesarrollo/genética , Temblor , Pez Cebra , Lactante , Preescolar , Niño , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad
3.
Am J Hum Genet ; 108(6): 1138-1150, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33909992

RESUMEN

ANKRD17 is an ankyrin repeat-containing protein thought to play a role in cell cycle progression, whose ortholog in Drosophila functions in the Hippo pathway as a co-factor of Yorkie. Here, we delineate a neurodevelopmental disorder caused by de novo heterozygous ANKRD17 variants. The mutational spectrum of this cohort of 34 individuals from 32 families is highly suggestive of haploinsufficiency as the underlying mechanism of disease, with 21 truncating or essential splice site variants, 9 missense variants, 1 in-frame insertion-deletion, and 1 microdeletion (1.16 Mb). Consequently, our data indicate that loss of ANKRD17 is likely the main cause of phenotypes previously associated with large multi-gene chromosomal aberrations of the 4q13.3 region. Protein modeling suggests that most of the missense variants disrupt the stability of the ankyrin repeats through alteration of core structural residues. The major phenotypic characteristic of our cohort is a variable degree of developmental delay/intellectual disability, particularly affecting speech, while additional features include growth failure, feeding difficulties, non-specific MRI abnormalities, epilepsy and/or abnormal EEG, predisposition to recurrent infections (mostly bacterial), ophthalmological abnormalities, gait/balance disturbance, and joint hypermobility. Moreover, many individuals shared similar dysmorphic facial features. Analysis of single-cell RNA-seq data from the developing human telencephalon indicated ANKRD17 expression at multiple stages of neurogenesis, adding further evidence to the assertion that damaging ANKRD17 variants cause a neurodevelopmental disorder.


Asunto(s)
Anomalías Craneofaciales/etiología , Heterocigoto , Discapacidad Intelectual/etiología , Trastornos del Desarrollo del Lenguaje/etiología , Mutación con Pérdida de Función , Proteínas de Unión al ARN/genética , Adolescente , Adulto , Niño , Preescolar , Anomalías Craneofaciales/patología , Femenino , Haploinsuficiencia , Humanos , Lactante , Discapacidad Intelectual/patología , Trastornos del Desarrollo del Lenguaje/patología , Masculino , Linaje , Fenotipo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Síndrome , Adulto Joven
4.
Genet Med ; 26(5): 101097, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38334070

RESUMEN

PURPOSE: Pathogenic variants of FIG4 generate enlarged lysosomes and neurological and developmental disorders. To identify additional genes regulating lysosomal volume, we carried out a genome-wide activation screen to detect suppression of enlarged lysosomes in FIG4-/- cells. METHODS: The CRISPR-a gene activation screen utilized sgRNAs from the promoters of protein-coding genes. Fluorescence-activated cell sorting separated cells with correction of the enlarged lysosomes from uncorrected cells. Patient variants of SLC12A9 were identified by exome or genome sequencing and studied by segregation analysis and clinical characterization. RESULTS: Overexpression of SLC12A9, a solute co-transporter, corrected lysosomal swelling in FIG4-/- cells. SLC12A9 (NP_064631.2) colocalized with LAMP2 at the lysosome membrane. Biallelic variants of SLC12A9 were identified in 3 unrelated probands with neurodevelopmental disorders. Common features included intellectual disability, skeletal and brain structural abnormalities, congenital heart defects, and hypopigmented hair. Patient 1 was homozygous for nonsense variant p.(Arg615∗), patient 2 was compound heterozygous for p.(Ser109Lysfs∗20) and a large deletion, and proband 3 was compound heterozygous for p.(Glu290Glyfs∗36) and p.(Asn552Lys). Fibroblasts from proband 1 contained enlarged lysosomes that were corrected by wild-type SLC12A9 cDNA. Patient variant p.(Asn552Lys) failed to correct the lysosomal defect. CONCLUSION: Impaired function of SLC12A9 results in enlarged lysosomes and a recessive disorder with a recognizable neurodevelopmental phenotype.


Asunto(s)
Lisosomas , Trastornos del Neurodesarrollo , Simportadores de Cloruro de Sodio-Potasio , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Alelos , Mutación con Pérdida de Función/genética , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Linaje , Fenotipo , Simportadores de Cloruro de Sodio-Potasio/genética
5.
Am J Hum Genet ; 107(1): 34-45, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32497488

RESUMEN

IFAP syndrome is a rare genetic disorder characterized by ichthyosis follicularis, atrichia, and photophobia. Previous research found that mutations in MBTPS2, encoding site-2-protease (S2P), underlie X-linked IFAP syndrome. The present report describes the identification via whole-exome sequencing of three heterozygous mutations in SREBF1 in 11 unrelated, ethnically diverse individuals with autosomal-dominant IFAP syndrome. SREBF1 encodes sterol regulatory element-binding protein 1 (SREBP1), which promotes the transcription of lipogenes involved in the biosynthesis of fatty acids and cholesterols. This process requires cleavage of SREBP1 by site-1-protease (S1P) and S2P and subsequent translocation into the nucleus where it binds to sterol regulatory elements (SRE). The three detected SREBF1 mutations caused substitution or deletion of residues 527, 528, and 530, which are crucial for S1P cleavage. In vitro investigation of SREBP1 variants demonstrated impaired S1P cleavage, which prohibited nuclear translocation of the transcriptionally active form of SREBP1. As a result, SREBP1 variants exhibited significantly lower transcriptional activity compared to the wild-type, as demonstrated via luciferase reporter assay. RNA sequencing of the scalp skin from IFAP-affected individuals revealed a dramatic reduction in transcript levels of low-density lipoprotein receptor (LDLR) and of keratin genes known to be expressed in the outer root sheath of hair follicles. An increased rate of in situ keratinocyte apoptosis, which might contribute to skin hyperkeratosis and hypotrichosis, was also detected in scalp samples from affected individuals. Together with previous research, the present findings suggest that SREBP signaling plays an essential role in epidermal differentiation, skin barrier formation, hair growth, and eye function.


Asunto(s)
Artrogriposis/genética , Mutación/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Regulación de la Expresión Génica/genética , Humanos , Queratosis/genética , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Adulto Joven
6.
Genet Med ; 25(8): 100885, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37165955

RESUMEN

PURPOSE: Missense variants clustering in the BTB domain region of RHOBTB2 cause a developmental and epileptic encephalopathy with early-onset seizures and severe intellectual disability. METHODS: By international collaboration, we assembled individuals with pathogenic RHOBTB2 variants and a variable spectrum of neurodevelopmental disorders. By western blotting, we investigated the consequences of missense variants in vitro. RESULTS: In accordance with previous observations, de novo heterozygous missense variants in the BTB domain region led to a severe developmental and epileptic encephalopathy in 16 individuals. Now, we also identified de novo missense variants in the GTPase domain in 6 individuals with apparently more variable neurodevelopmental phenotypes with or without epilepsy. In contrast to variants in the BTB domain region, variants in the GTPase domain do not impair proteasomal degradation of RHOBTB2 in vitro, indicating different functional consequences. Furthermore, we observed biallelic splice-site and truncating variants in 9 families with variable neurodevelopmental phenotypes, indicating that complete loss of RHOBTB2 is pathogenic as well. CONCLUSION: By identifying genotype-phenotype correlations regarding location and consequences of de novo missense variants in RHOBTB2 and by identifying biallelic truncating variants, we further delineate and expand the molecular and clinical spectrum of RHOBTB2-related phenotypes, including both autosomal dominant and recessive neurodevelopmental disorders.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Epilepsia/genética , Epilepsia/patología , Estudios de Asociación Genética , Discapacidad Intelectual/genética , Fenotipo , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/genética , Proteínas Supresoras de Tumor/genética
7.
Am J Hum Genet ; 105(2): 395-402, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31353022

RESUMEN

The glycosylphosphatidylinositol (GPI) anchor links over 150 proteins to the cell surface and is present on every cell type. Many of these proteins play crucial roles in neuronal development and function. Mutations in 18 of the 29 genes implicated in the biosynthesis of the GPI anchor have been identified as the cause of GPI biosynthesis deficiencies (GPIBDs) in humans. GPIBDs are associated with intellectual disability and seizures as their cardinal features. An essential component of the GPI transamidase complex is PIGU, along with PIGK, PIGS, PIGT, and GPAA1, all of which link GPI-anchored proteins (GPI-APs) onto the GPI anchor in the endoplasmic reticulum (ER). Here, we report two homozygous missense mutations (c.209T>A [p.Ile70Lys] and c.1149C>A [p.Asn383Lys]) in five individuals from three unrelated families. All individuals presented with global developmental delay, severe-to-profound intellectual disability, muscular hypotonia, seizures, brain anomalies, scoliosis, and mild facial dysmorphism. Using multicolor flow cytometry, we determined a characteristic profile for GPI transamidase deficiency. On granulocytes this profile consisted of reduced cell-surface expression of fluorescein-labeled proaerolysin (FLAER), CD16, and CD24, but not of CD55 and CD59; additionally, B cells showed an increased expression of free GPI anchors determined by T5 antibody. Moreover, computer-assisted facial analysis of different GPIBDs revealed a characteristic facial gestalt shared among individuals with mutations in PIGU and GPAA1. Our findings improve our understanding of the role of the GPI transamidase complex in the development of nervous and skeletal systems and expand the clinical spectrum of disorders belonging to the group of inherited GPI-anchor deficiencies.


Asunto(s)
Aciltransferasas/genética , Encefalopatías/etiología , Epilepsia/etiología , Glicosilfosfatidilinositoles/biosíntesis , Glicosilfosfatidilinositoles/deficiencia , Discapacidad Intelectual/etiología , Mutación , Convulsiones/patología , Adolescente , Adulto , Secuencia de Aminoácidos , Encefalopatías/patología , Niño , Preescolar , Epilepsia/patología , Femenino , Glicosilfosfatidilinositoles/genética , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/patología , Masculino , Linaje , Convulsiones/genética , Homología de Secuencia , Adulto Joven
8.
Am J Hum Genet ; 104(6): 1139-1157, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31155282

RESUMEN

Zimmermann-Laband syndrome (ZLS) is characterized by coarse facial features with gingival enlargement, intellectual disability (ID), hypertrichosis, and hypoplasia or aplasia of nails and terminal phalanges. De novo missense mutations in KCNH1 and KCNK4, encoding K+ channels, have been identified in subjects with ZLS and ZLS-like phenotype, respectively. We report de novo missense variants in KCNN3 in three individuals with typical clinical features of ZLS. KCNN3 (SK3/KCa2.3) constitutes one of three members of the small-conductance Ca2+-activated K+ (SK) channels that are part of a multiprotein complex consisting of the pore-forming channel subunits, the constitutively bound Ca2+ sensor calmodulin, protein kinase CK2, and protein phosphatase 2A. CK2 modulates Ca2+ sensitivity of the channels by phosphorylating SK-bound calmodulin. Patch-clamp whole-cell recordings of KCNN3 channel-expressing CHO cells demonstrated that disease-associated mutations result in gain of function of the mutant channels, characterized by increased Ca2+ sensitivity leading to faster and more complete activation of KCNN3 mutant channels. Pretreatment of cells with the CK2 inhibitor 4,5,6,7-tetrabromobenzotriazole revealed basal inhibition of wild-type and mutant KCNN3 channels by CK2. Analogous experiments with the KCNN3 p.Val450Leu mutant previously identified in a family with portal hypertension indicated basal constitutive channel activity and thus a different gain-of-function mechanism compared to the ZLS-associated mutant channels. With the report on de novo KCNK4 mutations in subjects with facial dysmorphism, hypertrichosis, epilepsy, ID, and gingival overgrowth, we propose to combine the phenotypes caused by mutations in KCNH1, KCNK4, and KCNN3 in a group of neurological potassium channelopathies caused by an increase in K+ conductance.


Asunto(s)
Anomalías Múltiples/etiología , Anomalías Craneofaciales/etiología , Fibromatosis Gingival/etiología , Mutación con Ganancia de Función , Deformidades Congénitas de la Mano/etiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Anomalías Múltiples/patología , Adulto , Secuencia de Aminoácidos , Animales , Células CHO , Niño , Preescolar , Anomalías Craneofaciales/patología , Cricetinae , Cricetulus , Femenino , Fibromatosis Gingival/patología , Deformidades Congénitas de la Mano/patología , Humanos , Activación del Canal Iónico , Masculino , Persona de Mediana Edad , Fenotipo , Conformación Proteica , Homología de Secuencia , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
9.
Clin Genet ; 100(6): 766-770, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34490615

RESUMEN

Neurological symptoms are frequent and often a leading feature of childhood-onset mitochondrial disorders (MD) but the exact incidence of MD in unselected neuropediatric patients is unknown. Their early detection is desirable due to a potentially rapid clinical decline and the availability of management options. In 491 children with neurological symptoms, a comprehensive diagnostic work-up including exome sequencing was performed. The success rate in terms of a molecular genetic diagnosis within our cohort was 51%. Disease-causing variants in a mitochondria-associated gene were detected in 12% of solved cases. In order to facilitate the clinical identification of MDs within neuropediatric cohorts, we have created an easy-to-use bedside-tool, the MDC-NP. In our cohort, the MDC-NP predicted disease conditions related to MDs with a sensitivity of 0.83, and a specificity of 0.96.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Mitocondriales/epidemiología , Enfermedades Mitocondriales/genética , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/genética , Factores de Edad , Alelos , Niño , Estudios de Cohortes , Genes Mitocondriales , Estudios de Asociación Genética , Genotipo , Humanos , Enfermedades Mitocondriales/diagnóstico , Mutación , Enfermedades del Sistema Nervioso/diagnóstico , Fenotipo , Prevalencia , Pronóstico
10.
Brain ; 143(8): 2437-2453, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32761064

RESUMEN

In pleiotropic diseases, multiple organ systems are affected causing a variety of clinical manifestations. Here, we report a pleiotropic disorder with a unique constellation of neurological, endocrine, exocrine, and haematological findings that is caused by biallelic MADD variants. MADD, the mitogen-activated protein kinase (MAPK) activating death domain protein, regulates various cellular functions, such as vesicle trafficking, activity of the Rab3 and Rab27 small GTPases, tumour necrosis factor-α (TNF-α)-induced signalling and prevention of cell death. Through national collaboration and GeneMatcher, we collected 23 patients with 21 different pathogenic MADD variants identified by next-generation sequencing. We clinically evaluated the series of patients and categorized the phenotypes in two groups. Group 1 consists of 14 patients with severe developmental delay, endo- and exocrine dysfunction, impairment of the sensory and autonomic nervous system, and haematological anomalies. The clinical course during the first years of life can be potentially fatal. The nine patients in Group 2 have a predominant neurological phenotype comprising mild-to-severe developmental delay, hypotonia, speech impairment, and seizures. Analysis of mRNA revealed multiple aberrant MADD transcripts in two patient-derived fibroblast cell lines. Relative quantification of MADD mRNA and protein in fibroblasts of five affected individuals showed a drastic reduction or loss of MADD. We conducted functional tests to determine the impact of the variants on different pathways. Treatment of patient-derived fibroblasts with TNF-α resulted in reduced phosphorylation of the extracellular signal-regulated kinases 1 and 2, enhanced activation of the pro-apoptotic enzymes caspase-3 and -7 and increased apoptosis compared to control cells. We analysed internalization of epidermal growth factor in patient cells and identified a defect in endocytosis of epidermal growth factor. We conclude that MADD deficiency underlies multiple cellular defects that can be attributed to alterations of TNF-α-dependent signalling pathways and defects in vesicular trafficking. Our data highlight the multifaceted role of MADD as a signalling molecule in different organs and reveal its physiological role in regulating the function of the sensory and autonomic nervous system and endo- and exocrine glands.


Asunto(s)
Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Discapacidades del Desarrollo/genética , Factores de Intercambio de Guanina Nucleótido/genética , Enfermedades del Sistema Nervioso/genética , Humanos , Mutación , Fenotipo , Transporte de Proteínas/genética , Transducción de Señal/genética
11.
J Neurochem ; 155(3): 250-263, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32202324

RESUMEN

Mutations in SHANK3, coding for a large scaffold protein of excitatory synapses in the CNS, are associated with neurodevelopmental disorders including autism spectrum disorders and intellectual disability (ID). Several cases have been identified in which the mutation leads to truncation of the protein, eliminating C-terminal sequences required for post-synaptic targeting of the protein. We identify here a patient with a truncating mutation in SHANK3, affected by severe global developmental delay and intellectual disability. By analyzing the subcellular distribution of this truncated form of Shank3, we identified a nuclear localization signal (NLS) in the N-terminal part of the protein which is responsible for targeting Shank3 fragments to the nucleus. To determine the relevance of Shank3 for nuclear signaling, we analyze how it affects signaling by ß-catenin, a component of the Wnt pathway. We show that full length as well as truncated variants of Shank3 interact with ß-catenin via the PDZ domain of Shank3, and the armadillo repeats of ß-catenin. As a result of this interaction, truncated forms of Shank3 and ß-catenin strictly co-localize in small intra-nuclear bodies both in 293T cells and in rat hippocampal neurons. On a functional level, the sequestration of both proteins in these nuclear bodies is associated with a strongly repressed transcriptional activation by ß-catenin owing to interaction with the truncated Shank3 fragment found in patients. Our data suggest that truncating mutations in SHANK3 may not only lead to a reduction in Shank3 protein available at postsynaptic sites but also negatively affect the Wnt signaling pathway.


Asunto(s)
Núcleo Celular/metabolismo , Discapacidades del Desarrollo/metabolismo , Mutación/fisiología , Proteínas del Tejido Nervioso/metabolismo , beta Catenina/metabolismo , Animales , Núcleo Celular/genética , Células Cultivadas , Discapacidades del Desarrollo/genética , Femenino , Células HEK293 , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Embarazo , Ratas , Ratas Wistar , Transducción de Señal/fisiología
12.
Am J Hum Genet ; 100(1): 117-127, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28017373

RESUMEN

From a GeneMatcher-enabled international collaboration, we identified ten individuals affected by intellectual disability, speech delay, ataxia, and facial dysmorphism and carrying a deleterious EBF3 variant detected by whole-exome sequencing. One 9-bp duplication and one splice-site, five missense, and two nonsense variants in EBF3 were found; the mutations occurred de novo in eight individuals, and the missense variant c.625C>T (p.Arg209Trp) was inherited by two affected siblings from their healthy mother, who is mosaic. EBF3 belongs to the early B cell factor family (also known as Olf, COE, or O/E) and is a transcription factor involved in neuronal differentiation and maturation. Structural assessment predicted that the five amino acid substitutions have damaging effects on DNA binding of EBF3. Transient expression of EBF3 mutant proteins in HEK293T cells revealed mislocalization of all but one mutant in the cytoplasm, as well as nuclear localization. By transactivation assays, all EBF3 mutants showed significantly reduced or no ability to activate transcription of the reporter gene CDKN1A, and in situ subcellular fractionation experiments demonstrated that EBF3 mutant proteins were less tightly associated with chromatin. Finally, in RNA-seq and ChIP-seq experiments, EBF3 acted as a transcriptional regulator, and mutant EBF3 had reduced genome-wide DNA binding and gene-regulatory activity. Our findings demonstrate that variants disrupting EBF3-mediated transcriptional regulation cause intellectual disability and developmental delay and are present in ∼0.1% of individuals with unexplained neurodevelopmental disorders.


Asunto(s)
Ataxia/genética , Cara/anomalías , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Mutación , Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Adolescente , Adulto , Sustitución de Aminoácidos , Niño , Preescolar , Cromatina/genética , Cromatina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Discapacidades del Desarrollo/genética , Exoma/genética , Femenino , Regulación de la Expresión Génica/genética , Genes Reporteros , Células HEK293 , Humanos , Masculino , Modelos Moleculares , Mosaicismo , Transporte de Proteínas/genética , Síndrome , Factores de Transcripción/química , Factores de Transcripción/metabolismo
13.
Am J Hum Genet ; 99(4): 991-999, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693232

RESUMEN

The ASXL genes (ASXL1, ASXL2, and ASXL3) participate in body patterning during embryogenesis and encode proteins involved in epigenetic regulation and assembly of transcription factors to specific genomic loci. Germline de novo truncating variants in ASXL1 and ASXL3 have been respectively implicated in causing Bohring-Opitz and Bainbridge-Ropers syndromes, which result in overlapping features of severe intellectual disability and dysmorphic features. ASXL2 has not yet been associated with a human Mendelian disorder. In this study, we performed whole-exome sequencing in six unrelated probands with developmental delay, macrocephaly, and dysmorphic features. All six had de novo truncating variants in ASXL2. A careful review enabled the recognition of a specific phenotype consisting of macrocephaly, prominent eyes, arched eyebrows, hypertelorism, a glabellar nevus flammeus, neonatal feeding difficulties, hypotonia, and developmental disabilities. Although overlapping features with Bohring-Opitz and Bainbridge-Ropers syndromes exist, features that distinguish the ASXL2-associated condition from ASXL1- and ASXL3-related disorders are macrocephaly, absence of growth retardation, and more variability in the degree of intellectual disabilities. We were also able to demonstrate with mRNA studies that these variants are likely to exert a dominant-negative effect, given that both alleles are expressed in blood and the mutated ASXL2 transcripts escape nonsense-mediated decay. In conclusion, de novo truncating variants in ASXL2 underlie a neurodevelopmental syndrome with a clinically recognizable phenotype. This report expands the germline disorders that are linked to the ASXL genes.


Asunto(s)
Fenotipo , Proteínas Represoras/genética , Niño , Preescolar , Discapacidades del Desarrollo/genética , Exoma/genética , Cejas/anomalías , Humanos , Hipertelorismo/genética , Lactante , Recién Nacido , Masculino , Megalencefalia/genética , Hipotonía Muscular/genética , ARN Mensajero/metabolismo , Síndrome
14.
Am J Hum Genet ; 99(3): 711-719, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27545680

RESUMEN

The overall understanding of the molecular etiologies of intellectual disability (ID) and developmental delay (DD) is increasing as next-generation sequencing technologies identify genetic variants in individuals with such disorders. However, detailed analyses conclusively confirming these variants, as well as the underlying molecular mechanisms explaining the diseases, are often lacking. Here, we report on an ID syndrome caused by de novo heterozygous loss-of-function (LoF) mutations in SON. The syndrome is characterized by ID and/or DD, malformations of the cerebral cortex, epilepsy, vision problems, musculoskeletal abnormalities, and congenital malformations. Knockdown of son in zebrafish resulted in severe malformation of the spine, brain, and eyes. Importantly, analyses of RNA from affected individuals revealed that genes critical for neuronal migration and cortex organization (TUBG1, FLNA, PNKP, WDR62, PSMD3, and HDAC6) and metabolism (PCK2, PFKL, IDH2, ACY1, and ADA) are significantly downregulated because of the accumulation of mis-spliced transcripts resulting from erroneous SON-mediated RNA splicing. Our data highlight SON as a master regulator governing neurodevelopment and demonstrate the importance of SON-mediated RNA splicing in human development.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Proteínas de Unión al ADN/genética , Genes Esenciales/genética , Discapacidad Intelectual/genética , Antígenos de Histocompatibilidad Menor/genética , Mutación/genética , Empalme del ARN/genética , Animales , Encéfalo/anomalías , Encéfalo/patología , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Discapacidades del Desarrollo/fisiopatología , Anomalías del Ojo/genética , Femenino , Haploinsuficiencia/genética , Cabeza/anomalías , Heterocigoto , Humanos , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Antígenos de Histocompatibilidad Menor/análisis , Antígenos de Histocompatibilidad Menor/metabolismo , Linaje , ARN Mensajero/análisis , Columna Vertebral/anomalías , Síndrome , Pez Cebra/anomalías , Pez Cebra/embriología , Pez Cebra/genética
15.
Am J Med Genet A ; 179(10): 2056-2066, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31407851

RESUMEN

Co-occurrence of primordial dwarfism and microcephaly together with particular skeletal findings are seen in a wide range of Mendelian syndromes including microcephaly micromelia syndrome (MMS, OMIM 251230), microcephaly, short stature, and limb abnormalities (MISSLA, OMIM 617604), and microcephalic primordial dwarfisms (MPDs). Genes associated with these syndromes encode proteins that have crucial roles in DNA replication or in other critical steps of the cell cycle that link DNA replication to cell division. We identified four unrelated families with five affected individuals having biallelic or de novo variants in DONSON presenting with a core phenotype of severe short stature (z score < -3 SD), additional skeletal abnormalities, and microcephaly. Two apparently unrelated families with identical homozygous c.631C > T p.(Arg211Cys) variant had clinical features typical of Meier-Gorlin syndrome (MGS), while two siblings with compound heterozygous c.346delG p.(Asp116Ile*62) and c.1349A > G p.(Lys450Arg) variants presented with Seckel-like phenotype. We also identified a de novo c.683G > T p.(Trp228Leu) variant in DONSON in a patient with prominent micrognathia, short stature and hypoplastic femur and tibia, clinically diagnosed with Femoral-Facial syndrome (FFS, OMIM 134780). Biallelic variants in DONSON have been recently described in individuals with microcephalic dwarfism. These studies also demonstrated that DONSON has an essential conserved role in the cell cycle. Here we describe novel biallelic and de novo variants that are associated with MGS, Seckel-like phenotype and FFS, the last of which has not been associated with any disease gene to date.


Asunto(s)
Alelos , Huesos/anomalías , Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Enanismo/genética , Microcefalia/genética , Proteínas Nucleares/genética , Niño , Preescolar , Enanismo/complicaciones , Familia , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Microcefalia/complicaciones , Linaje , Fenotipo
16.
Neurogenetics ; 19(3): 151-156, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29808465

RESUMEN

The human WWOX (WW domain-containing oxidoreductase) gene, originally known as a tumor suppressor gene, has been shown to be important for brain function and development. In recent years, mutations in WWOX have been associated with a wide phenotypic spectrum of autosomal recessively inherited neurodevelopmental disorders. Whole exome sequencing was completed followed by Sanger sequencing to verify segregation of the identified variants. Functional WWOX analysis was performed in fibroblasts of one patient. Transcription and translation were assessed by quantitative real-time PCR and Western blotting. We report two related patients who presented with early epilepsy refractory to treatment, progressive microcephaly, profound developmental delay, and brain MRI abnormalities. Additionally, one of the patients showed bilateral optic atrophy. Whole exome sequencing revealed homozygosity for a novel missense variant affecting the evolutionary conserved amino acid Gln230 in the catalytic short-chain dehydrogenase/reductase (SDR) domain of WWOX in both girls. Functional studies showed normal levels of WWOX transcripts but absence of WWOX protein. To our knowledge, our patients are the first individuals presenting the more severe end of the phenotypic spectrum of WWOX deficiency, although they were only affected by a single missense variant of WWOX. This could be explained by the functional data indicating an impaired translation or premature degradation of the WWOX protein.


Asunto(s)
Discapacidades del Desarrollo/genética , Mutación Missense , Espasmos Infantiles/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Oxidorreductasa que Contiene Dominios WW/deficiencia , Oxidorreductasa que Contiene Dominios WW/genética , Afganistán , Edad de Inicio , Células Cultivadas , Niño , Consanguinidad , Discapacidades del Desarrollo/complicaciones , Epilepsia/complicaciones , Epilepsia/genética , Familia , Femenino , Células HEK293 , Humanos , Recién Nacido , Linaje , Dominios Proteicos/genética , Estabilidad del ARN/genética , Índice de Severidad de la Enfermedad , Espasmos Infantiles/complicaciones , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW/química , Oxidorreductasa que Contiene Dominios WW/metabolismo
17.
Am J Med Genet A ; 176(4): 992-996, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29575616

RESUMEN

Robinow syndrome is a clinically and genetically heterogeneous disorder characterized by mesomelic limb shortening, distinctive facial features, and variable oral, cardiac, vertebral, and urogenital malformations. We identified the novel de novo splice acceptor mutation c.1715-2A > C in DVL3 in a 15-year-old female patient with typical features of Robinow syndrome. By studying DVL3 transcripts in this patient, we confirmed expression of both wild-type and mutant alleles. Mutant DVL3 mRNAs were found to harbor a deletion of four nucleotides at the beginning of exon 15 and encode a protein product with a distinct -1 reading-frame C-terminus. The data demonstrate that mutant DVL3 proteins associated with Robinow syndrome show truncation of the C-terminus and share 83 novel amino acid residues before the stop codon confirming highly specific DVL3 alterations to be associated with this syndrome. The phenotype of the Robinow syndrome-affected female reported here is typical as she shows mesomelia and mild hand anomalies as well as characteristic facial anomalies. She also exhibited a supraumbilical midline abdominal raphe which has not been observed in other patients with Robinow syndrome. In contrast to the clinical data of four previously reported individuals with DVL3-related Robinow syndrome, short stature was not present in this individual at the age of 15 years. These findings expand the clinical spectrum of Robinow syndrome associated with DVL3 mutations. To date, comparison of clinical data of DVL3 mutation-positive individuals with those of patients with genetically different forms did not allow delineation of gene-specific phenotypes.


Asunto(s)
Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/genética , Proteínas Dishevelled/genética , Enanismo/diagnóstico , Enanismo/genética , Estudios de Asociación Genética , Deformidades Congénitas de las Extremidades/diagnóstico , Deformidades Congénitas de las Extremidades/genética , Mutación , Empalme del ARN , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adolescente , Alelos , Sustitución de Aminoácidos , Exones , Facies , Femenino , Humanos , Fenotipo , Sitios de Empalme de ARN , Radiografía , Eliminación de Secuencia
18.
Am J Med Genet A ; 170(7): 1813-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27144914

RESUMEN

Hereditary congenital facial paresis (HCFP) belongs to the congenital cranial dysinnervation disorders. HCFP is characterized by the isolated dysfunction of the seventh cranial nerve and can be associated with hearing loss, strabismus, and orofacial anomalies. Möbius syndrome shares facial palsy with HCFP, but is additionally characterized by limited abduction of the eye(s). Genetic heterogeneity has been documented for HCFP as one locus mapped to chromosome 3q21-q22 (HCFP1) and a second to 10q21.3-q22.1 (HCFP2). The only known causative gene for HCFP is HOXB1 (17q21; HCFP3), encoding a homeodomain-containing transcription factor of the HOX gene family, which are master regulators of early developmental processes. The previously reported HOXB1 mutations change arginine 207 to another residue in the homeodomain and alter binding capacity of HOXB1 for transcriptional co-regulators and DNA. We performed whole exome sequencing in HCFP-affected individuals of a large consanguineous Moroccan family. The homozygous nonsense variant c.66C>G/p.(Tyr22*) in HOXB1 was identified in the four patients with HCFP and ear malformations, while healthy family members carried the mutation in the heterozygous state. This is the first disease-associated HOXB1 mutation with a likely loss-of-function effect suggesting that all HOXB1 variants reported so far also have severe impact on activity of this transcriptional regulator. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Exoma/genética , Parálisis Facial/congénito , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Adulto , Niño , Consanguinidad , Parálisis Facial/genética , Parálisis Facial/fisiopatología , Femenino , Heterogeneidad Genética , Heterocigoto , Homocigoto , Humanos , Masculino , Mutación , Transcripción Genética
20.
Eur J Hum Genet ; 31(4): 461-468, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36747006

RESUMEN

Haploinsufficiency of TRIP12 causes a neurodevelopmental disorder characterized by intellectual disability associated with epilepsy, autism spectrum disorder and dysmorphic features, also named Clark-Baraitser syndrome. Only a limited number of cases have been reported to date. We aimed to further delineate the TRIP12-associated phenotype and objectify characteristic facial traits through GestaltMatcher image analysis based on deep-learning algorithms in order to establish a TRIP12 gestalt. 38 individuals between 3 and 66 years (F = 20, M = 18) - 1 previously published and 37 novel individuals - were recruited through an ERN ITHACA call for collaboration. 35 TRIP12 variants were identified, including frameshift (n = 15) and nonsense (n = 6) variants, as well as missense (n = 5) and splice (n = 3) variants, intragenic deletions (n = 4) and two multigene deletions disrupting TRIP12. Though variable in severity, global developmental delay was noted in all individuals, with language deficit most pronounced. About half showed autistic features and susceptibility to obesity seemed inherent to this disorder. A more severe expression was noted in individuals with a missense variant. Facial analysis showed a clear gestalt including deep-set eyes with narrow palpebral fissures and fullness of the upper eyelids, downturned corners of the mouth and large, often low-set ears with prominent earlobes. We report the largest cohort to date of individuals with TRIP12 variants, further delineating the associated phenotype and introducing a facial gestalt. These findings will improve future counseling and patient guidance.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Trastorno del Espectro Autista/genética , Discapacidad Intelectual/genética , Fenotipo , Trastornos del Neurodesarrollo/genética , Mutación Missense , Proteínas Portadoras/genética , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA