Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(22): 3123-3134, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37166351

RESUMEN

Germline pathogenic variants in two genes encoding the lysine-specific histone methyltransferase genes SETD1A and SETD2 are associated with neurodevelopmental disorders (NDDs) characterized by developmental delay and congenital anomalies. The SETD1A and SETD2 gene products play a critical role in chromatin-mediated regulation of gene expression. Specific methylation episignatures have been detected for a range of chromatin gene-related NDDs and have impacted clinical practice by improving the interpretation of variant pathogenicity. To investigate if SETD1A and/or SETD2-related NDDs are associated with a detectable episignature, we undertook targeted genome-wide methylation profiling of > 2 M CpGs using a next-generation sequencing-based assay. A comparison of methylation profiles in patients with SETD1A variants (n = 6) did not reveal evidence of a strong methylation episignature. A review of the clinical and genetic features of the SETD2 patient group revealed that, as reported previously, there were phenotypic differences between patients with truncating mutations (n = 4, Luscan-Lumish syndrome; MIM:616831) and those with missense codon 1740 variants [p.Arg1740Trp (n = 4) and p.Arg1740Gln (n = 2)]. Both SETD2 subgroups demonstrated a methylation episignature, which was characterized by hypomethylation and hypermethylation events, respectively. Within the codon 1740 subgroup, both the methylation changes and clinical phenotype were more severe in those with p.Arg1740Trp variants. We also noted that two of 10 cases with a SETD2-NDD had developed a neoplasm. These findings reveal novel epigenotype-genotype-phenotype correlations in SETD2-NDDs and predict a gain-of-function mechanism for SETD2 codon 1740 pathogenic variants.


Asunto(s)
Cromatina , Trastornos del Neurodesarrollo , Humanos , Cromatina/genética , Metilación de ADN/genética , Mutación , Trastornos del Neurodesarrollo/genética , Estudios de Asociación Genética , Codón
2.
N Engl J Med ; 384(25): 2406-2417, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34161705

RESUMEN

BACKGROUND: Autophagy is the major intracellular degradation route in mammalian cells. Systemic ablation of core autophagy-related (ATG) genes in mice leads to embryonic or perinatal lethality, and conditional models show neurodegeneration. Impaired autophagy has been associated with a range of complex human diseases, yet congenital autophagy disorders are rare. METHODS: We performed a genetic, clinical, and neuroimaging analysis involving five families. Mechanistic investigations were conducted with the use of patient-derived fibroblasts, skeletal muscle-biopsy specimens, mouse embryonic fibroblasts, and yeast. RESULTS: We found deleterious, recessive variants in human ATG7, a core autophagy-related gene encoding a protein that is indispensable to classical degradative autophagy. Twelve patients from five families with distinct ATG7 variants had complex neurodevelopmental disorders with brain, muscle, and endocrine involvement. Patients had abnormalities of the cerebellum and corpus callosum and various degrees of facial dysmorphism. These patients have survived with impaired autophagic flux arising from a diminishment or absence of ATG7 protein. Although autophagic sequestration was markedly reduced, evidence of basal autophagy was readily identified in fibroblasts and skeletal muscle with loss of ATG7. Complementation of different model systems by deleterious ATG7 variants resulted in poor or absent autophagic function as compared with the reintroduction of wild-type ATG7. CONCLUSIONS: We identified several patients with a neurodevelopmental disorder who have survived with a severe loss or complete absence of ATG7, an essential effector enzyme for autophagy without a known functional paralogue. (Funded by the Wellcome Centre for Mitochondrial Research and others.).


Asunto(s)
Anomalías Múltiples/genética , Ataxia/genética , Proteína 7 Relacionada con la Autofagia/genética , Autofagia/genética , Discapacidades del Desarrollo/genética , Mutación Missense , Adolescente , Adulto , Autofagia/fisiología , Proteína 7 Relacionada con la Autofagia/fisiología , Células Cultivadas , Cerebelo/anomalías , Simulación por Computador , Cara/anomalías , Femenino , Fibroblastos , Genes Recesivos , Humanos , Lactante , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Malformaciones del Sistema Nervioso/genética , Linaje , Fenotipo
3.
J Med Genet ; 58(7): 453-464, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32631816

RESUMEN

BACKGROUND: Cilia are dynamic cellular extensions that generate and sense signals to orchestrate proper development and tissue homeostasis. They rely on the underlying polarisation of cells to participate in signalling. Cilia dysfunction is a well-known cause of several diseases that affect multiple organ systems including the kidneys, brain, heart, respiratory tract, skeleton and retina. METHODS: Among individuals from four unrelated families, we identified variants in discs large 5 (DLG5) that manifested in a variety of pathologies. In our proband, we also examined patient tissues. We depleted dlg5 in Xenopus tropicalis frog embryos to generate a loss-of-function model. Finally, we tested the pathogenicity of DLG5 patient variants through rescue experiments in the frog model. RESULTS: Patients with variants of DLG5 were found to have a variety of phenotypes including cystic kidneys, nephrotic syndrome, hydrocephalus, limb abnormalities, congenital heart disease and craniofacial malformations. We also observed a loss of cilia in cystic kidney tissue of our proband. Knockdown of dlg5 in Xenopus embryos recapitulated many of these phenotypes and resulted in a loss of cilia in multiple tissues. Unlike introduction of wildtype DLG5 in frog embryos depleted of dlg5, introduction of DLG5 patient variants was largely ineffective in restoring proper ciliation and tissue morphology in the kidney and brain suggesting that the variants were indeed detrimental to function. CONCLUSION: These findings in both patient tissues and Xenopus shed light on how mutations in DLG5 may lead to tissue-specific manifestations of disease. DLG5 is essential for cilia and many of the patient phenotypes are in the ciliopathy spectrum.


Asunto(s)
Ciliopatías/genética , Anomalías Congénitas/genética , Proteínas de la Membrana/genética , Mutación , Proteínas Supresoras de Tumor/genética , Animales , Encéfalo/patología , Niño , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Feto/anomalías , Técnicas de Silenciamiento del Gen , Proteínas Hedgehog/metabolismo , Humanos , Riñón/patología , Masculino , Linaje , Transducción de Señal , Secuenciación del Exoma , Xenopus
4.
Hum Mol Genet ; 28(20): 3391-3405, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31363758

RESUMEN

Reversible detyrosination of tubulin, the building block of microtubules, is crucial for neuronal physiology. Enzymes responsible for detyrosination were recently identified as complexes of vasohibins (VASHs) one or two with small VASH-binding protein (SVBP). Here we report three consanguineous families, each containing multiple individuals with biallelic inactivation of SVBP caused by truncating variants (p.Q28* and p.K13Nfs*18). Affected individuals show brain abnormalities with microcephaly, intellectual disability and delayed gross motor and speech development. Immunoblot testing in cells with pathogenic SVBP variants demonstrated that the encoded proteins were unstable and non-functional, resulting in a complete loss of VASH detyrosination activity. Svbp knockout mice exhibit drastic accumulation of tyrosinated tubulin and a reduction of detyrosinated tubulin in brain tissue. Similar alterations in tubulin tyrosination levels were observed in cultured neurons and associated with defects in axonal differentiation and architecture. Morphological analysis of the Svbp knockout mouse brains by anatomical magnetic resonance imaging showed a broad impact of SVBP loss, with a 7% brain volume decrease, numerous structural defects and a 30% reduction of some white matter tracts. Svbp knockout mice display behavioural defects, including mild hyperactivity, lower anxiety and impaired social behaviour. They do not, however, show prominent memory defects. Thus, SVBP-deficient mice recapitulate several features observed in human patients. Altogether, our data demonstrate that deleterious variants in SVBP cause this neurodevelopmental pathology, by leading to a major change in brain tubulin tyrosination and alteration of microtubule dynamics and neuron physiology.


Asunto(s)
Encéfalo/anomalías , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neuronas/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Proteínas Portadoras/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Femenino , Humanos , Immunoblotting , Imagen por Resonancia Magnética , Ratones , Microcefalia/genética , Microcefalia/metabolismo , Tirosina/metabolismo
5.
Am J Hum Genet ; 102(6): 1195-1203, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861108

RESUMEN

Next-generation sequencing is a powerful tool for the discovery of genes related to neurodevelopmental disorders (NDDs). Here, we report the identification of a distinct syndrome due to de novo or inherited heterozygous mutations in Tousled-like kinase 2 (TLK2) in 38 unrelated individuals and two affected mothers, using whole-exome and whole-genome sequencing technologies, matchmaker databases, and international collaborations. Affected individuals had a consistent phenotype, characterized by mild-borderline neurodevelopmental delay (86%), behavioral disorders (68%), severe gastro-intestinal problems (63%), and facial dysmorphism including blepharophimosis (82%), telecanthus (74%), prominent nasal bridge (68%), broad nasal tip (66%), thin vermilion of the upper lip (62%), and upslanting palpebral fissures (55%). Analysis of cell lines from three affected individuals showed that mutations act through a loss-of-function mechanism in at least two case subjects. Genotype-phenotype analysis and comparison of computationally modeled faces showed that phenotypes of these and other individuals with loss-of-function variants significantly overlapped with phenotypes of individuals with other variant types (missense and C-terminal truncating). This suggests that haploinsufficiency of TLK2 is the most likely underlying disease mechanism, leading to a consistent neurodevelopmental phenotype. This work illustrates the power of international data sharing, by the identification of 40 individuals from 26 different centers in 7 different countries, allowing the identification, clinical delineation, and genotype-phenotype evaluation of a distinct NDD caused by mutations in TLK2.


Asunto(s)
Estudios de Asociación Genética , Patrón de Herencia/genética , Mutación con Pérdida de Función/genética , Trastornos del Neurodesarrollo/genética , Proteínas Quinasas/genética , Adolescente , Adulto , Secuencia de Bases , Línea Celular , Niño , Preescolar , Facies , Femenino , Humanos , Lactante , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Translocación Genética , Adulto Joven
6.
Am J Hum Genet ; 102(3): 494-504, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29478781

RESUMEN

ATP synthase, H+ transporting, mitochondrial F1 complex, δ subunit (ATP5F1D; formerly ATP5D) is a subunit of mitochondrial ATP synthase and plays an important role in coupling proton translocation and ATP production. Here, we describe two individuals, each with homozygous missense variants in ATP5F1D, who presented with episodic lethargy, metabolic acidosis, 3-methylglutaconic aciduria, and hyperammonemia. Subject 1, homozygous for c.245C>T (p.Pro82Leu), presented with recurrent metabolic decompensation starting in the neonatal period, and subject 2, homozygous for c.317T>G (p.Val106Gly), presented with acute encephalopathy in childhood. Cultured skin fibroblasts from these individuals exhibited impaired assembly of F1FO ATP synthase and subsequent reduced complex V activity. Cells from subject 1 also exhibited a significant decrease in mitochondrial cristae. Knockdown of Drosophila ATPsynδ, the ATP5F1D homolog, in developing eyes and brains caused a near complete loss of the fly head, a phenotype that was fully rescued by wild-type human ATP5F1D. In contrast, expression of the ATP5F1D c.245C>T and c.317T>G variants rescued the head-size phenotype but recapitulated the eye and antennae defects seen in other genetic models of mitochondrial oxidative phosphorylation deficiency. Our data establish c.245C>T (p.Pro82Leu) and c.317T>G (p.Val106Gly) in ATP5F1D as pathogenic variants leading to a Mendelian mitochondrial disease featuring episodic metabolic decompensation.


Asunto(s)
Alelos , Enfermedades Metabólicas/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Mutación/genética , Subunidades de Proteína/genética , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Mutación con Pérdida de Función/genética , Masculino , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , ATPasas de Translocación de Protón Mitocondriales/química , Subunidades de Proteína/química
7.
Acta Neuropathol ; 141(3): 431-453, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33449170

RESUMEN

Mutations in the sarcomeric protein titin, encoded by TTN, are emerging as a common cause of myopathies. The diagnosis of a TTN-related myopathy is, however, often not straightforward due to clinico-pathological overlap with other myopathies and the prevalence of TTN variants in control populations. Here, we present a combined clinico-pathological, genetic and biophysical approach to the diagnosis of TTN-related myopathies and the pathogenicity ascertainment of TTN missense variants. We identified 30 patients with a primary TTN-related congenital myopathy (CM) and two truncating variants, or one truncating and one missense TTN variant, or homozygous for one TTN missense variant. We found that TTN-related myopathies show considerable overlap with other myopathies but are strongly suggested by a combination of certain clinico-pathological features. Presentation was typically at birth with the clinical course characterized by variable progression of weakness, contractures, scoliosis and respiratory symptoms but sparing of extraocular muscles. Cardiac involvement depended on the variant position. Our biophysical analyses demonstrated that missense mutations associated with CMs are strongly destabilizing and exert their effect when expressed on a truncating background or in homozygosity. We hypothesise that destabilizing TTN missense mutations phenocopy truncating variants and are a key pathogenic feature of recessive titinopathies that might be amenable to therapeutic intervention.


Asunto(s)
Conectina/genética , Miotonía Congénita/diagnóstico , Miotonía Congénita/genética , Miotonía Congénita/patología , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación Missense , Adulto Joven
8.
Am J Hum Genet ; 101(6): 1021-1033, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29220674

RESUMEN

ACTB encodes ß-actin, an abundant cytoskeletal housekeeping protein. In humans, postulated gain-of-function missense mutations cause Baraitser-Winter syndrome (BRWS), characterized by intellectual disability, cortical malformations, coloboma, sensorineural deafness, and typical facial features. To date, the consequences of loss-of-function ACTB mutations have not been proven conclusively. We describe heterozygous ACTB deletions and nonsense and frameshift mutations in 33 individuals with developmental delay, apparent intellectual disability, increased frequency of internal organ malformations (including those of the heart and the renal tract), growth retardation, and a recognizable facial gestalt (interrupted wavy eyebrows, dense eyelashes, wide nose, wide mouth, and a prominent chin) that is distinct from characteristics of individuals with BRWS. Strikingly, this spectrum overlaps with that of several chromatin-remodeling developmental disorders. In wild-type mouse embryos, ß-actin expression was prominent in the kidney, heart, and brain. ACTB mRNA expression levels in lymphoblastic lines and fibroblasts derived from affected individuals were decreased in comparison to those in control cells. Fibroblasts derived from an affected individual and ACTB siRNA knockdown in wild-type fibroblasts showed altered cell shape and migration, consistent with known roles of cytoplasmic ß-actin. We also demonstrate that ACTB haploinsufficiency leads to reduced cell proliferation, altered expression of cell-cycle genes, and decreased amounts of nuclear, but not cytoplasmic, ß-actin. In conclusion, we show that heterozygous loss-of-function ACTB mutations cause a distinct pleiotropic malformation syndrome with intellectual disability. Our biological studies suggest that a critically reduced amount of this protein alters cell shape, migration, proliferation, and gene expression to the detriment of brain, heart, and kidney development.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Discapacidades del Desarrollo/genética , Haploinsuficiencia/genética , Actinas/biosíntesis , Adolescente , Adulto , Anciano , Animales , Ciclo Celular/genética , Proliferación Celular/genética , Niño , Preescolar , Codón sin Sentido/genética , Coloboma/genética , Facies , Femenino , Mutación del Sistema de Lectura/genética , Eliminación de Gen , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/genética , Masculino , Malformaciones del Desarrollo Cortical/genética , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Adulto Joven
9.
Clin Genet ; 98(2): 191-197, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32530092

RESUMEN

Structural eye disorders are increasingly recognised as having a genetic basis, although current genetic testing is limited in its success. De novo missense variants in WDR37 are a recently described cause of a multisystemic syndromic disorder featuring ocular coloboma. This study characterises the phenotypic spectrum of this disorder and reports 2 de novo heterozygous variants (p.Thr115Ile, p.Ser119Tyr) in three unrelated Caucasian individuals. All had a clinical phenotype consisting of bilateral iris and retinal coloboma, developmental delay and additional, variable multisystem features. The variants fall within a highly conserved region upstream of the WD-repeat domains, within an apparent mutation cluster. Consistent with the literature, intellectual disability, structural eye disorders, epilepsy, congenital heart disease, genitorenal anomalies and dysmorphic facial features were observed. In addition, a broader developmental profile is reported with a more specific musculoskeletal phenotype described in association with the novel variant (p.Thr115Ile). We further expand the phenotypic spectrum of WDR37-related disorders to include those with milder developmental delay and strengthen the association of ocular coloboma and musculoskeletal features. We promote the inclusion of WDR37 on gene panels for intellectual disability, epilepsy and structural eye disorders.


Asunto(s)
Coloboma/genética , Oftalmopatías/genética , Discapacidad Intelectual/genética , Anomalías Musculoesqueléticas/genética , Proteínas Nucleares/genética , Adolescente , Adulto , Niño , Preescolar , Coloboma/complicaciones , Coloboma/patología , Epilepsia/complicaciones , Epilepsia/genética , Epilepsia/patología , Oftalmopatías/complicaciones , Oftalmopatías/patología , Femenino , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Anomalías Musculoesqueléticas/complicaciones , Anomalías Musculoesqueléticas/patología , Mutación/genética , Mutación Missense/genética , Fenotipo , Adulto Joven
10.
PLoS Genet ; 13(8): e1006957, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28859103

RESUMEN

Deletions at chromosome 2p25.3 are associated with a syndrome consisting of intellectual disability and obesity. The smallest region of overlap for deletions at 2p25.3 contains PXDN and MYT1L. MYT1L is expressed only within the brain in humans. We hypothesized that single nucleotide variants (SNVs) in MYT1L would cause a phenotype resembling deletion at 2p25.3. To examine this we sought MYT1L SNVs in exome sequencing data from 4, 296 parent-child trios. Further variants were identified through a genematcher-facilitated collaboration. We report 9 patients with MYT1L SNVs (4 loss of function and 5 missense). The phenotype of SNV carriers overlapped with that of 2p25.3 deletion carriers. To identify the transcriptomic consequences of MYT1L loss of function we used CRISPR-Cas9 to create a knockout cell line. Gene Ontology analysis in knockout cells demonstrated altered expression of genes that regulate gene expression and that are localized to the nucleus. These differentially expressed genes were enriched for OMIM disease ontology terms "mental retardation". To study the developmental effects of MYT1L loss of function we created a zebrafish knockdown using morpholinos. Knockdown zebrafish manifested loss of oxytocin expression in the preoptic neuroendocrine area. This study demonstrates that MYT1L variants are associated with syndromic obesity in humans. The mechanism is related to dysregulated expression of neurodevelopmental genes and altered development of the neuroendocrine hypothalamus.


Asunto(s)
Regulación de la Expresión Génica/genética , Hipotálamo/fisiología , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Obesidad/genética , Factores de Transcripción/genética , Adulto , Animales , Sistemas CRISPR-Cas , Línea Celular , Niño , Deleción Cromosómica , Cromosomas Humanos Par 2/genética , Femenino , Técnicas de Inactivación de Genes , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Discapacidad Intelectual/fisiopatología , Masculino , Mutación , Obesidad/fisiopatología , Polimorfismo de Nucleótido Simple/genética , Pez Cebra
11.
PLoS Genet ; 13(3): e1006683, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28346496

RESUMEN

Schinzel-Giedion syndrome (SGS) is a rare developmental disorder characterized by multiple malformations, severe neurological alterations and increased risk of malignancy. SGS is caused by de novo germline mutations clustering to a 12bp hotspot in exon 4 of SETBP1. Mutations in this hotspot disrupt a degron, a signal for the regulation of protein degradation, and lead to the accumulation of SETBP1 protein. Overlapping SETBP1 hotspot mutations have been observed recurrently as somatic events in leukemia. We collected clinical information of 47 SGS patients (including 26 novel cases) with germline SETBP1 mutations and of four individuals with a milder phenotype caused by de novo germline mutations adjacent to the SETBP1 hotspot. Different mutations within and around the SETBP1 hotspot have varying effects on SETBP1 stability and protein levels in vitro and in in silico modeling. Substitutions in SETBP1 residue I871 result in a weak increase in protein levels and mutations affecting this residue are significantly more frequent in SGS than in leukemia. On the other hand, substitutions in residue D868 lead to the largest increase in protein levels. Individuals with germline mutations affecting D868 have enhanced cell proliferation in vitro and higher incidence of cancer compared to patients with other germline SETBP1 mutations. Our findings substantiate that, despite their overlap, somatic SETBP1 mutations driving malignancy are more disruptive to the degron than germline SETBP1 mutations causing SGS. Additionally, this suggests that the functional threshold for the development of cancer driven by the disruption of the SETBP1 degron is higher than for the alteration in prenatal development in SGS. Drawing on previous studies of somatic SETBP1 mutations in leukemia, our results reveal a genotype-phenotype correlation in germline SETBP1 mutations spanning a molecular, cellular and clinical phenotype.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Portadoras/genética , Anomalías Craneofaciales/genética , Predisposición Genética a la Enfermedad/genética , Deformidades Congénitas de la Mano/genética , Neoplasias Hematológicas/genética , Discapacidad Intelectual/genética , Mutación , Uñas Malformadas/genética , Proteínas Nucleares/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Western Blotting , Proteínas Portadoras/metabolismo , Línea Celular , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Niño , Preescolar , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Femenino , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Mutación de Línea Germinal , Células HEK293 , Deformidades Congénitas de la Mano/metabolismo , Deformidades Congénitas de la Mano/patología , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Masculino , Uñas Malformadas/metabolismo , Uñas Malformadas/patología , Proteínas Nucleares/metabolismo , Fenotipo
12.
Hum Mol Genet ; 26(3): 519-526, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28053047

RESUMEN

Haploinsufficiency in DYRK1A is associated with a recognizable developmental syndrome, though the mechanism of action of pathogenic missense mutations is currently unclear. Here we present 19 de novo mutations in this gene, including five missense mutations, identified by the Deciphering Developmental Disorder study. Protein structural analysis reveals that the missense mutations are either close to the ATP or peptide binding-sites within the kinase domain, or are important for protein stability, suggesting they lead to a loss of the protein's function mechanism. Furthermore, there is some correlation between the magnitude of the change and the severity of the resultant phenotype. A comparison of the distribution of the pathogenic mutations along the length of DYRK1A with that of natural variants, as found in the ExAC database, confirms that mutations in the N-terminal end of the kinase domain are more disruptive of protein function. In particular, pathogenic mutations occur in significantly closer proximity to the ATP and the substrate peptide than the natural variants. Overall, we suggest that de novo dominant mutations in DYRK1A account for nearly 0.5% of severe developmental disorders due to substantially reduced kinase function.


Asunto(s)
Trastorno Autístico/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Trastorno Autístico/patología , Discapacidades del Desarrollo/fisiopatología , Femenino , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/patología , Masculino , Mutación , Mutación Missense , Linaje , Fenotipo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas/química , Relación Estructura-Actividad , Quinasas DyrK
13.
Am J Hum Genet ; 99(4): 860-876, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693233

RESUMEN

Mutations in SLC25A4 encoding the mitochondrial ADP/ATP carrier AAC1 are well-recognized causes of mitochondrial disease. Several heterozygous SLC25A4 mutations cause adult-onset autosomal-dominant progressive external ophthalmoplegia associated with multiple mitochondrial DNA deletions, whereas recessive SLC25A4 mutations cause childhood-onset mitochondrial myopathy and cardiomyopathy. Here, we describe the identification by whole-exome sequencing of seven probands harboring dominant, de novo SLC25A4 mutations. All affected individuals presented at birth, were ventilator dependent and, where tested, revealed severe combined mitochondrial respiratory chain deficiencies associated with a marked loss of mitochondrial DNA copy number in skeletal muscle. Strikingly, an identical c.239G>A (p.Arg80His) mutation was present in four of the seven subjects, and the other three case subjects harbored the same c.703C>G (p.Arg235Gly) mutation. Analysis of skeletal muscle revealed a marked decrease of AAC1 protein levels and loss of respiratory chain complexes containing mitochondrial DNA-encoded subunits. We show that both recombinant AAC1 mutant proteins are severely impaired in ADP/ATP transport, affecting most likely the substrate binding and mechanics of the carrier, respectively. This highly reduced capacity for transport probably affects mitochondrial DNA maintenance and in turn respiration, causing a severe energy crisis. The confirmation of the pathogenicity of these de novo SLC25A4 mutations highlights a third distinct clinical phenotype associated with mutation of this gene and demonstrates that early-onset mitochondrial disease can be caused by recurrent de novo mutations, which has significant implications for the application and analysis of whole-exome sequencing data in mitochondrial disease.


Asunto(s)
Translocador 1 del Nucleótido Adenina/genética , Variaciones en el Número de Copia de ADN/genética , ADN Mitocondrial/genética , Genes Dominantes/genética , Enfermedades Mitocondriales/genética , Mutación , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Edad de Inicio , Arilamina N-Acetiltransferasa/genética , Niño , Preescolar , Transporte de Electrón/genética , Exoma/genética , Femenino , Humanos , Lactante , Recién Nacido , Isoenzimas/genética , Masculino , Enfermedades Mitocondriales/patología , Músculo Esquelético/metabolismo
14.
Am J Hum Genet ; 96(3): 412-24, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25728776

RESUMEN

Non-homologous end joining (NHEJ) is a key cellular process ensuring genome integrity. Mutations in several components of the NHEJ pathway have been identified, often associated with severe combined immunodeficiency (SCID), consistent with the requirement for NHEJ during V(D)J recombination to ensure diversity of the adaptive immune system. In contrast, we have recently found that biallelic mutations in LIG4 are a common cause of microcephalic primordial dwarfism (MPD), a phenotype characterized by prenatal-onset extreme global growth failure. Here we provide definitive molecular genetic evidence supported by biochemical, cellular, and immunological data for mutations in XRCC4, encoding the obligate binding partner of LIG4, causing MPD. We report the identification of biallelic mutations in XRCC4 in five families. Biochemical and cellular studies demonstrate that these alterations substantially decrease XRCC4 protein levels leading to reduced cellular ligase IV activity. Consequently, NHEJ-dependent repair of ionizing-radiation-induced DNA double-strand breaks is compromised in XRCC4 cells. Similarly, immunoglobulin junctional diversification is impaired in cells. However, immunoglobulin levels are normal, and individuals lack overt signs of immunodeficiency. Additionally, in contrast to individuals with LIG4 mutations, pancytopenia leading to bone marrow failure has not been observed. Hence, alterations that alter different NHEJ proteins give rise to a phenotypic spectrum, from SCID to extreme growth failure, with deficiencies in certain key components of this repair pathway predominantly exhibiting growth deficits, reflecting differential developmental requirements for NHEJ proteins to support growth and immune maturation.


Asunto(s)
Proteínas de Unión al ADN/genética , Enanismo Hipofisario/genética , Enanismo/genética , Microcefalia/genética , Mutación , Alelos , Secuencia de Aminoácidos , Niño , Preescolar , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP) , ADN Ligasas/genética , ADN Ligasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Electroforesis en Gel de Campo Pulsado , Exoma , Facies , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Fenotipo , Conformación Proteica , Inmunodeficiencia Combinada Grave/genética
15.
Mol Genet Metab ; 123(1): 28-42, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29331171

RESUMEN

BACKGROUND: Mitochondrial diseases, a group of multi-systemic disorders often characterized by tissue-specific phenotypes, are usually progressive and fatal disorders resulting from defects in oxidative phosphorylation. MTO1 (Mitochondrial tRNA Translation Optimization 1), an evolutionarily conserved protein expressed in high-energy demand tissues has been linked to human early-onset combined oxidative phosphorylation deficiency associated with hypertrophic cardiomyopathy, often referred to as combined oxidative phosphorylation deficiency-10 (COXPD10). MATERIAL AND METHODS: Thirty five cases of MTO1 deficiency were identified and reviewed through international collaboration. The cases of two female siblings, who presented at 1 and 2years of life with seizures, global developmental delay, hypotonia, elevated lactate and complex I and IV deficiency on muscle biopsy but without cardiomyopathy, are presented in detail. RESULTS: For the description of phenotypic features, the denominator varies as the literature was insufficient to allow for complete ascertainment of all data for the 35 cases. An extensive review of all known MTO1 deficiency cases revealed the most common features at presentation to be lactic acidosis (LA) (21/34; 62% cases) and hypertrophic cardiomyopathy (15/34; 44% cases). Eventually lactic acidosis and hypertrophic cardiomyopathy are described in 35/35 (100%) and 27/34 (79%) of patients with MTO1 deficiency, respectively; with global developmental delay/intellectual disability present in 28/29 (97%), feeding difficulties in 17/35 (49%), failure to thrive in 12/35 (34%), seizures in 12/35 (34%), optic atrophy in 11/21 (52%) and ataxia in 7/34 (21%). There are 19 different pathogenic MTO1 variants identified in these 35 cases: one splice-site, 3 frameshift and 15 missense variants. None have bi-allelic variants that completely inactivate MTO1; however, patients where one variant is truncating (i.e. frameshift) while the second one is a missense appear to have a more severe, even fatal, phenotype. These data suggest that complete loss of MTO1 is not viable. A ketogenic diet may have exerted a favourable effect on seizures in 2/5 patients. CONCLUSION: MTO1 deficiency is lethal in some but not all cases, and a genotype-phenotype relation is suggested. Aside from lactic acidosis and cardiomyopathy, developmental delay and other phenotypic features affecting multiple organ systems are often present in these patients, suggesting a broader spectrum than hitherto reported. The diagnosis should be suspected on clinical features and the presence of markers of mitochondrial dysfunction in body fluids, especially low residual complex I, III and IV activity in muscle. Molecular confirmation is required and targeted genomic testing may be the most efficient approach. Although subjective clinical improvement was observed in a small number of patients on therapies such as ketogenic diet and dichloroacetate, no evidence-based effective therapy exists.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Encefalopatía Hepática/genética , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/genética , Adolescente , Biopsia , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/metabolismo , Niño , Preescolar , Femenino , Mutación del Sistema de Lectura , Encefalopatía Hepática/diagnóstico por imagen , Encefalopatía Hepática/fisiopatología , Humanos , Lactante , Recién Nacido , Masculino , Errores Innatos del Metabolismo/diagnóstico por imagen , Errores Innatos del Metabolismo/fisiopatología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Fosforilación Oxidativa , Proteínas de Unión al ARN
16.
Neuropediatrics ; 49(5): 330-338, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29940663

RESUMEN

BACKGROUND: Primary microcephaly and profound global developmental delay have been considered the core clinical phenotype in patients with bi-allelic PRUNE1 mutations. METHODS: Linkage analysis and whole-exome sequencing (WES) in a multiplex family and extraction of further cases from a WES repository containing 571 children with severe developmental disabilities and neurologic symptoms. RESULTS: We identified bi-allelic PRUNE1 mutations in twelve children from six unrelated families. All patients who survived beyond the first 6 months of life had early-onset global developmental delay, bilateral spastic paresis, dysphagia and difficult-to-treat seizures, while congenital or later-evolving microcephaly was not a consistent finding. Brain MRI showed variable anomalies with progressive cerebral and cerebellar atrophies and T2-hyperintense brain stem lesions. Peripheral neuropathy was documented in five cases. Disease course was progressive in all patients and eight children died in the first or early second decade of life. In addition to the previously reported missense mutation p.(Asp106Asn), we observed a novel homozygous missense variant p.(Leu172Pro) and a homozygous contiguous gene deletion encompassing most of the PRUNE1 gene and part of the neighboring BNIPL gene. CONCLUSIONS: PRUNE1 deficiency causes severe early-onset disease affecting the central and peripheral nervous systems. Microcephaly is probably not a universal feature.


Asunto(s)
Encéfalo/patología , Discapacidades del Desarrollo , Progresión de la Enfermedad , Epilepsia Refractaria , Errores Innatos del Metabolismo , Microcefalia , Espasticidad Muscular , Paresia , Monoéster Fosfórico Hidrolasas , Niño , Preescolar , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/genética , Epilepsia Refractaria/etiología , Epilepsia Refractaria/genética , Femenino , Ligamiento Genético , Humanos , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Errores Innatos del Metabolismo/fisiopatología , Microcefalia/etiología , Microcefalia/genética , Espasticidad Muscular/etiología , Espasticidad Muscular/genética , Mutación Missense , Paresia/etiología , Paresia/genética , Linaje , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Secuenciación del Exoma
17.
Prenat Diagn ; 38(1): 33-43, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096039

RESUMEN

OBJECTIVE: Rare genetic disorders resulting in prenatal or neonatal death are genetically heterogeneous, but testing is often limited by the availability of fetal DNA, leaving couples without a potential prenatal test for future pregnancies. We describe our novel strategy of exome sequencing parental DNA samples to diagnose recessive monogenic disorders in an audit of the first 50 couples referred. METHOD: Exome sequencing was carried out in a consecutive series of 50 couples who had 1 or more pregnancies affected with a lethal or prenatal-onset disorder. In all cases, there was insufficient DNA for exome sequencing of the affected fetus. Heterozygous rare variants (MAF < 0.001) in the same gene in both parents were selected for analysis. Likely, disease-causing variants were tested in fetal DNA to confirm co-segregation. RESULTS: Parental exome analysis identified heterozygous pathogenic (or likely pathogenic) variants in 24 different genes in 26/50 couples (52%). Where 2 or more fetuses were affected, a genetic diagnosis was obtained in 18/29 cases (62%). In most cases, the clinical features were typical of the disorder, but in others, they result from a hypomorphic variant or represent the most severe form of a variable phenotypic spectrum. CONCLUSION: We conclude that exome sequencing of parental samples is a powerful strategy with high clinical utility for the genetic diagnosis of lethal or prenatal-onset recessive disorders. © 2017 The Authors Prenatal Diagnosis published by John Wiley & Sons Ltd.


Asunto(s)
Anomalías Congénitas/genética , Secuenciación del Exoma , Enfermedades Genéticas Congénitas/diagnóstico , Padres , Diagnóstico Prenatal/métodos , Femenino , Genes Recesivos , Humanos , Masculino , Embarazo
18.
Neurogenetics ; 18(4): 227-235, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29075935

RESUMEN

Mitochondrial diseases are characterised by clinical, molecular and functional heterogeneity, reflecting their bi-genomic control. The nuclear gene GFM2 encodes mtEFG2, a protein with an essential role during the termination stage of mitochondrial translation. We present here two unrelated patients harbouring different and previously unreported compound heterozygous (c.569G>A, p.(Arg190Gln); c.636delA, p.(Glu213Argfs*3)) and homozygous (c.275A>C, p.(Tyr92Ser)) recessive variants in GFM2 identified by whole exome sequencing (WES) together with histochemical and biochemical findings to support the diagnoses of pathological GFM2 variants in each case. Both patients presented similarly in early childhood with global developmental delay, raised CSF lactate and abnormalities on cranial MRI. Sanger sequencing of familial samples confirmed the segregation of bi-allelic GFM2 variants with disease, while investigations into steady-state mitochondrial protein levels revealed respiratory chain subunit defects and loss of mtEFG2 protein in muscle. These data demonstrate the effects of defective mtEFG2 function, caused by previously unreported variants, confirming pathogenicity and expanding the clinical phenotypes associated with GFM2 variants.


Asunto(s)
Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Factor G de Elongación Peptídica/genética , Niño , Exoma/genética , Femenino , Homocigoto , Humanos , Masculino , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Mutación/genética , Linaje , Fenotipo
19.
J Inherit Metab Dis ; 40(1): 121-130, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27696117

RESUMEN

Mitochondrial diseases collectively represent one of the most heterogeneous group of metabolic disorders. Symptoms can manifest at any age, presenting with isolated or multiple-organ involvement. Advances in next-generation sequencing strategies have greatly enhanced the diagnosis of patients with mitochondrial disease, particularly where a mitochondrial aetiology is strongly suspected yet OXPHOS activities in biopsied tissue samples appear normal. We used whole exome sequencing (WES) to identify the molecular basis of an early-onset mitochondrial syndrome-pathogenic biallelic variants in the HTRA2 gene, encoding a mitochondria-localised serine protease-in five subjects from two unrelated families characterised by seizures, neutropenia, hypotonia and cardio-respiratory problems. A unifying feature in all affected children was 3-methylglutaconic aciduria (3-MGA-uria), a common biochemical marker observed in some patients with mitochondrial dysfunction. Although functional studies of HTRA2 subjects' fibroblasts and skeletal muscle homogenates showed severely decreased levels of mutant HTRA2 protein, the structural subunits and complexes of the mitochondrial respiratory chain appeared normal. We did detect a profound defect in OPA1 processing in HTRA2-deficient fibroblasts, suggesting a role for HTRA2 in the regulation of mitochondrial dynamics and OPA1 proteolysis. In addition, investigated subject fibroblasts were more susceptible to apoptotic insults. Our data support recent studies that described important functions for HTRA2 in programmed cell death and confirm that patients with genetically-unresolved 3-MGA-uria should be screened by WES with pathogenic variants in the HTRA2 gene prioritised for further analysis.


Asunto(s)
Variación Genética/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas/genética , Errores Innatos del Metabolismo/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Muerte Celular/genética , Células Cultivadas , Niño , Exoma/genética , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas Mitocondriales/genética , Músculo Esquelético/metabolismo , Serina Proteasas/genética , Síndrome
20.
Hum Mol Genet ; 23(10): 2752-68, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24381304

RESUMEN

Rare copy number variants (CNVs) disrupting ASTN2 or both ASTN2 and TRIM32 have been reported at 9q33.1 by genome-wide studies in a few individuals with neurodevelopmental disorders (NDDs). The vertebrate-specific astrotactins, ASTN2 and its paralog ASTN1, have key roles in glial-guided neuronal migration during brain development. To determine the prevalence of astrotactin mutations and delineate their associated phenotypic spectrum, we screened ASTN2/TRIM32 and ASTN1 (1q25.2) for exonic CNVs in clinical microarray data from 89 985 individuals across 10 sites, including 64 114 NDD subjects. In this clinical dataset, we identified 46 deletions and 12 duplications affecting ASTN2. Deletions of ASTN1 were much rarer. Deletions near the 3' terminus of ASTN2, which would disrupt all transcript isoforms (a subset of these deletions also included TRIM32), were significantly enriched in the NDD subjects (P = 0.002) compared with 44 085 population-based controls. Frequent phenotypes observed in individuals with such deletions include autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), speech delay, anxiety and obsessive compulsive disorder (OCD). The 3'-terminal ASTN2 deletions were significantly enriched compared with controls in males with NDDs, but not in females. Upon quantifying ASTN2 human brain RNA, we observed shorter isoforms expressed from an alternative transcription start site of recent evolutionary origin near the 3' end. Spatiotemporal expression profiling in the human brain revealed consistently high ASTN1 expression while ASTN2 expression peaked in the early embryonic neocortex and postnatal cerebellar cortex. Our findings shed new light on the role of the astrotactins in psychopathology and their interplay in human neurodevelopment.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Trastornos Generalizados del Desarrollo Infantil/genética , Glicoproteínas/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Preescolar , Cromosomas Humanos Par 9 , Variaciones en el Número de Copia de ADN , Exones , Femenino , Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glicoproteínas/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Fenotipo , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Riesgo , Eliminación de Secuencia , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA