Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Am J Hum Genet ; 109(2): 345-360, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35045343

RESUMO

Free oligosaccharides (fOSs) are soluble oligosaccharide species generated during N-glycosylation of proteins. Although little is known about fOS metabolism, the recent identification of NGLY1 deficiency, a congenital disorder of deglycosylation (CDDG) caused by loss of function of an enzyme involved in fOS metabolism, has elicited increased interest in fOS processing. The catabolism of fOSs has been linked to the activity of a specific cytosolic mannosidase, MAN2C1, which cleaves α1,2-, α1,3-, and α1,6-mannose residues. In this study, we report the clinical, biochemical, and molecular features of six individuals, including two fetuses, with bi-allelic pathogenic variants in MAN2C1; the individuals are from four different families. These individuals exhibit dysmorphic facial features, congenital anomalies such as tongue hamartoma, variable degrees of intellectual disability, and brain anomalies including polymicrogyria, interhemispheric cysts, hypothalamic hamartoma, callosal anomalies, and hypoplasia of brainstem and cerebellar vermis. Complementation experiments with isogenic MAN2C1-KO HAP1 cells confirm the pathogenicity of three of the identified MAN2C1 variants. We further demonstrate that MAN2C1 variants lead to accumulation and delay in the processing of fOSs in proband-derived cells. These results emphasize the involvement of MAN2C1 in human neurodevelopmental disease and the importance of fOS catabolism.


Assuntos
Cistos do Sistema Nervoso Central/genética , Defeitos Congênitos da Glicosilação/genética , Hamartoma/genética , Deficiência Intelectual/genética , Oligossacarídeos/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/deficiência , Polimicrogiria/genética , alfa-Manosidase/genética , Adolescente , Alelos , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Linhagem Celular Tumoral , Cistos do Sistema Nervoso Central/metabolismo , Cistos do Sistema Nervoso Central/patologia , Vermis Cerebelar/metabolismo , Vermis Cerebelar/patologia , Criança , Pré-Escolar , Defeitos Congênitos da Glicosilação/metabolismo , Defeitos Congênitos da Glicosilação/patologia , Feminino , Feto , Glicosilação , Hamartoma/metabolismo , Hamartoma/patologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Leucócitos/metabolismo , Leucócitos/patologia , Masculino , Manose/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/metabolismo , Polimicrogiria/metabolismo , Polimicrogiria/patologia , Língua/metabolismo , Língua/patologia , alfa-Manosidase/deficiência
2.
J Med Genet ; 61(2): 103-108, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-37879892

RESUMO

The Aristaless-related homeobox (ARX) gene is located on the X chromosome and encodes a transcription factor that is essential for brain development. While the clinical spectrum of ARX-related disorders is well described in males, from X linked lissencephaly with abnormal genitalia syndrome to syndromic and non-syndromic intellectual disability (ID), its phenotypic delineation in females is incomplete. Carrier females in ARX families are usually asymptomatic, but ID has been reported in some of them, as well as in others with de novo variants. In this study, we collected the clinical and molecular data of 10 unpublished female patients with de novo ARX pathogenic variants and reviewed the data of 63 females from the literature with either de novo variants (n=10), inherited variants (n=33) or variants of unknown inheritance (n=20). Altogether, the clinical spectrum of females with heterozygous pathogenic ARX variants is broad: 42.5% are asymptomatic, 16.4% have isolated agenesis of the corpus callosum (ACC) or mild symptoms (learning disabilities, autism spectrum disorder, drug-responsive epilepsy) without ID, whereas 41% present with a severe phenotype (ie, ID or developmental and epileptic encephalopathy (DEE)). The ID/DEE phenotype was significantly more prevalent in females carrying de novo variants (75%, n=15/20) versus in those carrying inherited variants (27.3%, n=9/33). ACC was observed in 66.7% (n=24/36) of females who underwent a brain MRI. By refining the clinical spectrum of females carrying ARX pathogenic variants, we show that ID is a frequent sign in females with this X linked condition.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Masculino , Humanos , Feminino , Genes Homeobox , Proteínas de Homeodomínio/genética , Transtorno do Espectro Autista/genética , Mutação/genética , Fatores de Transcrição/genética , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Fenótipo , Agenesia do Corpo Caloso/genética
3.
Am J Med Genet A ; 194(5): e63510, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38135344

RESUMO

Aicardi-Goutières syndrome (AGS) is a genetic interferonopathy classically characterized by early onset of severe neurologic injury with basal ganglia calcifications, white matter abnormalities, and progressive cerebral atrophy, along with lymphocytosis and raised interferon alpha (INFα) in the cerebrospinal fluid (CSF). Here, we report a 31/2 year-old patient born with prenatal onset AGS, first manifesting as intra-uterine growth retardation. Cranial ultrasonography and cerebral MRI revealed ventriculomegaly and periventricular and basal ganglia calcifications, along with cerebral atrophy. Perinatal infections and known metabolic disorders were excluded. Both CSF lymphocytosis and raised INFα were present. Molecular analysis disclosed two already described compound heterozygous pathogenic variants in TREX1 (c. 309dup, p.(Thr104Hisfs*53) and c. 506G > A, p.(Arg169His)). The evolution was marked by severe global developmental delay with progressive microcephaly. Promptly, the patient developed irritability, quadri-paretic dyskinetic movements, and subsequently tonic seizures. Sensorineural hearing loss was detected as well as glaucoma. Initially, he was symptomatically treated with trihexyphenidyl followed by levetiracetam and topiramate. At age 22 months, baricitinib (0.4 mg/kg/day) was introduced, leading to normal serum INFα levels. Clinically, dyskinetic movements significantly decreased as well as irritability and sleep disturbance. We confirmed that baricitinib was a useful treatment with no major side effect.


Assuntos
Doenças Autoimunes do Sistema Nervoso , Azetidinas , Doenças dos Gânglios da Base , Calcinose , Inibidores de Janus Quinases , Linfocitose , Malformações do Sistema Nervoso , Purinas , Pirazóis , Sulfonamidas , Masculino , Gravidez , Feminino , Humanos , Lactente , Linfocitose/líquido cefalorraquidiano , Linfocitose/genética , Malformações do Sistema Nervoso/tratamento farmacológico , Malformações do Sistema Nervoso/genética , Doenças dos Gânglios da Base/diagnóstico , Doenças dos Gânglios da Base/tratamento farmacológico , Doenças dos Gânglios da Base/genética , Doenças Autoimunes do Sistema Nervoso/tratamento farmacológico , Doenças Autoimunes do Sistema Nervoso/genética , Calcinose/genética , Atrofia
4.
Dev Med Child Neurol ; 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38394064

RESUMO

Malformations of cortical development (MCDs) represent a heterogeneous spectrum of disorders characterized by atypical development of the cerebral cortex. MCDs are most often diagnosed on the basis of imaging, although subtle lesions, such as focal cortical dysplasia, may only be revealed on neuropathology. Different subtypes have been defined, including lissencephaly, heterotopia, cobblestone malformation, polymicrogyria, and dysgyria. Many MCDs are of genetic origin, although acquired factors, such as congenital cytomegalovirus infections and twinning sequence, can lead to similar phenotypes. In this narrative review, we provide an overview of the diagnostic approach to MCDs, which is illustrated with clinical vignettes, on diagnostic pitfalls such as somatic mosaicism and consanguinity, and recognizable phenotypes on imaging, such as tubulinopathies, the lissencephaly spectrum, tuberous sclerosis complex, and FLNA-related periventricular nodular heterotopia.

5.
J Med Genet ; 60(2): 183-192, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35393335

RESUMO

BACKGROUND: Malformations of cortical development (MCDs) have been reported in a subset of patients with pathogenic heterozygous variants in GRIN1 or GRIN2B, genes which encode for subunits of the N-methyl-D-aspartate receptor (NMDAR). The aim of this study was to further define the phenotypic spectrum of NMDAR-related MCDs. METHODS: We report the clinical, radiological and molecular features of 7 new patients and review data on 18 previously reported individuals with NMDAR-related MCDs. Neuropathological findings for two individuals with heterozygous variants in GRIN1 are presented. We report the clinical and neuropathological features of one additional individual with homozygous pathogenic variants in GRIN1. RESULTS: Heterozygous variants in GRIN1 and GRIN2B were associated with overlapping severe clinical and imaging features, including global developmental delay, epilepsy, diffuse dysgyria, dysmorphic basal ganglia and hippocampi. Neuropathological examination in two fetuses with heterozygous GRIN1 variants suggests that proliferation as well as radial and tangential neuronal migration are impaired. In addition, we show that neuronal migration is also impaired by homozygous GRIN1 variants in an individual with microcephaly with simplified gyral pattern. CONCLUSION: These findings expand our understanding of the clinical and imaging features of the 'NMDARopathy' spectrum and contribute to our understanding of the likely underlying pathogenic mechanisms leading to MCD in these patients.


Assuntos
Epilepsia , Microcefalia , Receptores de N-Metil-D-Aspartato , Humanos , Heterozigoto , Homozigoto , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética
6.
Brain ; 145(12): 4232-4245, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-35139179

RESUMO

RAC1 is a highly conserved Rho GTPase critical for many cellular and developmental processes. De novo missense RAC1 variants cause a highly variable neurodevelopmental disorder. Some of these variants have previously been shown to have a dominant negative effect. Most previously reported patients with this disorder have either severe microcephaly or severe macrocephaly. Here, we describe eight patients with pathogenic missense RAC1 variants affecting residues between Q61 and R68 within the switch II region of RAC1. These patients display variable combinations of developmental delay, intellectual disability, brain anomalies such as polymicrogyria and cardiovascular defects with normocephaly or relatively milder micro- or macrocephaly. Pulldown assays, NIH3T3 fibroblast spreading assays and staining for activated PAK1/2/3 and WAVE2 suggest that these variants increase RAC1 activity and over-activate downstream signalling targets. Axons of neurons isolated from Drosophila embryos expressing the most common of the activating variants are significantly shorter, with an increased density of filopodial protrusions. In vivo, these embryos exhibit frequent defects in axonal organization. Class IV dendritic arborization neurons expressing this variant exhibit a significant reduction in the total area of the dendritic arbour, increased branching and failure of self-avoidance. RNAi knock down of the WAVE regulatory complex component Cyfip significantly rescues these morphological defects. These results establish that activating substitutions affecting residues Q61-R68 within the switch II region of RAC1 cause a developmental syndrome. Our findings reveal that these variants cause altered downstream signalling, resulting in abnormal neuronal morphology and reveal the WAVE regulatory complex/Arp2/3 pathway as a possible therapeutic target for activating RAC1 variants. These insights also have the potential to inform the mechanism and therapy for other disorders caused by variants in genes encoding other Rho GTPases, their regulators and downstream effectors.


Assuntos
Megalencefalia , Transtornos do Neurodesenvolvimento , Proteínas rac1 de Ligação ao GTP , Animais , Camundongos , Megalencefalia/genética , Transtornos do Neurodesenvolvimento/genética , Neurônios , Células NIH 3T3 , Transdução de Sinais/genética
7.
Genet Med ; 24(9): 1941-1951, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35678782

RESUMO

PURPOSE: WNK3 kinase (PRKWNK3) has been implicated in the development and function of the brain via its regulation of the cation-chloride cotransporters, but the role of WNK3 in human development is unknown. METHOD: We ascertained exome or genome sequences of individuals with rare familial or sporadic forms of intellectual disability (ID). RESULTS: We identified a total of 6 different maternally-inherited, hemizygous, 3 loss-of-function or 3 pathogenic missense variants (p.Pro204Arg, p.Leu300Ser, p.Glu607Val) in WNK3 in 14 male individuals from 6 unrelated families. Affected individuals had ID with variable presence of epilepsy and structural brain defects. WNK3 variants cosegregated with the disease in 3 different families with multiple affected individuals. This included 1 large family previously diagnosed with X-linked Prieto syndrome. WNK3 pathogenic missense variants localize to the catalytic domain and impede the inhibitory phosphorylation of the neuronal-specific chloride cotransporter KCC2 at threonine 1007, a site critically regulated during the development of synaptic inhibition. CONCLUSION: Pathogenic WNK3 variants cause a rare form of human X-linked ID with variable epilepsy and structural brain abnormalities and implicate impaired phospho-regulation of KCC2 as a pathogenic mechanism.


Assuntos
Deficiência Intelectual Ligada ao Cromossomo X , Proteínas Serina-Treonina Quinases , Simportadores , Encéfalo/anormalidades , Domínio Catalítico/genética , Hemizigoto , Humanos , Mutação com Perda de Função , Masculino , Herança Materna/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Mutação de Sentido Incorreto , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Simportadores/metabolismo
8.
J Med Genet ; 58(1): 33-40, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32571897

RESUMO

BACKGROUND: Variants in genes belonging to the tubulin superfamily account for a heterogeneous spectrum of brain malformations referred to as tubulinopathies. Variants in TUBB2A have been reported in 10 patients with a broad spectrum of brain imaging features, ranging from a normal cortex to polymicrogyria, while one patient has been reported with progressive atrophy of the cerebellar vermis. METHODS: In order to further refine the phenotypical spectrum associated with TUBB2A, clinical and imaging features of 12 patients with pathogenic TUBB2A variants, recruited via the international network of the authors, were reviewed. RESULTS: We report 12 patients with eight novel and one recurrent variants spread throughout the TUBB2A gene but encoding for amino acids clustering at the protein surface. Eleven patients (91.7%) developed seizures in early life. All patients suffered from intellectual disability, and 11 patients had severe motor developmental delay, with 4 patients (36.4 %) being non-ambulatory. The cerebral cortex was normal in five individuals and showed dysgyria of variable severity in seven patients. Associated brain malformations were less frequent in TUBB2A patients compared with other tubulinopathies. None of the patients had progressive cerebellar atrophy. CONCLUSION: The imaging phenotype associated with pathogenic variants in TUBB2A is highly variable, ranging from a normal cortex to extensive dysgyria with associated brain malformations. For recurrent variants, no clear genotype-phenotype correlations could be established, suggesting the role of additional modifiers.


Assuntos
Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Malformações do Sistema Nervoso/genética , Polimicrogiria/genética , Tubulina (Proteína)/genética , Adolescente , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Vermis Cerebelar/diagnóstico por imagem , Vermis Cerebelar/patologia , Criança , Pré-Escolar , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/patologia , Feminino , Predisposição Genética para Doença , Humanos , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/patologia , Masculino , Mutação de Sentido Incorreto/genética , Malformações do Sistema Nervoso/diagnóstico por imagem , Malformações do Sistema Nervoso/patologia , Neuroimagem/métodos , Fenótipo , Polimicrogiria/diagnóstico por imagem , Polimicrogiria/patologia , Tubulina (Proteína)/deficiência , Adulto Jovem
9.
J Med Genet ; 58(10): 712-716, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32820033

RESUMO

OBJECTIVE: To determine the potential disease association between variants in LMBRD2 and complex multisystem neurological and developmental delay phenotypes. METHODS: Here we describe a series of de novo missense variants in LMBRD2 in 10 unrelated individuals with overlapping features. Exome sequencing or genome sequencing was performed on all individuals, and the cohort was assembled through GeneMatcher. RESULTS: LMBRD2 encodes an evolutionary ancient and widely expressed transmembrane protein with no known disease association, although two paralogues are involved in developmental and metabolic disorders. Exome or genome sequencing revealed rare de novo LMBRD2 missense variants in 10 individuals with developmental delay, intellectual disability, thin corpus callosum, microcephaly and seizures. We identified five unique variants and two recurrent variants, c.1448G>A (p.Arg483His) in three cases and c.367T>C (p.Trp123Arg) in two cases. All variants are absent from population allele frequency databases, and most are predicted to be deleterious by multiple in silico damage-prediction algorithms. CONCLUSION: These findings indicate that rare de novo variants in LMBRD2 can lead to a previously unrecognised early-onset neurodevelopmental disorder. Further investigation of individuals harbouring LMBRD2 variants may lead to a better understanding of the function of this ubiquitously expressed gene.


Assuntos
Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Transtornos das Habilidades Motoras/diagnóstico , Transtornos das Habilidades Motoras/genética , Mutação de Sentido Incorreto , Malformações do Sistema Nervoso/diagnóstico , Malformações do Sistema Nervoso/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Alelos , Substituição de Aminoácidos , Estudos de Coortes , Predisposição Genética para Doença , Genótipo , Humanos , Fenótipo
10.
Ann Neurol ; 88(2): 348-362, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32515017

RESUMO

OBJECTIVE: Pathogenic variants in SCN3A, encoding the voltage-gated sodium channel subunit Nav1.3, cause severe childhood onset epilepsy and malformation of cortical development. Here, we define the spectrum of clinical, genetic, and neuroimaging features of SCN3A-related neurodevelopmental disorder. METHODS: Patients were ascertained via an international collaborative network. We compared sodium channels containing wild-type versus variant Nav1.3 subunits coexpressed with ß1 and ß2 subunits using whole-cell voltage clamp electrophysiological recordings in a heterologous mammalian system (HEK-293T cells). RESULTS: Of 22 patients with pathogenic SCN3A variants, most had treatment-resistant epilepsy beginning in the first year of life (16/21, 76%; median onset, 2 weeks), with severe or profound developmental delay (15/20, 75%). Many, but not all (15/19, 79%), exhibited malformations of cortical development. Pathogenic variants clustered in transmembrane segments 4 to 6 of domains II to IV. Most pathogenic missense variants tested (10/11, 91%) displayed gain of channel function, with increased persistent current and/or a leftward shift in the voltage dependence of activation, and all variants associated with malformation of cortical development exhibited gain of channel function. One variant (p.Ile1468Arg) exhibited mixed effects, with gain and partial loss of function. Two variants demonstrated loss of channel function. INTERPRETATION: Our study defines SCN3A-related neurodevelopmental disorder along a spectrum of severity, but typically including epilepsy and severe or profound developmental delay/intellectual disability. Malformations of cortical development are a characteristic feature of this unusual channelopathy syndrome, present in >75% of affected individuals. Gain of function at the channel level in developing neurons is likely an important mechanism of disease pathogenesis. ANN NEUROL 2020;88:348-362.


Assuntos
Encéfalo/diagnóstico por imagem , Epilepsia/diagnóstico por imagem , Epilepsia/genética , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Transtornos do Neurodesenvolvimento/diagnóstico por imagem , Transtornos do Neurodesenvolvimento/genética , Canais de Sódio/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Feto/diagnóstico por imagem , Variação Genética/genética , Células HEK293 , Humanos , Lactente , Masculino
11.
Brain ; 142(4): 867-884, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30879067

RESUMO

Recessive mutations in RTTN, encoding the protein rotatin, were originally identified as cause of polymicrogyria, a cortical malformation. With time, a wide variety of other brain malformations has been ascribed to RTTN mutations, including primary microcephaly. Rotatin is a centrosomal protein possibly involved in centriolar elongation and ciliogenesis. However, the function of rotatin in brain development is largely unknown and the molecular disease mechanism underlying cortical malformations has not yet been elucidated. We performed both clinical and cell biological studies, aimed at clarifying rotatin function and pathogenesis. Review of the 23 published and five unpublished clinical cases and genomic mutations, including the effect of novel deep intronic pathogenic mutations on RTTN transcripts, allowed us to extrapolate the core phenotype, consisting of intellectual disability, short stature, microcephaly, lissencephaly, periventricular heterotopia, polymicrogyria and other malformations. We show that the severity of the phenotype is related to residual function of the protein, not only the level of mRNA expression. Skin fibroblasts from eight affected individuals were studied by high resolution immunomicroscopy and flow cytometry, in parallel with in vitro expression of RTTN in HEK293T cells. We demonstrate that rotatin regulates different phases of the cell cycle and is mislocalized in affected individuals. Mutant cells showed consistent and severe mitotic failure with centrosome amplification and multipolar spindle formation, leading to aneuploidy and apoptosis, which could relate to depletion of neuronal progenitors often observed in microcephaly. We confirmed the role of rotatin in functional and structural maintenance of primary cilia and determined that the protein localized not only to the basal body, but also to the axoneme, proving the functional interconnectivity between ciliogenesis and cell cycle progression. Proteomics analysis of both native and exogenous rotatin uncovered that rotatin interacts with the neuronal (non-muscle) myosin heavy chain subunits, motors of nucleokinesis during neuronal migration, and in human induced pluripotent stem cell-derived bipolar mature neurons rotatin localizes at the centrosome in the leading edge. This illustrates the role of rotatin in neuronal migration. These different functions of rotatin explain why RTTN mutations can lead to heterogeneous cerebral malformations, both related to proliferation and migration defects.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Adulto , Encéfalo/patologia , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Cílios/metabolismo , Feminino , Estudos de Associação Genética/métodos , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Recém-Nascido , Masculino , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/metabolismo , Microcefalia/genética , Mutação , Malformações do Sistema Nervoso/genética , Polimicrogiria/etiologia , Polimicrogiria/patologia
12.
Am J Hum Genet ; 99(1): 217-27, 2016 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-27374774

RESUMO

Complex I deficiency is the most common biochemical phenotype observed in individuals with mitochondrial disease. With 44 structural subunits and over 10 assembly factors, it is unsurprising that complex I deficiency is associated with clinical and genetic heterogeneity. Massively parallel sequencing (MPS) technologies including custom, targeted gene panels or unbiased whole-exome sequencing (WES) are hugely powerful in identifying the underlying genetic defect in a clinical diagnostic setting, yet many individuals remain without a genetic diagnosis. These individuals might harbor mutations in poorly understood or uncharacterized genes, and their diagnosis relies upon characterization of these orphan genes. Complexome profiling recently identified TMEM126B as a component of the mitochondrial complex I assembly complex alongside proteins ACAD9, ECSIT, NDUFAF1, and TIMMDC1. Here, we describe the clinical, biochemical, and molecular findings in six cases of mitochondrial disease from four unrelated families affected by biallelic (c.635G>T [p.Gly212Val] and/or c.401delA [p.Asn134Ilefs(∗)2]) TMEM126B variants. We provide functional evidence to support the pathogenicity of these TMEM126B variants, including evidence of founder effects for both variants, and establish defects within this gene as a cause of complex I deficiency in association with either pure myopathy in adulthood or, in one individual, a severe multisystem presentation (chronic renal failure and cardiomyopathy) in infancy. Functional experimentation including viral rescue and complexome profiling of subject cell lines has confirmed TMEM126B as the tenth complex I assembly factor associated with human disease and validates the importance of both genome-wide sequencing and proteomic approaches in characterizing disease-associated genes whose physiological roles have been previously undetermined.


Assuntos
Alelos , Complexo I de Transporte de Elétrons/deficiência , Proteínas de Membrana/genética , Doenças Mitocondriais/genética , Mutação/genética , Fenótipo , Adolescente , Adulto , Idade de Início , Sequência de Aminoácidos , Criança , Complexo I de Transporte de Elétrons/genética , Feminino , Humanos , Lactente , Masculino , Proteínas de Membrana/química , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
13.
J Assist Reprod Genet ; 36(3): 491-497, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30483911

RESUMO

PURPOSE: We aim to investigate whether there is a genetic predisposition in women who developed ovarian hyperstimulation syndrome (OHSS) after GnRH antagonist protocol with GnRH agonist trigger and freeze-all approach. METHODS: Four patients with OHSS after GnRH agonist trigger and freeze-all approach were gathered from the worldwide patient population. These patients were analyzed through Whole Exome Sequencing. In this study known causes of OHSS were investigated and new causes present in at least two individuals were searched for. RESULTS: In the first part of the study, we evaluated the presence of mutations in genes already known to be involved in OHSS. In PGR and TP53, heterozygous alterations were detected. PGR is predicted to be involved in progesterone resistance with a recessive inheritance pattern and is, therefore, not considered as being causal. The consequences of the variant detected in TP53 currently remain unknown. In part 2 of the study, we assessed the clinical significance of variants in genes previously not linked to OHSS. We especially focused on genes with variants present in ≥ 2 patients. Two patients have variants in the FLT4 gene. Mutations in this gene are linked to hereditary lymphedema, but no link to OHSS has been described. CONCLUSIONS: Defining a genetic predisposition for OHSS is essential in view of prevention. In this study, a potential link between the FLT4 gene and OHSS has been suggested. Future functional studies are essential to define a more precise involvement of the detected variants in the development of OHSS.


Assuntos
Fertilização in vitro , Hormônio Liberador de Gonadotropina/genética , Síndrome de Hiperestimulação Ovariana/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Adulto , Gonadotropina Coriônica/genética , Gonadotropina Coriônica/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Heterozigoto , Antagonistas de Hormônios/administração & dosagem , Humanos , Mutação , Síndrome de Hiperestimulação Ovariana/tratamento farmacológico , Síndrome de Hiperestimulação Ovariana/fisiopatologia , Indução da Ovulação/métodos , Gravidez , Taxa de Gravidez , Proteína Supressora de Tumor p53/genética , Sequenciamento do Exoma
14.
J Med Genet ; 54(6): 432-440, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28258187

RESUMO

BACKGROUND: Collagens are one of the major constituents of the pial membrane, which plays a crucial role in neuronal migration and cortical lamination during brain development. Type III procollagen, the chains of which are encoded by COL3A1, is the ligand of the G protein-coupled receptor 56 (GPR56), also known as adhesion G protein-coupled receptor G1. Bi-allelic mutations in GPR56 give rise to cobblestone-like malformation, white matter changes and cerebellar dysplasia. This report shows that bi-allelic mutations in COL3A1 are associated with a similar phenotype. METHODS: Exome analysis was performed in a family consisting of two affected and two non-affected siblings. Brain imaging studies of this family and of two previously reported individuals with bi-allelic mutations in COL3A1 were reviewed. Functional assays were performed on dermal fibroblasts. RESULTS: Exome analysis revealed a novel homozygous variant c.145C>G (p.Pro49Ala) in exon 2 of COL3A1. Brain MRI in the affected siblings as well as in the two previously reported individuals with bi-allelic COL3A1 mutations showed a brain phenotype similar to that associated with mutations in GPR56. CONCLUSION: Homozygous or compound heterozygous mutations in COL3A1 are associated with cobblestone-like malformation in all three families reported to date. The variability of the phenotype across patients suggests that genetic alterations in distinct domains of type III procollagen can lead to different outcomes. The presence of cobblestone-like malformation in patients with bi-allelic COL3A1 mutations emphasises the critical role of the type III collagen-GPR56 axis and the pial membrane in the regulation of brain development and cortical lamination.


Assuntos
Colágeno Tipo III/genética , Cistos/genética , Malformações do Desenvolvimento Cortical/genética , Receptores Acoplados a Proteínas G/genética , Substância Branca/patologia , Adulto , Alelos , Células Cultivadas , Doenças Cerebelares/genética , Doenças Cerebelares/patologia , Criança , Pré-Escolar , Cistos/patologia , Exoma/genética , Éxons/genética , Feminino , Fibroblastos/patologia , Humanos , Ligantes , Imageamento por Ressonância Magnética/métodos , Masculino , Malformações do Desenvolvimento Cortical/patologia , Mutação/genética , Fenótipo , Adulto Jovem
15.
Bioinformatics ; 32(3): 447-9, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26454277

RESUMO

SUMMARY: Today's genome browsers and protein databanks supply vast amounts of information about proteins. The challenge is to concisely bring together this information in an interactive and easy to generate format. AVAILABILITY AND IMPLEMENTATION: We have developed an interactive CIRCOS module called i-PV to visualize user supplied protein sequence, conservation and SNV data in a live presentable format. I-PV can be downloaded from http://www.i-pv.org. CONTACT: ibrahim.tanyalcin@i-pv.org, itanyalc@vub.ac.be or support@i-pv.org SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Aminoácidos/química , Gráficos por Computador , Proteínas/metabolismo , Análise de Sequência de Proteína/métodos , Software , Animais , Bases de Dados de Proteínas , Humanos , Internet , Camundongos , Polimorfismo Genético/genética , Proteínas/química , Proteínas/genética , Interface Usuário-Computador
16.
BMC Bioinformatics ; 17(1): 425, 2016 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-27760515

RESUMO

BACKGROUND: Predict whether a mutation is deleterious based on the custom 3D model of a protein. RESULTS: We have developed MODICT, a mutation prediction tool which is based on per residue RMSD (root mean square deviation) values of superimposed 3D protein models. Our mathematical algorithm was tested for 42 described mutations in multiple genes including renin (REN), beta-tubulin (TUBB2B), biotinidase (BTD), sphingomyelin phosphodiesterase-1 (SMPD1), phenylalanine hydroxylase (PAH) and medium chain Acyl-Coa dehydrogenase (ACADM). Moreover, MODICT scores corresponded to experimentally verified residual enzyme activities in mutated biotinidase, phenylalanine hydroxylase and medium chain Acyl-CoA dehydrogenase. Several commercially available prediction algorithms were tested and results were compared. The MODICT PERL package and the manual can be downloaded from https://github.com/IbrahimTanyalcin/MODICT . CONCLUSIONS: We show here that MODICT is capable tool for mutation effect prediction at the protein level, using superimposed 3D protein models instead of sequence based algorithms used by POLYPHEN and SIFT.


Assuntos
Biologia Computacional/métodos , Modelos Moleculares , Mutação/genética , Proteínas/química , Proteínas/genética , Software , Acil-CoA Desidrogenase/genética , Humanos , Conformação Proteica , Renina/genética , Tubulina (Proteína)/genética
17.
J Assist Reprod Genet ; 32(7): 1063-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25982422

RESUMO

PURPOSE: We report on the results of the whole-genome analysis performed in a patient who developed severe ovarian hyperstimulation syndrome (OHSS) following gonadotropin-releasing hormone (GnRH) agonist triggering in a "freeze-all" protocol. METHODS: A 30-year-old patient with polycystic ovary syndrome who developed severe early-onset OHSS with clinical ascites, and slight renal and hepatic dysfunction was admitted for monitoring and treatment with cabergoline and intravenous albumin. Exome sequencing to assess for any known genetic predisposition for OHSS was performed. RESULTS: No known genetic variants associated with OHSS predisposition were found. CONCLUSIONS: Case reports of severe OHSS following a "freeze-all" strategy are starting to arise, showing that OHSS has not been completely eliminated with this approach. Further studies should be conducted to confirm if such cases may be due to genetic predisposition or not.


Assuntos
Predisposição Genética para Doença , Síndrome de Hiperestimulação Ovariana/genética , Adulto , Feminino , Genoma Humano , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Masculino , Recuperação de Oócitos , Síndrome do Ovário Policístico/genética , Gravidez , Injeções de Esperma Intracitoplásmicas
18.
Nat Commun ; 15(1): 1232, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38336715

RESUMO

Children conceived through assisted reproductive technologies (ART) have an elevated risk of lower birthweight, yet the underlying cause remains unclear. Our study explores mitochondrial DNA (mtDNA) variants as contributors to birthweight differences by impacting mitochondrial function during prenatal development. We deep-sequenced the mtDNA of 451 ART and spontaneously conceived (SC) individuals, 157 mother-child pairs and 113 individual oocytes from either natural menstrual cycles or after ovarian stimulation (OS) and find that ART individuals carried a different mtDNA genotype than SC individuals, with more de novo non-synonymous variants. These variants, along with rRNA variants, correlate with lower birthweight percentiles, independent of conception mode. Their higher occurrence in ART individuals stems from de novo mutagenesis associated with maternal aging and OS-induced oocyte cohort size. Future research will establish the long-term health consequences of these changes and how these findings will impact the clinical practice and patient counselling in the future.


Assuntos
Recém-Nascido Prematuro , Nascimento Prematuro , Gravidez , Recém-Nascido , Feminino , Humanos , Resultado da Gravidez , Gravidez Múltipla , Nascimento Prematuro/epidemiologia , Peso ao Nascer , Mitocôndrias/genética , DNA Mitocondrial/genética
19.
Biochim Biophys Acta ; 1822(12): 1864-72, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22634129

RESUMO

The X and Y chromosomes, the sex chromosomes, are important key players in germ cell development. Both chromosomes contain genes that are uniquely expressed in male spermatogenesis. Furthermore, these chromosomes are special because men only have a single copy of them. These features make the sex chromosomes interesting for studying in view of spermatogenesis defects. The role of the Y chromosome, together with the presence of Yq microdeletions, in male infertility is well established. Less well-understood are the X-linked genes, their expression patterns and potential impact on male infertility. This review provides an overview of the current knowledge on potential spermatogenesis genes that are located on the mouse and human X chromosomes. A summary is given on knock-out mice models in which X-linked genes have been shown to alter spermatogenesis, and on genes that have been studied in humans. Finally, new research areas like miRNA analysis, Genome Wide Association Studies (GWAS) and array comparative genomic hybridisation (CGH) studies are discussed. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.


Assuntos
Mutação , Espermatogênese/genética , Cromossomo X , Animais , Hibridização Genômica Comparativa , Genes Ligados ao Cromossomo X , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Camundongos Knockout , Cromossomo Y
20.
Eur J Paediatr Neurol ; 37: 155-164, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34535379

RESUMO

Clinical manifestations of malformations of cortical development (MCD) are variable and can range from mild to severe intellectual disability, cerebral palsy and drug-resistant epilepsy. Besides common clinical features, non-specific or more subtle clinical symptoms may be present in association with different types of MCD. Especially in severely affected individuals, subtle but specific underlying clinical symptoms can be overlooked or overshadowed by the global clinical presentation. To facilitate the interpretation of genetic variants detailed clinical information is indispensable. Detailed (neurological) examination can be helpful in assisting with the diagnostic trajectory, both when referring for genetic work-up as well as when interpreting data from molecular genetic testing. This systematic literature review focusses on different clues derived from the neurological examination and potential further work-up triggered by these signs and symptoms in genetically defined MCDs. A concise overview of specific neurological findings and their associations with MCD subtype and genotype are presented, easily applicable in daily clinical practice. The following pathologies will be discussed: neuropathy, myopathy, muscular dystrophies and spastic paraplegia. In the discussion section, tips and pitfalls are illustrated to improve clinical outcome in the future.


Assuntos
Epilepsia Resistente a Medicamentos , Epilepsia , Malformações do Desenvolvimento Cortical , Malformações do Sistema Nervoso , Córtex Cerebral , Epilepsia Resistente a Medicamentos/complicações , Epilepsia/genética , Humanos , Malformações do Desenvolvimento Cortical/genética , Malformações do Sistema Nervoso/complicações , Malformações do Sistema Nervoso/diagnóstico , Malformações do Sistema Nervoso/genética , Sistema Nervoso Periférico/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA