Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neuroscience ; 445: 190-206, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32360592

RESUMO

Disruptions in the gene encoding methyl-CpG binding protein 2 (MECP2) underlie complex neurodevelopmental disorders including Rett Syndrome (RTT), MECP2 duplication disorder, intellectual disabilities, and autism. Significant progress has been made on the molecular and cellular basis of MECP2-related disorders providing a new framework for understanding how altered epigenetic landscape can derail the formation and refinement of neuronal circuits in early postnatal life and proper neurological function. This review will summarize selected major findings from the past years and particularly highlight the integrated and multidisciplinary work done at eight NIH-funded Intellectual and Developmental Disabilities Research Centers (IDDRC) across the US. Finally, we will outline a path forward with identification of reliable biomarkers and outcome measures, longitudinal preclinical and clinical studies, reproducibility of results across centers as a synergistic effort to decode and treat the pathogenesis of the complex MeCP2 disorders.


Assuntos
Proteína 2 de Ligação a Metil-CpG , Síndrome de Rett , Proteínas de Transporte , Criança , Deficiências do Desenvolvimento , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Mutação , Reprodutibilidade dos Testes , Síndrome de Rett/genética
2.
Proc Natl Acad Sci U S A ; 117(38): 23298-23303, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-31332003

RESUMO

Neurodevelopmental spectrum disorders like autism (ASD) are diagnosed, on average, beyond age 4 y, after multiple critical periods of brain development close and behavioral intervention becomes less effective. This raises the urgent need for quantitative, noninvasive, and translational biomarkers for their early detection and tracking. We found that both idiopathic (BTBR) and genetic (CDKL5- and MeCP2-deficient) mouse models of ASD display an early, impaired cholinergic neuromodulation as reflected in altered spontaneous pupil fluctuations. Abnormalities were already present before the onset of symptoms and were rescued by the selective expression of MeCP2 in cholinergic circuits. Hence, we trained a neural network (ConvNetACh) to recognize, with 97% accuracy, patterns of these arousal fluctuations in mice with enhanced cholinergic sensitivity (LYNX1-deficient). ConvNetACh then successfully detected impairments in all ASD mouse models tested except in MeCP2-rescued mice. By retraining only the last layers of ConvNetACh with heart rate variation data (a similar proxy of arousal) directly from Rett syndrome patients, we generated ConvNetPatients, a neural network capable of distinguishing them from typically developing subjects. Even with small cohorts of rare patients, our approach exhibited significant accuracy before (80% in the first and second year of life) and into regression (88% in stage III patients). Thus, transfer learning across species and modalities establishes spontaneous arousal fluctuations combined with deep learning as a robust noninvasive, quantitative, and sensitive translational biomarker for the rapid and early detection of neurodevelopmental disorders before major symptom onset.


Assuntos
Acetilcolina/metabolismo , Nível de Alerta , Transtorno Autístico/psicologia , Aprendizado Profundo , Animais , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Pupila/fisiologia , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatologia , Síndrome de Rett/psicologia
3.
J Neurodev Disord ; 11(1): 15, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31362710

RESUMO

BACKGROUND: Rett syndrome is a neurodevelopmental disorder caused by a mutation in the X-linked MECP2 gene. Individuals with Rett syndrome typically develop normally until around 18 months of age before undergoing a developmental regression, and the disorder can lead to cognitive, motor, sensory, and autonomic dysfunction. Understanding the mechanism of developmental regression represents a unique challenge when viewed through a neuroscience lens. Are circuits that were previously established erased, and are new ones built to supplant old ones? One way to examine circuit-level changes is with the use of electroencephalography (EEG). Previous studies of the EEG in individuals with Rett syndrome have focused on morphological characteristics, but few have explored spectral power, including power as an index of brain function or disease severity. This study sought to determine if EEG power differs in girls with Rett syndrome and typically developing girls and among girls with Rett syndrome based on various clinical characteristics in order to better understand neural connectivity and cortical organization in individuals with this disorder. METHODS: Resting state EEG data were acquired from girls with Rett syndrome (n = 57) and typically developing children without Rett syndrome (n = 37). Clinical data were also collected for girls with Rett syndrome. EEG power across several brain regions in numerous frequency bands was then compared between girls with Rett syndrome and typically developing children and power in girls with Rett syndrome was compared based on these clinical measures. 1/ƒ slope was also compared between groups. RESULTS: Girls with Rett syndrome demonstrate significantly lower power in the middle frequency bands across multiple brain regions. Additionally, girls with Rett syndrome that are postregression demonstrate significantly higher power in the lower frequency delta and theta bands and a significantly more negative slope of the power spectrum. Increased power in these bands, as well as a more negative 1/ƒ slope, trended with lower cognitive assessment scores. CONCLUSIONS: Increased power in lower frequency bands is consistent with previous studies demonstrating a "slowing" of the background EEG in Rett syndrome. This increase, particularly in the delta band, could represent abnormal cortical inhibition due to dysfunctional GABAergic signaling and could potentially be used as a marker of severity due to associations with more severe Rett syndrome phenotypes.


Assuntos
Ondas Encefálicas/fisiologia , Córtex Cerebral/fisiopatologia , Eletroencefalografia/métodos , Síndrome de Rett/fisiopatologia , Biomarcadores , Feminino , Neuroimagem Funcional , Humanos , Lactente , Índice de Gravidade de Doença
4.
Intellect Dev Disabil ; 55(6): 419-431, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29194024

RESUMO

Rett Syndrome (RTT) is characterized by severe impairment in fine motor (FM) and expressive language (EL) function, making accurate evaluations of development difficult with standardized assessm ents. In this study, the administration and scoring of the Mullen Scales of Early Learning (MSEL) were adapted to eliminate the confounding effects of FM and EL impairments in assessing development. Forty-seven girls with RTT were assessed with the Adapted-MSEL (MSEL-A), a subset (n = 30) was also assessed using the Vineland Adaptive Behavior Scales-Second Edition (Vineland-II) and a further subset (n = 17) was assessed using an eye-tracking version of the MSEL (MSEL-ET). Participants performed better on the visual reception (VR) and receptive language (RL) domains compared to the FM and EL domains on the MSEL-A. Individual performance on each domain was independent of other domains. Corresponding MSEL-A and Vineland-II domains were significantly correlated. The MSEL-ET was as accurate as the MSEL-A in assessing VR and RL, yet took a 44% less time. Results suggested that the MSEL-A and the MSEL-ET could be viable measures for accurately assessing developmental domains in children with RTT.


Assuntos
Desenvolvimento Infantil/fisiologia , Deficiências do Desenvolvimento/psicologia , Aprendizagem/fisiologia , Síndrome de Rett/psicologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Psicometria , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
5.
Mol Autism ; 7: 30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27354901

RESUMO

BACKGROUND: Duplication and deletion of the chromosomal region 16p11.2 cause a broad range of impairments, including intellectual disability, language disorders, and sensory symptoms. However, it is unclear how changes in 16p11.2 dosage affect cortical circuitry during development. The aim of this study was to investigate whether the visual evoked potential (VEP) could be used as a noninvasive quantitative measure of cortical processing in children with 16p11.2 copy number variation. METHODS: Pattern-reversal VEPs were successfully recorded in 19 deletion carriers, 9 duplication carriers, and 13 typically developing children between the ages of 3 and 14 years. The stimulus was a black and white checkerboard (60') that reversed contrast at 2 Hz. VEP responses were extracted from continuous EEG recorded using a high-density elasticized electrode net. RESULTS: Quantitative analysis of the VEP waveform revealed that, relative to controls, deletion carriers displayed increased amplitude and duplication carriers displayed diminished amplitude. Latencies of the VEP waveform components were unaffected by 16p11.2 status. P1 amplitude did not correlate with age, IQ, or head circumference. CONCLUSIONS: The results of this study suggest that recording VEP is a useful method to assay cortical processing in children with 16p11.2 copy number variation. There is a gene dosage-dependent effect on P1 amplitude that merits further investigation. The VEP is directly translatable to animal models, offering a promising way to probe the neurobiological mechanisms underlying cortical dysfunction in this developmental disorder.


Assuntos
Cromossomos Humanos Par 16/genética , Potenciais Evocados Visuais/fisiologia , Adolescente , Criança , Pré-Escolar , Variações do Número de Cópias de DNA , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/fisiopatologia , Eletroencefalografia , Feminino , Deleção de Genes , Duplicação Gênica , Humanos , Masculino , Córtex Visual/diagnóstico por imagem
6.
Ann Neurol ; 78(5): 775-86, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26332183

RESUMO

OBJECTIVE: Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutation of the X-linked MECP2 gene and characterized by developmental regression during the first few years of life. The objective of this study was to investigate if the visual evoked potential (VEP) could be used as an unbiased, quantitative biomarker to monitor brain function in RTT. METHODS: We recorded pattern-reversal VEPs in Mecp2 heterozygous female mice and 34 girls with RTT. The amplitudes and latencies of VEP waveform components were quantified, and were related to disease stage, clinical severity, and MECP2 mutation type in patients. Visual acuity was also assessed in both mice and patients by modulating the spatial frequency of the stimuli. RESULTS: Mecp2 heterozygous female mice and RTT patients exhibited a similar decrease in VEP amplitude that was most striking in the later stages of the disorder. RTT patients also displayed a slower recovery from the principal peak of the VEP response that was impacted by MECP2 mutation type. When the spatial frequency of the stimulus was increased, both patients and mice displayed a deficit in discriminating smaller patterns, indicating lower visual spatial acuity in RTT. INTERPRETATION: VEP is a method that can be used to assess brain function across species and in children with severe disabilities like RTT. Our findings support the introduction of standardized VEP analysis in clinical and research settings to probe the neurobiological mechanism underlying functional impairment and to longitudinally monitor progression of the disorder and response to treatment.


Assuntos
Potenciais Evocados Visuais/fisiologia , Síndrome de Rett/fisiopatologia , Córtex Visual/fisiopatologia , Envelhecimento/fisiologia , Animais , Biomarcadores , Criança , Pré-Escolar , Progressão da Doença , Eletroencefalografia , Feminino , Humanos , Lactente , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Estimulação Luminosa , Acuidade Visual
7.
J Neurosci ; 33(45): 17789-96, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24198369

RESUMO

Visual circuits mature and are refined by sensory experience. However, significant gaps remain in our understanding how deprivation influences the development of visual acuity in mice. Here, we perform a longitudinal study assessing the effects of chronic deprivation on the development of the mouse subcortical and cortical visual circuits using a combination of behavioral optomotor testing, in vivo visual evoked responses (VEP) and single-unit cortical recordings. As previously reported, orientation tuning was degraded and onset of ocular dominance plasticity was delayed and remained open in chronically deprived mice. Surprisingly, we found that the development of optomotor threshold and VEP acuity can occur in an experience-independent manner, although at a significantly slower rate. Moreover, monocular deprivation elicited amblyopia only during a discrete period of development in the dark. The rate of recovery of optomotor threshold upon exposure of deprived mice to light confirmed a maturational transition regardless of visual input. Together our results revealed a dissociable developmental trajectory for visual receptive-field properties in dark-reared mice suggesting a differential role for spontaneous activity within thalamocortical and intracortical circuits.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Privação Sensorial/fisiologia , Visão Monocular/fisiologia , Acuidade Visual/fisiologia , Córtex Visual/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Visual/crescimento & desenvolvimento
8.
Cell ; 147(3): 577-89, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22036566

RESUMO

BMP and Wnt signaling pathways control essential cellular responses through activation of the transcription factors SMAD (BMP) and TCF (Wnt). Here, we show that regeneration of hematopoietic lineages following acute injury depends on the activation of each of these signaling pathways to induce expression of key blood genes. Both SMAD1 and TCF7L2 co-occupy sites with master regulators adjacent to hematopoietic genes. In addition, both SMAD1 and TCF7L2 follow the binding of the predominant lineage regulator during differentiation from multipotent hematopoietic progenitor cells to erythroid cells. Furthermore, induction of the myeloid lineage regulator C/EBPα in erythroid cells shifts binding of SMAD1 to sites newly occupied by C/EBPα, whereas expression of the erythroid regulator GATA1 directs SMAD1 loss on nonerythroid targets. We conclude that the regenerative response mediated by BMP and Wnt signaling pathways is coupled with the lineage master regulators to control the gene programs defining cellular identity.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Hematopoese , Transdução de Sinais , Via de Sinalização Wnt , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Regeneração , Proteína Smad1/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Peixe-Zebra
9.
Neural Plast ; 2011: 921680, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21826280

RESUMO

Cortical circuits in the brain are refined by experience during critical periods early in postnatal life. Critical periods are regulated by the balance of excitatory and inhibitory (E/I) neurotransmission in the brain during development. There is now increasing evidence of E/I imbalance in autism, a complex genetic neurodevelopmental disorder diagnosed by abnormal socialization, impaired communication, and repetitive behaviors or restricted interests. The underlying cause is still largely unknown and there is no fully effective treatment or cure. We propose that alteration of the expression and/or timing of critical period circuit refinement in primary sensory brain areas may significantly contribute to autistic phenotypes, including cognitive and behavioral impairments. Dissection of the cellular and molecular mechanisms governing well-established critical periods represents a powerful tool to identify new potential therapeutic targets to restore normal plasticity and function in affected neuronal circuits.


Assuntos
Transtorno Autístico/psicologia , Período Crítico Psicológico , Animais , Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Transtorno Autístico/fisiopatologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Humanos , Rede Nervosa/crescimento & desenvolvimento , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Sensação , Ácido gama-Aminobutírico/metabolismo
10.
Cell ; 142(1): 133-43, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20603019

RESUMO

Recent genome-wide studies have demonstrated that pausing of RNA polymerase II (Pol II) occurred on many vertebrate genes. By genetic studies in the zebrafish tif1gamma mutant moonshine we found that loss of function of Pol II-associated factors PAF or DSIF rescued erythroid gene transcription in tif1gamma-deficient animals. Biochemical analysis established physical interactions among TIF1gamma, the blood-specific SCL transcription complex, and the positive elongation factors p-TEFb and FACT. Chromatin immunoprecipitation assays in human CD34(+) cells supported a TIF1gamma-dependent recruitment of positive elongation factors to erythroid genes to promote transcription elongation by counteracting Pol II pausing. Our study establishes a mechanism for regulating tissue cell fate and differentiation through transcription elongation.


Assuntos
Eritropoese , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Células Eritroides/metabolismo , Humanos , RNA Polimerase II/metabolismo , Peixe-Zebra/metabolismo
11.
J Neurodev Disord ; 1(2): 172-81, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20664807

RESUMO

UNLABELLED: One unifying explanation for the complexity of Autism Spectrum Disorders (ASD) may lie in the disruption of excitatory/inhibitory (E/I) circuit balance during critical periods of development. We examined whether Parvalbumin (PV)-positive inhibitory neurons, which normally drive experience-dependent circuit refinement (Hensch Nat Rev Neurosci 6:877-888, 1), are disrupted across heterogeneous ASD mouse models. We performed a meta-analysis of PV expression in previously published ASD mouse models and analyzed two additional models, reflecting an embryonic chemical insult (prenatal valproate, VPA) or single-gene mutation identified in human patients (Neuroligin-3, NL-3 R451C). PV-cells were reduced in the neocortex across multiple ASD mouse models. In striking contrast to controls, both VPA and NL-3 mouse models exhibited an asymmetric PV-cell reduction across hemispheres in parietal and occipital cortices (but not the underlying area CA1). ASD mouse models may share a PV-circuit disruption, providing new insight into circuit development and potential prevention by treatment of autism. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11689-009-9023-x) contains supplementary material, which is available to authorized users.

12.
Cell Stem Cell ; 2(2): 183-9, 2008 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-18371439

RESUMO

The zebrafish is a useful model for understanding normal and cancer stem cells, but analysis has been limited to embryogenesis due to the opacity of the adult fish. To address this, we have created a transparent adult zebrafish in which we transplanted either hematopoietic stem/progenitor cells or tumor cells. In a hematopoiesis radiation recovery assay, transplantation of GFP-labeled marrow cells allowed for striking in vivo visual assessment of engraftment from 2 hr-5 weeks posttransplant. Using FACS analysis, both transparent and wild-type fish had equal engraftment, but this could only be visualized in the transparent recipient. In a tumor engraftment model, transplantation of RAS-melanoma cells allowed for visualization of tumor engraftment, proliferation, and distant metastases in as little as 5 days, which is not seen in wild-type recipients until 3 to 4 weeks. This transparent adult zebrafish serves as the ideal combination of both sensitivity and resolution for in vivo stem cell analyses.


Assuntos
Modelos Animais , Pigmentação/genética , Transplante de Células-Tronco , Células-Tronco/ultraestrutura , Peixe-Zebra , Albinismo/genética , Animais , Citometria de Fluxo , Luz , Transplante de Neoplasias
13.
Blood ; 110(10): 3627-36, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17698971

RESUMO

Vascular endothelial growth factor A (VEGFA) and the type III receptor tyrosine kinase receptors (RTKs) are both required for the differentiation of endothelial cells (vasculogenesis) and for the sprouting of new capillaries (angiogenesis). We have isolated a duplicated zebrafish VegfA locus, termed VegfAb, and a duplicate RTK locus with homology to KDR/FLK1 (named Kdrb). Morpholino-disrupted VegfAb embryos develop a normal circulatory system until approximately 2 to 3 days after fertilization (dpf), when defects in angiogenesis permit blood to extravasate into many tissues. Unlike the VegfAa(121) and VegfAa(165) isoforms, the VegfAb isoforms VegfAb(171) and VegfAb(210) are not normally secreted when expressed in mammalian tissue culture cells. The Kdrb locus encodes a 1361-amino acid transmembrane receptor with strong homology to mammalian KDR. Combined knockdown of both RTKs leads to defects in vascular development, suggesting that they cooperate in mediating the vascular effects of VegfA in zebrafish development. Both VegfAa and VegfAb can individually bind and promote phosphorylation of both Flk1 (Kdra) and Kdrb proteins in vitro. Taken together, our data support a model in the zebrafish, in which duplicated VegfA and multiple type III RTKs mediate vascular development.


Assuntos
Vasos Sanguíneos/embriologia , Genes Duplicados , Homologia de Sequência , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Chlorocebus aethiops , Clonagem Molecular , Cricetinae , Cricetulus , Embrião não Mamífero , Isoenzimas/genética , Modelos Biológicos , Dados de Sequência Molecular , Neovascularização Fisiológica/genética , Filogenia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA