Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 162(2): 375-390, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26186191

RESUMO

Autism spectrum disorder (ASD) is a disorder of brain development. Most cases lack a clear etiology or genetic basis, and the difficulty of re-enacting human brain development has precluded understanding of ASD pathophysiology. Here we use three-dimensional neural cultures (organoids) derived from induced pluripotent stem cells (iPSCs) to investigate neurodevelopmental alterations in individuals with severe idiopathic ASD. While no known underlying genomic mutation could be identified, transcriptome and gene network analyses revealed upregulation of genes involved in cell proliferation, neuronal differentiation, and synaptic assembly. ASD-derived organoids exhibit an accelerated cell cycle and overproduction of GABAergic inhibitory neurons. Using RNA interference, we show that overexpression of the transcription factor FOXG1 is responsible for the overproduction of GABAergic neurons. Altered expression of gene network modules and FOXG1 are positively correlated with symptom severity. Our data suggest that a shift toward GABAergic neuron fate caused by FOXG1 is a developmental precursor of ASD.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/patologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Telencéfalo/embriologia , Feminino , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas , Masculino , Megalencefalia/genética , Megalencefalia/patologia , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Organoides/patologia , Telencéfalo/patologia
2.
Cell ; 158(2): 263-276, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24998929

RESUMO

Autism spectrum disorder (ASD) is a heterogeneous disease in which efforts to define subtypes behaviorally have met with limited success. Hypothesizing that genetically based subtype identification may prove more productive, we resequenced the ASD-associated gene CHD8 in 3,730 children with developmental delay or ASD. We identified a total of 15 independent mutations; no truncating events were identified in 8,792 controls, including 2,289 unaffected siblings. In addition to a high likelihood of an ASD diagnosis among patients bearing CHD8 mutations, characteristics enriched in this group included macrocephaly, distinct faces, and gastrointestinal complaints. chd8 disruption in zebrafish recapitulates features of the human phenotype, including increased head size as a result of expansion of the forebrain/midbrain and impairment of gastrointestinal motility due to a reduction in postmitotic enteric neurons. Our findings indicate that CHD8 disruptions define a distinct ASD subtype and reveal unexpected comorbidities between brain development and enteric innervation.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , Adolescente , Sequência de Aminoácidos , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Criança , Transtornos Globais do Desenvolvimento Infantil/classificação , Transtornos Globais do Desenvolvimento Infantil/patologia , Pré-Escolar , Proteínas de Ligação a DNA/metabolismo , Feminino , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiopatologia , Humanos , Macaca mulatta , Masculino , Megalencefalia/patologia , Dados de Sequência Molecular , Mutação , Alinhamento de Sequência , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Am J Med Genet A ; 194(7): e63585, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38459620

RESUMO

Germline gain of function variations in the AKT3 gene cause brain overgrowth syndrome with megalencephaly and diffuse bilateral cortical malformations. Here we report a child with megalencephaly, who is a carrier of a novel heterozygous missense variant in the AKT3 gene NM_005465.7:c.964G>T,p.Asp322Tyr. The phenotype of this patient is associated with pituitary deficiencies diagnosed at 2 years of age: growth hormone (GH) deficiency responsible for growth delay and central hypothyroidism. After 6 months of GH treatment, intracranial hypertension was noted, confirmed by the observation of papilledema and increased intracranial pressure, requiring the initiation of acetazolamide treatment and the discontinuation of GH treatment. This is the second reported patient described with megalencephaly and AKT3 gene variant associated with GH deficiency . Other endocrine disorders have also been reported in few cases with hypothyroidism and hypoglycemia. Pituitary deficiency may be a part of the of megalencephaly phenotype secondary to germline variant in the AKT3 gene. Special attention should be paid to growth in these patients and search for endocrine deficiency is necessary in case of growth retardation or hypoglycemia.


Assuntos
Mutação em Linhagem Germinativa , Megalencefalia , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas c-akt , Humanos , Megalencefalia/genética , Megalencefalia/patologia , Mutação de Sentido Incorreto/genética , Proteínas Proto-Oncogênicas c-akt/genética , Mutação em Linhagem Germinativa/genética , Masculino , Pré-Escolar , Fenótipo , Hipotireoidismo/genética , Hipotireoidismo/patologia , Hipotireoidismo/complicações , Feminino , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/genética
4.
Genes Chromosomes Cancer ; 62(12): 703-709, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37395289

RESUMO

Heterozygous germline or somatic variants in AKT3 gene can cause isolated malformations of cortical development (MCDs) such as focal cortical dysplasia, megalencephaly (MEG), Hemimegalencephaly (HME), dysplastic megalencephaly, and syndromic forms like megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome, and megalencephaly-capillary malformation syndrome. This report describes a new case of HME and capillary malformation caused by a somatic AKT3 variant that differs from the common p.E17K variant described in literature. The patient's skin biopsy from the angiomatous region revealed an heterozygous likely pathogenic variant AKT3:c.241_243dup, p.(T81dup) that may affect the binding domain and downstream pathways. Compared to previously reported cases with a common E17K mosaic variant, the phenotype is milder and patients showed segmental overgrowth, an uncommon characteristic in AKT3 variant cases. These findings suggest that the severity of the disease may be influenced not only by the level of mosaicism but also by the type of variant. This report expands the phenotypic spectrum associated with AKT3 variants and highlights the importance of genomic analysis in patients with capillary malformation and MCDs.


Assuntos
Megalencefalia , Malformações Vasculares , Humanos , Mutação , Megalencefalia/genética , Megalencefalia/patologia , Malformações Vasculares/genética , Fenótipo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
5.
Semin Cell Dev Biol ; 111: 15-22, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32741653

RESUMO

Genetic studies identified multiple mutations associated with malformations of cortical development (MCD) in humans. When analyzing the underlying mechanisms in non-human experimental models it became increasingly evident, that these mutations accumulate in genes, which functions evolutionary progressed from rodents to humans resulting in an incomplete reflection of the molecular and cellular alterations in these models. Human brain organoids derived from human pluripotent stem cells resemble early aspects of human brain development to a remarkable extent making them an attractive model to investigate MCD. Here we review how human brain organoids enable the generation of fundamental new insight about the underlying pathomechanisms of MCD. We show how phenotypic features of these diseases are reflected in human brain organoids and discuss challenges and future considerations but also limitations for the use of human brain organoids to model human brain development and associated disorders.


Assuntos
Córtex Cerebral/metabolismo , Lisencefalia/genética , Megalencefalia/genética , Microcefalia/genética , Proteínas do Tecido Nervoso/genética , Organoides/metabolismo , Heterotopia Nodular Periventricular/genética , Diferenciação Celular , Córtex Cerebral/anormalidades , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiopatologia , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Regulação da Expressão Gênica , Humanos , Lisencefalia/metabolismo , Lisencefalia/patologia , Lisencefalia/fisiopatologia , Megalencefalia/metabolismo , Megalencefalia/patologia , Megalencefalia/fisiopatologia , Microcefalia/metabolismo , Microcefalia/patologia , Microcefalia/fisiopatologia , Modelos Biológicos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Organoides/patologia , Heterotopia Nodular Periventricular/metabolismo , Heterotopia Nodular Periventricular/patologia , Heterotopia Nodular Periventricular/fisiopatologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Cultura Primária de Células
6.
J Biol Chem ; 296: 100313, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33482199

RESUMO

Functional genomic approaches have facilitated the discovery of rare genetic disorders and improved efforts to decipher their underlying etiology. PPP2R5D-related disorder is an early childhood onset condition characterized by intellectual disability, hypotonia, autism-spectrum disorder, macrocephaly, and dysmorphic features. The disorder is caused by de novo single nucleotide changes in PPP2R5D, which generate heterozygous dominant missense variants. PPP2R5D is known to encode a B'-type (B'56δ) regulatory subunit of a PP2A-serine/threonine phosphatase. To help elucidate the molecular mechanisms altered in PPP2R5D-related disorder, we used a CRISPR-single-base editor to generate HEK-293 cells in which a single transition (c.1258G>A) was introduced into one allele, precisely recapitulating a clinically relevant E420K variant. Unbiased quantitative proteomic and phosphoproteomic analyses of endogenously expressed proteins revealed heterozygous-dominant changes in kinase/phosphatase signaling. These data combined with orthogonal validation studies revealed a previously unrecognized interaction of PPP2R5D with AKT in human cells, leading to constitutively active AKT-mTOR signaling, increased cell size, and uncoordinated cellular growth in E420K-variant cells. Rapamycin reduced cell size and dose-dependently reduced RPS6 phosphorylation in E420K-variant cells, suggesting that inhibition of mTOR1 can suppress both the observed RPS6 hyperphosphorylation and increased cell size. Together, our findings provide a deeper understanding of PPP2R5D and insight into how the E420K-variant alters signaling networks influenced by PPP2R5D. Our comprehensive approach, which combines precise genome editing, isobaric tandem mass tag labeling of peptides generated from endogenously expressed proteins, and concurrent liquid chromatography-mass spectrometry (LC-MS3), also provides a roadmap that can be used to rapidly explore the etiologies of additional genetic disorders.


Assuntos
Doenças Genéticas Inatas/genética , Predisposição Genética para Doença , Proteína Fosfatase 2/genética , Proteômica , Serina-Treonina Quinases TOR/genética , Transtorno Autístico/genética , Transtorno Autístico/patologia , Sistemas CRISPR-Cas/genética , Doenças Genéticas Inatas/patologia , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Megalencefalia/genética , Megalencefalia/patologia , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas c-akt/genética
7.
Am J Med Genet A ; 188(7): 2246-2250, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35338599

RESUMO

Noonan syndrome-like disorder with loose anagen hair (NSLH) is a rare disease characterized by typical features of Noonan syndrome with additional findings of relative or absolute macrocephaly, loose anagen hair, and a higher incidence of intellectual disability. NSLH1 is caused by a heterozygous mutation in the SHOC2 gene on chromosome 10q25, and NLSH2 is caused by a heterozygous mutation in the Protein phosphatase one catalytic subunit beta (PPP1CB) gene on chromosome 2p23. Protein phosphatase1 (PP1), encoded by PPP1CB, forms a complex with SHOC2 and dephosphorylates RAFs, which results in activation of the signaling cascade and contribution to Noonan syndrome pathogenesis. Here, we report two genetically confirmed Japanese patients with NSLH2 having the same de novo mutation in PPP1CB presenting prominent-hyperteloric-appearing eyes and a tall forehead similar to individuals carrying a mutation in PPP1CB, c.146C > G; p.Pro49Arg, which is different from typical facial features of Noonan syndrome. They also showed short stature, absolute macrocephaly, and loose anagen hair like NSLH1: however, growth hormone deficiency often seen in NSLH1 caused by SHOC2 mutation was absent. Although a number of Noonan syndrome and NSLH1 patients have shown blunted or no response to GH therapy, linear growth was promoted by recombinant human growth hormone (rhGH) in one of our patients. Since another NSLH2 patient with good response to rhGH treatment was reported, rhGH therapy may be effective in patients with NSLH2.


Assuntos
Anormalidades Múltiplas , Hormônio do Crescimento Humano , Síndrome dos Cabelos Anágenos Frouxos , Megalencefalia , Síndrome de Noonan , Anormalidades Múltiplas/patologia , Cabelo/patologia , Hormônio do Crescimento Humano/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Japão , Síndrome dos Cabelos Anágenos Frouxos/diagnóstico , Síndrome dos Cabelos Anágenos Frouxos/genética , Síndrome dos Cabelos Anágenos Frouxos/patologia , Megalencefalia/patologia , Mutação , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Síndrome de Noonan/patologia
8.
Ann Neurol ; 88(6): 1077-1094, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32856318

RESUMO

OBJECTIVE: Mutations in phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) complex have been associated with a broad spectrum of brain and organ overgrowth syndromes. For example, mutations in phosphatidylinositol-3-kinase regulatory subunit 2 (PIK3R2) have been identified in human patients with megalencephaly polymicrogyria polydactyly hydrocephalus (MPPH) syndrome, which includes brain overgrowth. To better understand the pathogenesis of PIK3R2-related mutations, we have developed and characterized a murine model. METHODS: We generated a knock-in mouse model for the most common human PIK3R2 mutation, p.G373R (p.G367R in mice) using CRISPR/Cas9. The mouse phenotypes, including brain size, seizure activity, cortical lamination, cell proliferation/size/density, interneuron migration, and PI3K pathway activation, were analyzed using standard methodologies. For human patients with PIK3R2 mutations, clinical data (occipitofrontal circumference [OFC] and epilepsy) were retrospectively obtained from our clinical records (published / unpublished). RESULTS: The PI3K-AKT pathway was hyperactivated in these mice, confirming the p.G367R mutation is an activating mutation in vivo. Similar to human patients with PIK3R2 mutations, these mice have enlarged brains. We found cell size to be increased but not cell numbers. The embryonic brain showed mild defects in cortical lamination, although not observed in the mature brain. Furthermore, electroencephalogram (EEG) recordings from mutant mice showed background slowing and rare seizures, again similar to our observations in human patients. INTERPRETATION: We have generated a PIK3R2 mouse model that exhibits megalencephaly and EEG changes, both of which overlap with human patients. Our data provide novel insight into the pathogenesis of the human disease caused by PIK3R2 p.G373R mutation. We anticipate this model will be valuable in testing therapeutic options for human patients with MPPH. ANN NEUROL 2020;88:1077-1094.


Assuntos
Encéfalo/patologia , Megalencefalia/patologia , Fosfatidilinositol 3-Quinases/genética , Convulsões/genética , Animais , Eletroencefalografia , Epilepsia/diagnóstico , Feminino , Técnicas de Introdução de Genes/métodos , Humanos , Masculino , Megalencefalia/genética , Camundongos , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais/genética , Síndrome
9.
Am J Med Genet A ; 185(9): 2719-2738, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34087052

RESUMO

Cyclin D2 (CCND2) is a critical cell cycle regulator and key member of the cyclin D2-CDK4 (DC) complex. De novo variants of CCND2 clustering in the distal part of the protein have been identified as pathogenic causes of brain overgrowth (megalencephaly, MEG) and severe cortical malformations in children including the megalencephaly-polymicrogyria-polydactyly-hydrocephalus (MPPH) syndrome. Megalencephaly-associated CCND2 variants are localized to the terminal exon and result in accumulation of degradation-resistant protein. We identified five individuals from three unrelated families with novel variants in the proximal region of CCND2 associated with microcephaly, mildly simplified cortical gyral pattern, symmetric short stature, and mild developmental delay. Identified variants include de novo frameshift variants and a dominantly inherited stop-gain variant segregating with the phenotype. This is the first reported association between proximal CCND2 variants and microcephaly, to our knowledge. This series expands the phenotypic spectrum of CCND2-related disorders and suggests that distinct classes of CCND2 variants are associated with reciprocal effects on human brain growth (microcephaly and megalencephaly due to possible loss or gain of protein function, respectively), adding to the growing paradigm of inverse phenotypes due to dysregulation of key brain growth genes.


Assuntos
Encéfalo/anormalidades , Ciclina D2/genética , Hidrocefalia/patologia , Megalencefalia/patologia , Mutação , Polidactilia/patologia , Polimicrogiria/patologia , Adolescente , Adulto , Criança , Feminino , Humanos , Hidrocefalia/genética , Lactente , Masculino , Megalencefalia/genética , Polidactilia/genética , Polimicrogiria/genética
10.
Am J Med Genet A ; 185(7): 2084-2093, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33973697

RESUMO

Nuclear factor I A (NFIA) is a transcription factor that belongs to the NFI family. Truncating variants or intragenic deletion of the NFIA gene are known to cause the human neurodevelopmental disorder known as NFIA-related disorder, but no patient heterozygous for a missense mutation has been reported. Here, we document two unrelated patients with typical phenotypic features of the NFIA-related disorder who shared a missense variant p.Lys125Glu (K125E) in the NFIA gene. Patient 1 was a 6-year-old female with global developmental delay, corpus callosum anomaly, macrocephaly, and dysmorphic facial features. Patient 2 was a 14-month-old male with corpus callosum anomaly and macrocephaly. By using Drosophila and zebrafish models, we functionally evaluated the effect of the K125E substitution. Ectopic expression of wild-type human NFIA in Drosophila caused developmental defects such as eye malformation and premature death, while that of human NFIA K125E variant allele did not. nfia-deficient zebrafish embryos showed defects of midline-crossing axons in the midbrain/hindbrain boundary. This impairment of commissural neurons was rescued by expression of wild-type human NFIA, but not by that of mutant variant harboring K125E substitution. In accordance with these in vivo functional analyses, we showed that the K125E mutation impaired the transcriptional regulation of HES1 promoter in cultured cells. Taken together, we concluded that the K125E variant in the NFIA gene is a loss-of-function mutation.


Assuntos
Predisposição Genética para Doença , Megalencefalia/genética , Fatores de Transcrição NFI/genética , Transtornos do Neurodesenvolvimento/genética , Alelos , Substituição de Aminoácidos/genética , Animais , Criança , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Drosophila/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Lactente , Masculino , Megalencefalia/patologia , Mutação de Sentido Incorreto/genética , Transtornos do Neurodesenvolvimento/patologia , Peixe-Zebra/genética
11.
Am J Med Genet A ; 185(5): 1538-1543, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33595182

RESUMO

We describe a patient with Temple syndrome resulting from maternal uniparental disomy of chromosome 14 who also has low-level mosaicism for trisomy 14. UPD was initially suspected when SNP microarray analysis detected a large region of homozygosity on chromosome 14 and the patient's clinical features were consistent with the phenotype of upd(14)mat. However, SNP arrays cannot prove UPD, as homozygosity may also result from identity by descent. Methylation assays diagnose imprinting disorders such as Prader-Willi, Angelman and Temple syndromes; they detect methylation defects that occur in imprinted loci, which have parent-of-origin-specific expression and have the advantage of making a diagnosis without parental samples. However, in this patient methylation analysis using endpoint PCR detected biparental inheritance. Therefore, sequencing analysis was performed and diagnosed upd(14)mat. Re-examination of the microarray suggested that the explanation for the discrepancy between the array and methylation testing was low-level mosaicism for trisomy 14 and fluorescence in situ hybridization testing detected a trisomic cell line. Thus, this patient's Temple syndrome is a result of a maternal M1 error, which gave a trisomic zygote, followed by loss of the paternal chromosome 14 in an early mitotic division to give maternal UPD with low-level mosaicism for trisomy 14. The methylation assay detected the paternal allele in the trisomic line. The diagnostic failure of the methylation assay in this patient highlights a significant shortcoming of methylation endpoint analysis, especially for Temple syndrome, and underscores the need to use other methods in cases with mosaicism.


Assuntos
Megalencefalia/diagnóstico , Síndrome de Prader-Willi/diagnóstico , Trissomia/genética , Dissomia Uniparental/genética , Cromossomos Humanos Par 14/genética , Metilação de DNA/genética , Feminino , Impressão Genômica/genética , Humanos , Hibridização in Situ Fluorescente , Megalencefalia/genética , Megalencefalia/patologia , Análise em Microsséries , Mosaicismo , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/patologia , Trissomia/patologia , Dissomia Uniparental/patologia
12.
Am J Med Genet A ; 185(12): 3877-3883, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34357686

RESUMO

Kleefstra syndrome (KS) is a rare autosomic dominant genetic disorder caused by euchromatic histone methyltransferase 1 (EHMT1) alterations. Patients mainly present with moderate to severe intellectual disability, a severe delay in/or absence of speech, autism spectrum disorder, childhood hypotonia, neuropsychiatric anomalies, and distinctive dysmorphic features. Here, we report the cases of a male and a female, two younger siblings of three, with asymptomatic parents. An EHMT1 new mutation was identified. Both presented with a typical core phenotype. Some specific features were noted, such as macrocephaly (previously reported) and enuresis (not yet described). Parental analysis identified the mutation in the mosaic state in the father. Reverse phenotyping enabled us to highlight the pauci phenotype features of inguinal hernia, azoospermia, and possible behavioral disorders. This allowed us to adapt his follow-up and genetic counseling for the family. Our three reported cases provide a new description of KS with an intragenic EHMT1 mutation, whereas in the literature most reported cases have EHMT1 deletions. Moreover, in the areas of next-generation sequencing and trio techniques with parental segregation, it is important to remain cautious about disregarding variants based on an autosomal recessive hypothesis.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Craniofaciais/genética , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Histona-Lisina N-Metiltransferase/genética , Deficiência Intelectual/genética , Megalencefalia/genética , Anormalidades Múltiplas/patologia , Adolescente , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 9/genética , Hibridização Genômica Comparativa , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/patologia , Feminino , Aconselhamento Genético , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/patologia , Masculino , Megalencefalia/patologia , Mosaicismo , Mutação , Fenótipo , Adulto Jovem
13.
Am J Med Genet A ; 185(10): 3083-3091, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34061450

RESUMO

KCNT2 variants resulting in substitutions affecting the Arg190 residue have been shown to cause epileptic encephalopathy and a recognizable facial gestalt. We report two additional individuals with intellectual disability, dysmorphic features, hypertrichosis, macrocephaly and the same de novo KCNT2 missense variants affecting the Arg190 residue as previously described. Notably, neither patient has epilepsy. Homology modeling of these missense variants revealed that they are likely to disrupt the stabilization of a closed channel conformation of KCNT2 resulting in a constitutively open state. This is the first report of pathogenic variants in KCNT2 causing a developmental phenotype without epilepsy.


Assuntos
Epilepsia/genética , Hipertricose/genética , Deficiência Intelectual/genética , Megalencefalia/genética , Canais de Potássio Ativados por Sódio/genética , Adolescente , Arginina/genética , Criança , Pré-Escolar , Epilepsia/diagnóstico , Epilepsia/patologia , Feminino , Predisposição Genética para Doença , Humanos , Hipertricose/diagnóstico , Hipertricose/diagnóstico por imagem , Hipertricose/patologia , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/patologia , Megalencefalia/diagnóstico por imagem , Megalencefalia/patologia , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Mutação de Sentido Incorreto/genética , Fenótipo
14.
Am J Med Genet A ; 185(11): 3485-3493, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34477286

RESUMO

Bachmann-Bupp syndrome (BABS) is a rare syndrome caused by gain-of-function variants in the C-terminus of ornithine decarboxylase (ODC coded by the ODC1 gene). BABS is characterized by developmental delay, macrocephaly, macrosomia, and an unusual pattern of non-congenital alopecia. Recent diagnosis of four more BABS patients provides further characterization of the phenotype of this syndrome including late-onset seizures in the oldest reported patient at 23 years of age, representing the first report for this phenotype in BABS. Neuroimaging abnormalities continue to be an inconsistent feature of the syndrome. This may be related to the yet unknown impact of ODC/polyamine dysregulation on the developing brain in this syndrome. Variants continue to cluster, providing support to a universal biochemical mechanism related to elevated ODC protein, enzyme activity, and abnormalities in polyamine levels. Recommendations for medical management can now be suggested as well as the potential for targeted molecular or metabolic testing when encountering this unique phenotype. The natural history of this syndrome will evolve with difluoromethylornithine (DFMO) therapy and raise new questions for further study and understanding.


Assuntos
Alopecia/genética , Deficiências do Desenvolvimento/genética , Transportadores de Ácidos Dicarboxílicos/genética , Megalencefalia/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Adolescente , Adulto , Alopecia/diagnóstico , Alopecia/tratamento farmacológico , Alopecia/patologia , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Criança , Pré-Escolar , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/tratamento farmacológico , Eflornitina/uso terapêutico , Feminino , Predisposição Genética para Doença , Humanos , Lactente , Recém-Nascido , Masculino , Megalencefalia/diagnóstico por imagem , Megalencefalia/tratamento farmacológico , Megalencefalia/patologia , Neuroimagem , Fenótipo , Poliaminas/metabolismo , Convulsões/diagnóstico , Convulsões/tratamento farmacológico , Convulsões/genética , Convulsões/patologia , Adulto Jovem
15.
J Inherit Metab Dis ; 44(3): 629-638, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33274439

RESUMO

Glutaric aciduria type 1 (GA1) is a rare neurometabolic disorder, caused by inherited deficiency of glutaryl-CoA dehydrogenase, mostly affecting the brain. Early identification by newborn screening (NBS) significantly improves neurologic outcome. It has remained unclear whether recommended therapy, particular low lysine diet, is safe or negatively affects anthropometric long-term outcome. This national prospective, observational, multi-centre study included 79 patients identified by NBS and investigated effects of interventional and non-interventional parameters on body weight, body length, body mass index (BMI) and head circumference as well as neurological parameters. Adherence to recommended maintenance and emergency treatment (ET) had a positive impact on neurologic outcome and allowed normal anthropometric development until adulthood. In contrast, non-adherence to ET, resulting in increased risk of dystonia, had a negative impact on body weight (mean SDS -1.07; P = .023) and body length (mean SDS -1.34; P = -.016). Consistently, longitudinal analysis showed a negative influence of severe dystonia on weight and length development over time (P < .001). Macrocephaly was more often found in female (mean SDS 0.56) than in male patients (mean SDS -0.20; P = .049), and also in individuals with high excreter phenotype (mean SDS 0.44) compared to low excreter patients (mean SDS -0.68; P = .016). In GA1, recommended long-term treatment is effective and allows for normal anthropometric long-term development up to adolescence, with gender- and excreter type-specific variations. Delayed ET and severe movement disorder result in poor anthropometric outcome.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/terapia , Glutaril-CoA Desidrogenase/deficiência , Adolescente , Antropometria , Estatura , Índice de Massa Corporal , Peso Corporal , Criança , Pré-Escolar , Distonia/patologia , Tratamento de Emergência , Feminino , Alemanha , Humanos , Lactente , Recém-Nascido , Masculino , Megalencefalia/patologia , Triagem Neonatal , Estudos Prospectivos , Fatores Sexuais , Adulto Jovem
16.
BMC Med Genet ; 21(1): 219, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33167890

RESUMO

BACKGROUND: Damaging variants in TRIO have been associated with moderate to severe neurodevelopmental disorders in humans. While recent work has delineated the positional effect of missense variation on the resulting phenotype, the clinical spectrum associated with loss-of-function variation has yet to be fully defined. CASE PRESENTATION: We report on two probands with novel loss-of-function variants in TRIO. Patient 1 presents with a severe neurodevelopmental disorder and macrocephaly. The TRIO variant is inherited from his affected mother. Patient 2 presents with moderate developmental delays, microcephaly, and cutis aplasia with a frameshift variant of unknown inheritance. CONCLUSIONS: We describe two patients with neurodevelopmental disorder, macro/microcephaly, and cutis aplasia in one patient. Both patients have loss-of-function variants, helping to further characterize how these types of variants affect the phenotypic spectrum associated with TRIO. We also present the third reported case of autosomal dominant inheritance of a damaging variant in TRIO.


Assuntos
Transtorno Autístico/genética , Deficiências do Desenvolvimento/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Megalencefalia/genética , Microcefalia/genética , Proteínas Serina-Treonina Quinases/genética , Adolescente , Transtorno Autístico/diagnóstico , Transtorno Autístico/patologia , Pré-Escolar , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/patologia , Feminino , Mutação da Fase de Leitura , Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/deficiência , Humanos , Padrões de Herança , Mutação com Perda de Função , Masculino , Megalencefalia/diagnóstico , Megalencefalia/patologia , Microcefalia/diagnóstico , Microcefalia/patologia , Linhagem , Proteínas Serina-Treonina Quinases/deficiência
17.
Clin Genet ; 97(3): 467-476, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31972898

RESUMO

The proximal 19p13.3 microdeletion/microduplication (prox19p13.3del/dup) syndrome is a recently described disorder with common clinical features including developmental delay, intellectual disability, speech delay, facial dysmorphic features with ear defects, anomalies of the hands and feet, umbilical hernia and hypotonia. While deletions are associated with macrocephaly, patients with duplications have microcephaly. The smallest region of overlap in multiple patients (113.5 kb) included three genes and one pseudogene, with a suggested major role of PIAS4 in determination of the phenotype and head size in these patients. Here, we refine the prox19p13.3del/dup with four additional patients: two with microdeletions, one with microduplication and one family with single-nucleotide nonsense variant in PIAS4. The patient with the PIAS4 loss of function variant displayed a phenotype quite similar to deletion patients -including the macrocephaly and many other core features of the syndrome. Patient's SNV was inherited from her mother who is similarly affected. Thus, our data indicate that PIAS4 is a major contributor to the proximal 19p13.3del/dup syndrome phenotype. In summary, we report the first patient with a pathogenic variant in PIAS4- and three additional rearrangements at the proximal 19p13.3 locus. These observations add further evidence about the molecular basis of this microdeletion/microduplication syndrome.


Assuntos
Anormalidades Múltiplas/genética , Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas Inibidoras de STAT Ativados/genética , Anormalidades Múltiplas/patologia , Criança , Deleção Cromossômica , Duplicação Cromossômica/genética , Cromossomos Humanos Par 19/genética , Hibridização Genômica Comparativa , Deficiências do Desenvolvimento/patologia , Feminino , Humanos , Deficiência Intelectual/patologia , Masculino , Megalencefalia/genética , Megalencefalia/patologia , Microcefalia/patologia , Fenótipo
18.
Clin Genet ; 97(6): 890-901, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32266967

RESUMO

Primrose syndrome (PS; MIM# 259050) is characterized by intellectual disability (ID), macrocephaly, unusual facial features (frontal bossing, deeply set eyes, down-slanting palpebral fissures), calcified external ears, sparse body hair and distal muscle wasting. The syndrome is caused by de novo heterozygous missense variants in ZBTB20. Most of the 29 published patients are adults as characteristics appear more recognizable with age. We present 13 hitherto unpublished individuals and summarize the clinical and molecular findings in all 42 patients. Several signs and symptoms of PS develop during childhood, but the cardinal features, such as calcification of the external ears, cystic bone lesions, muscle wasting, and contractures typically develop between 10 and 16 years of age. Biochemically, anemia and increased alpha-fetoprotein levels are often present. Two adult males with PS developed a testicular tumor. Although PS should be regarded as a progressive entity, there are no indications that cognition becomes more impaired with age. No obvious genotype-phenotype correlation is present. A subgroup of patients with ZBTB20 variants may be associated with mild, nonspecific ID. Metabolic investigations suggest a disturbed mitochondrial fatty acid oxidation. We suggest a regular surveillance in all adult males with PS until it is clear whether or not there is a truly elevated risk of testicular cancer.


Assuntos
Anormalidades Múltiplas/genética , Calcinose/genética , Otopatias/genética , Predisposição Genética para Doença , Deficiência Intelectual/genética , Megalencefalia/genética , Atrofia Muscular/genética , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética , 3-Hidroxiacil-CoA Desidrogenases/genética , Anormalidades Múltiplas/patologia , Acetil-CoA C-Aciltransferase/genética , Adolescente , Adulto , Calcinose/patologia , Isomerases de Ligação Dupla Carbono-Carbono/genética , Criança , Pré-Escolar , Otopatias/patologia , Enoil-CoA Hidratase/genética , Face/anormalidades , Feminino , Estudos de Associação Genética , Heterozigoto , Humanos , Lactente , Deficiência Intelectual/patologia , Masculino , Megalencefalia/patologia , Pessoa de Meia-Idade , Mitocôndrias/genética , Mitocôndrias/patologia , Atrofia Muscular/patologia , Mutação , Mutação de Sentido Incorreto/genética , Fenótipo , Racemases e Epimerases/genética , Neoplasias Testiculares , Adulto Jovem
19.
Am J Med Genet A ; 182(4): 762-767, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31999056

RESUMO

Alteration of the KPTN gene, responsible for the coding of kaptin (a protein involved in actin cytoskeletal dynamics), causes a syndrome characterized by macrocephaly, neurodevelopmental delay and epileptic seizures. We report the first Brazilian case of KPTN gene variation, previously described in nine subjects from four interlinked families from an Amish community in Ohio, two Estonian siblings and a 9-year-old boy from Kansas City. We report a case of KPTN-related syndrome in a 5-year-old child which presented macrocephaly, muscular hypotonia, and global development delay. The neurological examination revealed below-expected performance in coordination and balance tests, dyspraxia, and hand-mouth synkinesia. Expressive language was characterized by phono-articulatory imprecision, abundance of phonological processes and morphosyntactic immaturity. Neuropsychological assessment revealed intellectual disability with impairment of verbal and executive functions. Exome sequencing was performed. Analysis revealed a homozygous 2-nucleotide duplication c.597_598dup p.(Ser200Ilefs*55) in the KPTN gene, which is predicted to lead to a translational frameshift and formation of a premature stop codon. The phenotypic profile is similar to the cases described in the other families. Presence of macrocephaly and delayed development indicate the possibility of KPTN gene variation. Genetic testing should be carried out at an early stage in order to reach a timely diagnosis.


Assuntos
Deficiências do Desenvolvimento/patologia , Homozigoto , Deficiência Intelectual/patologia , Megalencefalia/patologia , Proteínas dos Microfilamentos/genética , Hipotonia Muscular/patologia , Mutação , Brasil , Pré-Escolar , Deficiências do Desenvolvimento/genética , Feminino , Humanos , Deficiência Intelectual/genética , Megalencefalia/genética , Hipotonia Muscular/genética , Fenótipo , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA