Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 109(4): 601-617, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35395208

RESUMO

Neurodevelopmental disorders are highly heterogenous conditions resulting from abnormalities of brain architecture and/or function. FBXW7 (F-box and WD-repeat-domain-containing 7), a recognized developmental regulator and tumor suppressor, has been shown to regulate cell-cycle progression and cell growth and survival by targeting substrates including CYCLIN E1/2 and NOTCH for degradation via the ubiquitin proteasome system. We used a genotype-first approach and global data-sharing platforms to identify 35 individuals harboring de novo and inherited FBXW7 germline monoallelic chromosomal deletions and nonsense, frameshift, splice-site, and missense variants associated with a neurodevelopmental syndrome. The FBXW7 neurodevelopmental syndrome is distinguished by global developmental delay, borderline to severe intellectual disability, hypotonia, and gastrointestinal issues. Brain imaging detailed variable underlying structural abnormalities affecting the cerebellum, corpus collosum, and white matter. A crystal-structure model of FBXW7 predicted that missense variants were clustered at the substrate-binding surface of the WD40 domain and that these might reduce FBXW7 substrate binding affinity. Expression of recombinant FBXW7 missense variants in cultured cells demonstrated impaired CYCLIN E1 and CYCLIN E2 turnover. Pan-neuronal knockdown of the Drosophila ortholog, archipelago, impaired learning and neuronal function. Collectively, the data presented herein provide compelling evidence of an F-Box protein-related, phenotypically variable neurodevelopmental disorder associated with monoallelic variants in FBXW7.


Assuntos
Proteína 7 com Repetições F-Box-WD , Transtornos do Neurodesenvolvimento , Ubiquitinação , Proteína 7 com Repetições F-Box-WD/química , Proteína 7 com Repetições F-Box-WD/genética , Proteína 7 com Repetições F-Box-WD/metabolismo , Células Germinativas , Mutação em Linhagem Germinativa , Humanos , Transtornos do Neurodesenvolvimento/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
2.
Am J Hum Genet ; 108(5): 951-961, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33894126

RESUMO

The collapsin response mediator protein (CRMP) family proteins are intracellular mediators of neurotrophic factors regulating neurite structure/spine formation and are essential for dendrite patterning and directional axonal pathfinding during brain developmental processes. Among this family, CRMP5/DPYSL5 plays a significant role in neuronal migration, axonal guidance, dendrite outgrowth, and synapse formation by interacting with microtubules. Here, we report the identification of missense mutations in DPYSL5 in nine individuals with brain malformations, including corpus callosum agenesis and/or posterior fossa abnormalities, associated with variable degrees of intellectual disability. A recurrent de novo p.Glu41Lys variant was found in eight unrelated patients, and a p.Gly47Arg variant was identified in one individual from the first family reported with Ritscher-Schinzel syndrome. Functional analyses of the two missense mutations revealed impaired dendritic outgrowth processes in young developing hippocampal primary neuronal cultures. We further demonstrated that these mutations, both located in the same loop on the surface of DPYSL5 monomers and oligomers, reduced the interaction of DPYSL5 with neuronal cytoskeleton-associated proteins MAP2 and ßIII-tubulin. Our findings collectively indicate that the p.Glu41Lys and p.Gly47Arg variants impair DPYSL5 function on dendritic outgrowth regulation by preventing the formation of the ternary complex with MAP2 and ßIII-tubulin, ultimately leading to abnormal brain development. This study adds DPYSL5 to the list of genes implicated in brain malformation and in neurodevelopmental disorders.


Assuntos
Agenesia do Corpo Caloso/genética , Cerebelo/anormalidades , Mutação de Sentido Incorreto/genética , Transtornos do Neurodesenvolvimento/genética , Adulto , Agenesia do Corpo Caloso/diagnóstico por imagem , Cerebelo/diagnóstico por imagem , Criança , Pré-Escolar , Feminino , Humanos , Hidrolases/química , Hidrolases/genética , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/genética , Masculino , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Moleculares , Transtornos do Neurodesenvolvimento/diagnóstico por imagem , Tubulina (Proteína)/metabolismo , Adulto Jovem
3.
Cerebellum ; 22(2): 206-222, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35218524

RESUMO

Cerebellar hypoplasia and dysplasia encompass a group of clinically and genetically heterogeneous disorders frequently associated with neurodevelopmental impairment. The Neuron Navigator 2 (NAV2) gene (MIM: 607,026) encodes a member of the Neuron Navigator protein family, widely expressed within the central nervous system (CNS), and particularly abundant in the developing cerebellum. Evidence across different species supports a pivotal function of NAV2 in cytoskeletal dynamics and neurite outgrowth. Specifically, deficiency of Nav2 in mice leads to cerebellar hypoplasia with abnormal foliation due to impaired axonal outgrowth. However, little is known about the involvement of the NAV2 gene in human disease phenotypes. In this study, we identified a female affected with neurodevelopmental impairment and a complex brain and cardiac malformations in which clinical exome sequencing led to the identification of NAV2 biallelic truncating variants. Through protein expression analysis and cell migration assay in patient-derived fibroblasts, we provide evidence linking NAV2 deficiency to cellular migration deficits. In model organisms, the overall CNS histopathology of the Nav2 hypomorphic mouse revealed developmental anomalies including cerebellar hypoplasia and dysplasia, corpus callosum hypo-dysgenesis, and agenesis of the olfactory bulbs. Lastly, we show that the NAV2 ortholog in Drosophila, sickie (sick) is widely expressed in the fly brain, and sick mutants are mostly lethal with surviving escapers showing neurobehavioral phenotypes. In summary, our results unveil a novel human neurodevelopmental disorder due to genetic loss of NAV2, highlighting a critical conserved role of the NAV2 gene in brain and cerebellar development across species.


Assuntos
Encéfalo , Malformações do Sistema Nervoso , Animais , Feminino , Humanos , Camundongos , Cerebelo/anormalidades , Neurônios
4.
EMBO Rep ; 22(9): e51781, 2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34323349

RESUMO

During neocortical development, neural stem cells (NSCs) divide symmetrically to self-renew at the early stage and then divide asymmetrically to generate post-mitotic neurons. The molecular mechanisms regulating the balance between NSC self-renewal and neurogenesis are not fully understood. Using mouse in utero electroporation (IUE) technique and in vitro human NSC differentiation models including cerebral organoids (hCOs), we show here that regulator of cell cycle (RGCC) modulates NSC self-renewal and neuronal differentiation by affecting cell cycle regulation and spindle orientation. RGCC deficiency hampers normal cell cycle process and dysregulates the mitotic spindle, thus driving more cells to divide asymmetrically. These modulations diminish the NSC population and cause NSC pre-differentiation that eventually leads to brain developmental malformation in hCOs. We further show that RGCC might regulate NSC spindle orientation by affecting the organization of centrosome and microtubules. Our results demonstrate that RGCC is essential to maintain the NSC pool during cortical development and suggest that RGCC defects could have etiological roles in human brain malformations.


Assuntos
Neocórtex , Células-Tronco Neurais , Animais , Diferenciação Celular , Camundongos , Neurogênese , Neurônios
5.
Childs Nerv Syst ; 39(5): 1341-1345, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36574013

RESUMO

BACKGROUND: Dandy-Walker malformation (DWM) is a posterior fossa malformation characterized by a huge posterior fossa cyst in communication with the fourth ventricle. Hydrocephalus is associated with more than 80% of cases and is usually treated by shunting. Despite infection being a common complication of the shunt, abscess formation within the cyst was reported only once. CASE REPORT: A neonate affected by DWM developed a posterior fossa abscess following a shunt infection. The purulent collection was refractory to standard treatment (antibiotics and burr hole drainage); therefore, an endoscopic approach was performed in order to remove the purulent collection under direct vision. This material was aspirated with the help of an endoscopic ultrasonic aspirator. The outcome was favorable, with a resolution of infection and re-implantation of the ventriculo-peritoneal shunt. Surprisingly, post-operative radiological examination showed substantial modification of the anatomy of the posterior fossa with disappearing of the Dandy-Walker cyst. To the best of our knowledge, this is the first documented report of a true Dandy-Walker malformation that modified its anatomical appearance over time. DISCUSSION AND CONCLUSION: Endoscopic aspiration of intracranial purulent collection should be considered a valid option to manage complicated cases. An endoscopic ultrasonic aspirator may make the procedure more effective and faster.


Assuntos
Cistos , Síndrome de Dandy-Walker , Recém-Nascido , Humanos , Síndrome de Dandy-Walker/complicações , Síndrome de Dandy-Walker/diagnóstico por imagem , Síndrome de Dandy-Walker/cirurgia , Abscesso/cirurgia , Ultrassom , Ventriculostomia/métodos , Cistos/cirurgia , Imageamento por Ressonância Magnética
6.
Vet Med (Praha) ; 68(8): 343-348, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37982125

RESUMO

An intracranial arachnoid cyst (IAC) is a rare developmental disorder that is consistent with cerebrospinal fluid accumulation between the brain and the arachnoid membrane. A quadrigeminal cyst is a specific type of cyst that is identified based on its localization. To the best of our knowledge, this is the first study to report on this type of pathology in a Brussels Griffon. This case study describes an 11-month-old female Brussels Griffon with symptoms of reluctance to lower the head and eat from a bowl placed on the ground, combined with episodes of vocalization. The patient was subsequently diagnosed with a quadrigeminal cyst during a low-field magnetic resonance imaging (MRI) exam, and she tested positive for toxoplasmosis in the blood test. Arachnoid cysts are often described as incidental findings, but the characteristics of neurological symptoms in the presented patient suggest that the cyst was clinically significant. The currently known options of pharmacological and surgical treatment give some hope for symptomatic patients, although their definitive success rate is not yet fully known.

7.
Neurol Neurochir Pol ; 57(3): 269-281, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37078131

RESUMO

INTRODUCTION: Corpus callosum abnormalities are complex, aetiologically diverse, and clinically heterogeneous conditions. Counselling parents regarding their causes and associated syndromes, and predicting the neurodevelopmental and seizure risk prognosis, is challenging. MATERIAL AND METHODS: We describe the clinical characteristics, associated anomalies, and neurodevelopmental outcomes of children with agenesis of corpus callosum (ACC). Fifty-one neonates with ACC/hypoplasia of the corpus callosum were identified over a 17-year period, and their medical records were retrospectively reviewed. RESULTS: Patients were classified into two groups depending on the presence or absence of associated abnormalities. The first group (17 patients, 33.4%) presented with isolated callosal anomalies. The second group included 34 patients (66.6%) with associated cerebral and extracerebral anomalies. We achieved an identifiable genetic aetiology in 23.5% of our cohort. Magnetic resonance imaging was performed in 28 patients (55%), and of these 39.3% had additional brain anomalies. During the study period, five patients died early in the neonatal period and four were lost to follow up. Of the 42 followed patients, 13 (31%) showed normal neurodevelopment, 13 (31%) showed mild delay, and 16 (38%) had a severe delay. Fifteen (35.7%) had epilepsy. CONCLUSIONS AND CLINICAL IMPLICATIONS: We have confirmed that callosal defects are frequently accompanied by brain and somatic anomalies. Additional abnormalities were shown to be significantly associated with developmental delay and increased risk of epilepsy. We have highlighted essential clinical features that may provide diagnostic clues to physicians and we have given examples of underlying genetic disorders. We have provided recommendations about extended neuroimaging diagnostics and widespread genetic testing that may impact upon daily clinical practice. Paediatric neurologists may therefore use our findings to help base their decisions regarding this matter.


Assuntos
Encefalopatias , Corpo Caloso , Recém-Nascido , Humanos , Criança , Corpo Caloso/diagnóstico por imagem , Corpo Caloso/patologia , Estudos Retrospectivos , Agenesia do Corpo Caloso/diagnóstico por imagem , Agenesia do Corpo Caloso/genética , Agenesia do Corpo Caloso/patologia , Encéfalo/patologia , Encefalopatias/patologia , Imageamento por Ressonância Magnética
8.
Neuroimage ; 260: 119486, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35843515

RESUMO

T1-weighted magnetic resonance images provide a comprehensive view of the morphology of the human brain at the macro scale. These images are usually the input of a segmentation process that aims detecting the anatomical structures labeling them according to a predefined set of target tissues. Automated methods for brain tissue segmentation rely on anatomical priors of the human brain structures. This is the reason why their performance is quite accurate on healthy individuals. Nevertheless model-based tools become less accurate in clinical practice, specifically in the cases of severe lesions or highly distorted cerebral anatomy. More recently there are empirical evidences that a data-driven approach can be more robust in presence of alterations of brain structures, even though the learning model is trained on healthy brains. Our contribution is a benchmark to support an open investigation on how the tissue segmentation of distorted brains can be improved by adopting a supervised learning approach. We formulate a precise definition of the task and propose an evaluation metric for a fair and quantitative comparison. The training sample is composed of almost one thousand healthy individuals. Data include both T1-weighted MR images and their labeling of brain tissues. The test sample is a collection of several tens of individuals with severe brain distortions. Data and code are openly published on BrainLife, an open science platform for reproducible neuroscience data analysis.


Assuntos
Benchmarking , Processamento de Imagem Assistida por Computador , Encéfalo/anatomia & histologia , Criança , Humanos , Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos
9.
Am J Hum Genet ; 105(4): 734-746, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31585106

RESUMO

Disorders of somatic mosaicism (DoSM) are a diverse group of syndromic and non-syndromic conditions caused by mosaic variants in genes that regulate cell survival and proliferation. Despite overlap in gene space and technical requirements, few clinical labs specialize in DoSM compared to oncology. We adapted a high-sensitivity next-generation sequencing cancer assay for DoSM in 2014. Some 343 individuals have been tested over the past 5 years, 58% of which had pathogenic and likely pathogenic (P/LP) findings, for a total of 206 P/LP variants in 22 genes. Parameters associated with the high diagnostic yield were: (1) deep sequencing (∼2,000× coverage), (2) a broad gene set, and (3) testing affected tissues. Fresh and formalin-fixed paraffin embedded tissues performed equivalently for identification of P/LP variants (62% and 71% of individuals, respectively). Comparing cultured fibroblasts to skin biopsies suggested that culturing might boost the allelic fraction of variants that confer a growth advantage, specifically gain-of-function variants in PIK3CA. Buccal swabs showed high diagnostic sensitivity in case subjects where disease phenotypes manifested in the head or brain. Peripheral blood was useful as an unaffected comparator tissue to determine somatic versus constitutional origin but had poor diagnostic sensitivity. Descriptions of all tested individuals, specimens, and P/LP variants included in this cohort are available to further the study of the DoSM population.


Assuntos
Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mosaicismo , Biópsia , Estudos de Coortes , Humanos
10.
Am J Hum Genet ; 105(3): 631-639, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31353024

RESUMO

Notch signaling is an established developmental pathway for brain morphogenesis. Given that Delta-like 1 (DLL1) is a ligand for the Notch receptor and that a few individuals with developmental delay, intellectual disability, and brain malformations have microdeletions encompassing DLL1, we hypothesized that insufficiency of DLL1 causes a human neurodevelopmental disorder. We performed exome sequencing in individuals with neurodevelopmental disorders. The cohort was identified using known Matchmaker Exchange nodes such as GeneMatcher. This method identified 15 individuals from 12 unrelated families with heterozygous pathogenic DLL1 variants (nonsense, missense, splice site, and one whole gene deletion). The most common features in our cohort were intellectual disability, autism spectrum disorder, seizures, variable brain malformations, muscular hypotonia, and scoliosis. We did not identify an obvious genotype-phenotype correlation. Analysis of one splice site variant showed an in-frame insertion of 12 bp. In conclusion, heterozygous DLL1 pathogenic variants cause a variable neurodevelopmental phenotype and multi-systemic features. The clinical and molecular data support haploinsufficiency as a mechanism for the pathogenesis of this DLL1-related disorder and affirm the importance of DLL1 in human brain development.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Haploinsuficiência , Proteínas de Membrana/genética , Transtornos do Neurodesenvolvimento/genética , Estudos de Coortes , Feminino , Humanos , Ligantes , Masculino , Linhagem , Sequenciamento do Exoma
11.
Am J Med Genet A ; 188(8): 2331-2338, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35686685

RESUMO

The recent finding that some patients with fetal akinesia deformation sequence (FADS) carry variants in the TUBB2B gene has prompted us to add to the existing literature a first description of two fetal FADS cases carrying TUBA1A variants. Hitherto, only isolated cortical malformations have been described with TUBA1A mutation, including microlissencephaly, lissencephaly, central pachygyria and polymicrogyria-like cortical dysplasia, generalized polymicrogyria cortical dysplasia, and/or the "simplified" gyral pattern. The neuropathology of our fetal cases shows several common features of tubulinopathies, in particular, the dysmorphism of the basal ganglia, as the most pathognomonic sign. The cortical ribbon anomalies were extremely severe and concordant with the complex cortical malformation. In conclusion, we broaden the phenotypic spectrum of TUBA1A variants, to include FADS.


Assuntos
Artrogripose , Lisencefalia , Malformações do Desenvolvimento Cortical , Polimicrogiria , Artrogripose/diagnóstico , Artrogripose/genética , Humanos , Lisencefalia/genética , Malformações do Desenvolvimento Cortical/genética , Mutação , Tubulina (Proteína)/genética
12.
Am J Med Genet A ; 188(2): 648-657, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34761517

RESUMO

SMG8 (MIM *617315) is a regulatory subunit involved in nonsense-mediated mRNA decay (NMD), a cellular protective pathway that regulates mRNA transcription, transcript stability, and degrades transcripts containing premature stop codons. SMG8 binds SMG9 and SMG1 to form the SMG1C complex and inhibit the kinase activity of SMG1. Biallelic deleterious variants in SMG9 are known to cause a heart and brain malformation syndrome (HBMS; MIM #616920), whereas biallelic deleterious variants in SMG8 were recently described to cause a novel neurodevelopmental disorder (NDD) with dysmorphic facies and cataracts, now defined as Alzahrani-Kuwahara syndrome (ALKUS: MIM #619268). Only eight subjects from four families with ALKUS have been described to date. Through research reanalysis of a nondiagnostic clinical exome, we identified a subject from a fifth unrelated family with a homozygous deleterious variant in SMG8 and features consistent with ALKUS. Interestingly, the subject also had unilateral microphthalmia, a clinical feature that has been described in SMG9-related disorder. Our study expands the phenotypic spectrum of SMG8-related disorder, demonstrates an overlapping phenotype between SMG8- and SMG9-related rare disease traits, provides further evidence for the SMG8 and SMG9 protein interactions, and highlights the importance of revisiting nondiagnostic exome data to identify and affirm emerging novel genes for rare disease traits.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Degradação do RNAm Mediada por Códon sem Sentido , Alelos , Homozigoto , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fenótipo , Fosforilação
13.
J Xray Sci Technol ; 30(6): 1243-1260, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36155489

RESUMO

BACKGROUND: Standard planes (SPs) are crucial for the diagnosis of fetal brain malformation. However, it is very time-consuming and requires extensive experiences to acquire the SPs accurately due to the large difference in fetal posture and the complexity of SPs definitions. OBJECTIVE: This study aims to present a guiding approach that could assist sonographer to obtain the SPs more accurately and more quickly. METHODS: To begin with, sonographer uses the 3D probe to scan the fetal head to obtain 3D volume data, and then we used affine transformation to calibrate 3D volume data to the standard body position and established the corresponding 3D head model in 'real time'. When the sonographer uses the 2D probe to scan a plane, the position of current plane can be clearly show in 3D head model by our RLNet (regression location network), which can conduct the sonographer to obtain the three SPs more accurately. When the three SPs are located, the sagittal plane and the coronal planes can be automatically generated according to the spatial relationship with the three SPs. RESULTS: Experimental results conducted on 3200 2D US images show that the RLNet achieves average angle error of the transthalamic plane was 3.91±2.86°, which has a obvious improvement compared other published data. The automatically generated coronal and sagittal SPs conform the diagnostic criteria and the diagnostic requirements of fetal brain malformation. CONCLUSIONS: A guiding scanning method based deep learning for ultrasonic brain malformation screening is firstly proposed and it has a pragmatic value for future clinical application.


Assuntos
Cabeça , Ultrassonografia Pré-Natal , Gravidez , Feminino , Humanos , Ultrassonografia Pré-Natal/métodos , Encéfalo/diagnóstico por imagem , Ultrassonografia
14.
Am J Med Genet A ; 185(9): 2801-2808, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33938618

RESUMO

In this review, we explore evidence that hypoxia in the developing human fetus can lead not only to the more commonly accepted disruptive-type defects, but also patterns of anomalies that suggest that hypoxia can exert a more classic teratogenic effect, using the brain as one example. We review neuropathology in the context of intrauterine hypoxia, particularly as it relates to carbon monoxide poisoning, in utero strokes, and homozygous alpha-thalassemia. In general, the associated brain injuries resemble those seen with other causes of hypoxic-ischemic injury. Fetal strokes during development usually lead to loss of brain tissue in areas that do not follow a typical embryologic pattern, and therefore are considered disruptions. However, there is also evidence that fetal brain ischemia can cause more classically recognized patterns of abnormal embryonic neuronal migration and organization such as polymicrogyria, cortical dysplasia, or dysgenesis, including select types of focal cortical dysplasia. This study summarizes available literature and evidence to raise clinicians' awareness regarding the association between hypoxia and congenital anomalies, including brain malformations.


Assuntos
Anormalidades Múltiplas/patologia , Anormalidades Congênitas/patologia , Hipóxia/fisiopatologia , Teratogênese , Teratogênicos/química , Anormalidades Múltiplas/etiologia , Anormalidades Congênitas/etiologia , Humanos
15.
Ultrasound Obstet Gynecol ; 56(5): 732-739, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31710738

RESUMO

OBJECTIVE: To analyze a large retrospective cohort of fetuses in which the cavum septi pellucidi and vergae (CSPV) was not present or was not in its expected position on in-utero magnetic resonance imaging (iuMRI), in order to describe the possible causes of that finding and provide a diagnostic approach to assess such cases in clinical practice using iuMRI. METHODS: This was a retrospective study of fetuses that underwent iuMRI at a single institution, over an 18-year period (2000-2017 inclusive), in which the CSPV was not visualized or was abnormal. All iuMRI studies were reviewed and classified as CSPV being not present, disrupted (visualization of remnants of an otherwise normally placed CSPV) or malpositioned (CSPV was present, but not in its expected position). We describe the neuropathology present in each of the groups. RESULTS: Of the 270 fetuses that met the inclusion criteria, the CSPV was described as malpositioned in 150 (56%), disrupted in 71 (26%) and not present in 49 (18%). Malpositioned CSPV was present only in cases with agenesis of the corpus callosum and three specific patterns of malpositioning are described, depending on the location of the leaflets of the CSPV and fornix. Disrupted CSPV was present in fetuses with hydrocephalus or pathologies causing extensive brain parenchymal injury. Not present CSPV was found in cases with holoprosencephaly or when absence of the CSPV appeared to be an isolated finding. CONCLUSION: We have described a large cohort of fetuses with non-visualization of a normal CSPV on iuMRI and present a categorical classification system based on the CSPV being not present, disrupted or malpositioned. This approach should help in the diagnosis of the underlying cause of a CSPV abnormality. © 2019 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Feto/diagnóstico por imagem , Imageamento por Ressonância Magnética/classificação , Malformações do Sistema Nervoso/diagnóstico por imagem , Diagnóstico Pré-Natal/classificação , Septo Pelúcido/diagnóstico por imagem , Feminino , Feto/anormalidades , Humanos , Imageamento por Ressonância Magnética/métodos , Malformações do Sistema Nervoso/classificação , Malformações do Sistema Nervoso/embriologia , Gravidez , Diagnóstico Pré-Natal/métodos , Valores de Referência , Estudos Retrospectivos , Septo Pelúcido/anormalidades , Septo Pelúcido/embriologia
16.
Childs Nerv Syst ; 36(10): 2571-2596, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33048248

RESUMO

Neurocutaneous melanosis (NCM; MIM # 249400; ORPHA: 2481], first reported by the Bohemian pathologist Rokitansky in 1861, and now more precisely defined as neurocutaneous melanocytosis, is a rare, congenital syndrome characterised by the association of (1) congenital melanocytic nevi (CMN) of the skin with overlying hypertrichosis, presenting as (a) large (LCMN) or giant and/or multiple (MCMN) melanocytic lesions (or both; sometimes associated with smaller "satellite" nevi) or (b) as proliferative melanocytic nodules; and (2) melanocytosis (with infiltration) of the brain parenchyma and/or leptomeninges. CMN of the skin and leptomeningeal/nervous system infiltration are usually benign, more rarely may progress to melanoma or non-malignant melanosis of the brain. Approximately 12% of individuals with LCMN will develop NCM: wide extension and/or dorsal axial distribution of LCMN increases the risk of NCM. The CMN are recognised at birth and are distributed over the skin according to 6 or more patterns (6B patterns) in line with the archetypical patterns of distribution of mosaic skin disorders. Neurological manifestations can appear acutely in infancy, or more frequently later in childhood or adult life, and include signs/symptoms of intracranial hypertension, seizures/epilepsy, cranial nerve palsies, motor/sensory deficits, cognitive/behavioural abnormalities, sleep cycle anomalies, and eventually neurological deterioration. NMC patients may be symptomatic or asymptomatic, with or without evidence of the typical nervous system changes at MRI. Associated brain and spinal cord malformations include the Dandy-Walker malformation (DWM) complex, hemimegalencephaly, cortical dysplasia, arachnoid cysts, Chiari I and II malformations, syringomyelia, meningoceles, occult spinal dysraphism, and CNS lipoma/lipomatosis. There is no systemic involvement, or only rarely. Pathogenically, single postzygotic mutations in the NRAS (neuroblastoma RAS viral oncogene homologue; MIM # 164790; at 1p13.2) proto-oncogene explain the occurrence of single/multiple CMNs and melanocytic and non-melanocytic nervous system lesions in NCM: these disrupt the RAS/ERK/mTOR/PI3K/akt pathways. Diagnostic/surveillance work-ups require physical examination, ophthalmoscopy, brain/spinal cord magnetic resonance imaging (MRI) and angiography (MRA), positron emission tomography (PET), and video-EEG and IQ testing. Treatment strategies include laser therapy, chemical peeling, dermabrasion, and surgical removal/grafting for CMNs and shunt surgery and surgical removal/chemo/radiotherapy for CNS lesions. Biologically targeted therapies tailored (a) BRAF/MEK in NCM mice (MEK162) and GCMN (trametinib); (b) PI3K/mTOR (omipalisib/GSK2126458) in NMC cells; (c) RAS/MEK (vemurafenib and trametinib) in LCMNs cells; or created experimental NMC cells (YP-MEL).


Assuntos
Melanose , Síndromes Neurocutâneas , Nevo Pigmentado , Adulto , Animais , Humanos , Imageamento por Ressonância Magnética , Melanose/complicações , Camundongos , Síndromes Neurocutâneas/complicações , Síndromes Neurocutâneas/diagnóstico por imagem , Nevo Pigmentado/complicações , Fosfatidilinositol 3-Quinases , Proto-Oncogene Mas , Tomografia Computadorizada por Raios X
17.
Proc Natl Acad Sci U S A ; 114(8): 2036-2041, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28154132

RESUMO

Inositol hexakisphosphate kinase 1 (IP6K1), which generates 5-diphosphoinositol pentakisphosphate (5-IP7), physiologically mediates numerous functions. We report that IP6K1 deletion leads to brain malformation and abnormalities of neuronal migration. IP6K1 physiologically associates with α-actinin and localizes to focal adhesions. IP6K1 deletion disrupts α-actinin's intracellular localization and function. The IP6K1 deleted cells display substantial decreases of stress fiber formation and impaired cell migration and spreading. Regulation of α-actinin by IP6K1 requires its kinase activity. Deletion of IP6K1 abolishes α-actinin tyrosine phosphorylation, which is known to be regulated by focal adhesion kinase (FAK). FAK phosphorylation is substantially decreased in IP6K1 deleted cells. 5-IP7, a product of IP6K1, promotes FAK autophosphorylation. Pharmacologic inhibition of IP6K by TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl)purine] recapitulates the phenotype of IP6K1 deletion. These findings establish that IP6K1 physiologically regulates neuronal migration by binding to α-actinin and influencing phosphorylation of both FAK and α-actinin through its product 5-IP7.


Assuntos
Actinina/metabolismo , Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Neurônios/fisiologia , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Animais , Encéfalo/anormalidades , Encéfalo/enzimologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Fosfatos de Inositol/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Fosfotransferases (Aceptor do Grupo Fosfato)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo
18.
Pediatr Int ; 62(5): 593-599, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31886593

RESUMO

BACKGROUND: Holoprosencephaly (HPE) is a congenital malformation with an estimated prevalence of 0.10-6.06 per 10 000 births but with no nationwide data specific to Japan. METHODS: This nationwide retrospective questionnaire survey was conducted from 2011 to 2013. All 467 training hospitals for perinatal and neonatal care certified by the Japan Society of Perinatal and Neonatal Medicine were contacted. The birth prevalence rate (BPR) was assessed from the primary survey and clinical characteristics from the secondary survey. RESULTS: We received valid responses from 253 hospitals in the primary survey (54.6%). Of 390 342 live births, 60 were diagnosed with HPE (23 males and 37 females), resulting in an actual BPR of 1.54 per 10 000 live births. The point estimate for HPE cases was 100 (95% confidence interval [CI]: 80.7-120), and the estimated BPR of HPE was calculated to be 0.32 per 10 000 live births (95% CI: 0.26-0.38) based on 3 117 853 live births according to Japanese national statistics during the study period. In the secondary survey, we obtained data for 49 cases (19 males and 30 females). Of these, 20 were alobar (40.8%), 20 were semilobar (40.8%), five were lobar (10.4%), and four were of unknown type. Genetic examination was performed in 37 of the 49 HPE patients and revealed that chromosomes 13, 18, and 7 were affected in eight, six, and four patients, respectively. CONCLUSIONS: This is the most extensive survey on holoprosencephaly to date in Japan. The estimated BPR was consistent with that reported in previous research.


Assuntos
Holoprosencefalia/epidemiologia , Feminino , Testes Genéticos , Holoprosencefalia/genética , Humanos , Recém-Nascido , Japão/epidemiologia , Nascido Vivo/epidemiologia , Masculino , Prevalência , Estudos Retrospectivos , Inquéritos e Questionários
19.
Am J Med Genet A ; 179(11): 2257-2262, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31390136

RESUMO

INTRODUCTION: SMG9 deficiency is an extremely rare autosomal recessive condition originally described in three patients from two families harboring homozygous truncating SMG9 variants in a context of severe syndromic developmental disorder. To our knowledge, no additional patient has been described since this first report. METHODS: We performed exome sequencing in a patient exhibiting a syndromic developmental delay and in her unaffected parents and report the phenotypic features. RESULTS: Our patient presented with a syndromic association of severe global developmental delay and diverse malformations, including cleft lip and palate, facial dysmorphic features, brain abnormalities, heart defect, growth retardation, and severe infections. She carried a novel SMG9 homozygous variant NM_019108.3:c.1177C>T, p.(Gln393*), while her unaffected parents were both heterozygous. CONCLUSIONS: We confirm that bi-allelic truncating SMG9 variants cause a severe developmental syndrome including brain and heart malformations associated with facial dysmorphic features, severe growth and developmental delay with or without ophthalmological abnormalities, severe feeding difficulties, and life-threatening infections.


Assuntos
Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Alelos , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Pré-Escolar , Consanguinidade , Feminino , Estudos de Associação Genética/métodos , Homozigoto , Humanos , Linhagem , Fenótipo , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA