Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
ACS Appl Bio Mater ; 6(2): 566-577, 2023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36739562

RESUMO

Bicelles are discoidal lipid nanoparticles (LNPs) in which the planar bilayer and curved rim are, respectively, composed of long- and short-chain lipids. Bicellar LNPs have a hydrophobic core, allowing hydrophobic molecules and large molecular complexes such as quantum dots (QDs) to be encapsulated. In this study, CdSe/ZnS QDs were encapsulated in bicelles made of dipalmitoyl phosphatidylcholine, dihexanoyl phosphatidylcholine, dipalmitoyl phosphatidylglycerol, and distearoyl phosphatidylethanolamine conjugated with polyethylene glycerol amine 2000 to form a well-defined bicelle-QD nanocomplex (known as NANO2-QD or bicelle-QD). The bicelle-QD was then incubated with Hek293t cells and HeLa cells for different periods of time to determine changes in their cellular localization. Bicelle-QDs readily penetrated Hek293t cell membranes within 15 min of incubation, localized to the cytoplasm, and associated with mitochondria and intracellular vesicles. After 1 h, the bicelle-QDs enter the cell nucleus. Large aggregates form throughout the cell after 2 h and QDs are nearly absent from the nucleus by 4 h. Previous reports have demonstrated that CdSe/ZnS QDs can be toxic to cells, and we have found that encapsulating QDs in bicelles can attenuate but did not eliminate cytotoxicity. The present research outcome demonstrates the time-resolved pathway of bicelle-encapsulated QDs in Hek293t cells, morphological evolution in cells over time, and cytotoxicity of the bicelle-QDs, providing important insight into the potential application of the nanocomplex for cellular imaging.


Assuntos
Nanocompostos , Pontos Quânticos , Humanos , Células HeLa , Pontos Quânticos/toxicidade , Pontos Quânticos/química , Células HEK293 , Nanocompostos/toxicidade
2.
J Neurophysiol ; 123(6): 2449-2464, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401131

RESUMO

Somatic mutations have emerged as the likely cause of focal epilepsies associated with developmental malformations and epilepsy-associated glioneuronal tumors (GNT). Somatic BRAFV600E mutations in particular have been detected in the majority of low-grade neuroepithelial tumors (LNETS) and in neurons in focal cortical dysplasias adjacent to epilepsy-associated tumors. Furthermore, conditional expression of an activating BRAF mutation in neocortex causes seizures in mice. In this study we characterized the cellular electrophysiology of layer 2/3 neocortical pyramidal neurons induced to express BRAFV600E from neural progenitor stages. In utero electroporation of a piggyBac transposase plasmid system was used to introduce transgenes expressing BRAF wild type (BRAFwt), BRAFV600E, and/or enhanced green fluorescent protein (eGFP) and monomeric red fluorescent protein (mRFP) into radial glia progenitors in mouse embryonic cortex. Whole cell patch-clamp recordings of pyramidal neurons in slices prepared from both juvenile and adult mice showed that BRAFV600E resulted in neurons with a distinct hyperexcitable phenotype characterized by depolarized resting membrane potentials, increased input resistances, lowered action potential (AP) thresholds, and increased AP firing frequencies. Some of the BRAFV600E-expressing neurons normally destined for upper cortical layers by their birthdate were stalled in their migration and occupied lower cortical layers. BRAFV600E-expressing neurons also displayed increased hyperpolarization-induced inward currents (Ih) and decreased sustained potassium currents. Neurons adjacent to BRAFV600E transgene-expressing neurons, and neurons with TSC1 genetically deleted by CRISPR or those induced to carry PIK3CAE545K transgenes, did not show an excitability phenotype similar to that of BRAFV600E-expressing neurons. Together, these results indicate that BRAFV600E leads to a distinct hyperexcitable neuronal phenotype.NEW & NOTEWORTHY This study is the first to report the cell autonomous effects of BRAFV600E mutations on the intrinsic neuronal excitability. We show that BRAFV600E alters multiple electrophysiological parameters in neocortical neurons. Similar excitability changes did not occur in cells neighboring BRAFV600E-expressing neurons, after overexpression of wild-type BRAF transgenes, or after introduction of mutations affecting the mammalian target of rapamycin (mTOR) or the catalytic subunit of phosphoinositide 3-kinase (PIK3CA). We conclude that BRAFV600E causes a distinct, cell autonomous, highly excitable neuronal phenotype when introduced somatically into neocortical neuronal progenitors.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Neocórtex/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Células Piramidais/fisiologia , Animais , Excitabilidade Cortical/fisiologia , Fenômenos Eletrofisiológicos/genética , Eletroporação , Embrião de Mamíferos , Feminino , Masculino , Camundongos , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Gravidez , Proteínas Proto-Oncogênicas B-raf/genética , Células Piramidais/metabolismo
3.
Elife ; 82019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31025941

RESUMO

Dravet syndrome (DS) is a form of epilepsy with a high incidence of sudden unexpected death in epilepsy (SUDEP). Respiratory failure is a leading cause of SUDEP, and DS patients' frequently exhibit disordered breathing. Despite this, mechanisms underlying respiratory dysfunction in DS are unknown. We found that mice expressing a DS-associated Scn1a missense mutation (A1783V) conditionally in inhibitory neurons (Slc32a1cre/+::Scn1aA1783V fl/+; defined as Scn1aΔE26) exhibit spontaneous seizures, die prematurely and present a respiratory phenotype including hypoventilation, apnea, and a diminished ventilatory response to CO2. At the cellular level in the retrotrapezoid nucleus (RTN), we found inhibitory neurons expressing the Scn1a A1783V variant are less excitable, whereas glutamatergic chemosensitive RTN neurons, which are a key source of the CO2/H+-dependent drive to breathe, are hyper-excitable in slices from Scn1aΔE26 mice. These results show loss of Scn1a function can disrupt respiratory control at the cellular and whole animal levels.


Assuntos
Epilepsias Mioclônicas/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Respiração/genética , Convulsões/genética , Potenciais de Ação/genética , Animais , Dióxido de Carbono/toxicidade , Modelos Animais de Doenças , Epilepsias Mioclônicas/fisiopatologia , Humanos , Camundongos , Mutação de Sentido Incorreto/genética , Neurônios/metabolismo , Neurônios/patologia , Convulsões/fisiopatologia , Morte Súbita Inesperada na Epilepsia/patologia
4.
Cell Stem Cell ; 20(4): 421-422, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28388422

RESUMO

Cellular changes underlying malformations of human cortical development may be difficult to identify with traditional mouse models. Two recent Cell Stem Cell papers, Li et al. (2017) and Bershteyn et al. (2017), use human cerebral organoids to identify specific cellular defects in neurogenesis that may explain PTEN-related macrocephaly and Miller-Dieker lissencephaly.


Assuntos
Neocórtex , Organoides , Animais , Humanos , Masculino , Camundongos , Neurogênese , Especificidade da Espécie , Células-Tronco
5.
Brain Lang ; 172: 30-38, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-25989970

RESUMO

Dyslexia is a learning disability characterized by difficulty learning to read and write. The underlying biological and genetic etiology remains poorly understood. One candidate gene, dyslexia susceptibility 1 candidate 1 (DYX1C1), has been shown to be associated with deficits in short-term memory in dyslexic populations. The purpose of the current study was to examine the behavioral phenotype of a mouse model with a homozygous conditional (forebrain) knockout of the rodent homolog Dyx1c1. Twelve Dyx1c1 conditional homozygous knockouts, 7 Dyx1c1 conditional heterozygous knockouts and 6 wild-type controls were behaviorally assessed. Mice with the homozygous Dyx1c1 knockout showed deficits on memory and learning, but not on auditory or motor tasks. These findings affirm existing evidence that DYX1C1 may play an underlying role in the development of neural systems important to learning and memory, and disruption of this function could contribute to the learning deficits seen in individuals with dyslexia.


Assuntos
Dislexia/genética , Predisposição Genética para Doença , Aprendizagem/fisiologia , Transtornos da Memória/genética , Mutação , Proteínas do Tecido Nervoso/genética , Animais , Modelos Animais de Doenças , Genótipo , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Leitura
6.
J Comp Neurol ; 525(5): 1291-1311, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27804142

RESUMO

Collybistin (CB) is a guanine nucleotide exchange factor selectively localized to γ-aminobutyric acid (GABA)ergic and glycinergic postsynapses. Active CB interacts with gephyrin, inducing the submembranous clustering and the postsynaptic accumulation of gephyrin, which is a scaffold protein that recruits GABAA receptors (GABAA Rs) at the postsynapse. CB is expressed with or without a src homology 3 (SH3) domain. We have previously reported the effects on GABAergic synapses of the acute overexpression of CBSH3- or CBSH3+ in cultured hippocampal (HP) neurons. In the present communication, we are studying the effects on GABAergic synapses after chronic in vivo transgenic expression of CB2SH3- or CB2SH3+ in neurons of the adult rat cerebral cortex. The embryonic precursors of these cortical neurons were in utero electroporated with CBSH3- or CBSH3+ DNAs, migrated to the appropriate cortical layer, and became integrated in cortical circuits. The results show that: 1) the strength of inhibitory synapses in vivo can be enhanced by increasing the expression of CB in neurons; and 2) there are significant differences in the results between in vivo and in culture studies. J. Comp. Neurol. 525:1291-1311, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Córtex Cerebral/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Animais , Córtex Cerebral/crescimento & desenvolvimento , Embrião de Mamíferos , Feminino , Imunofluorescência , Processamento de Imagem Assistida por Computador , Masculino , Microscopia Confocal , Técnicas de Patch-Clamp , Ratos , Ratos Transgênicos , Ratos Wistar , Sinapses/metabolismo
7.
J Neurosci ; 36(17): 4895-906, 2016 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27122044

RESUMO

UNLABELLED: Dyslexia is the most common developmental language disorder and is marked by deficits in reading and phonological awareness. One theory of dyslexia suggests that the phonological awareness deficit is due to abnormal auditory processing of speech sounds. Variants in DCDC2 and several other neural migration genes are associated with dyslexia and may contribute to auditory processing deficits. In the current study, we tested the hypothesis that RNAi suppression of Dcdc2 in rats causes abnormal cortical responses to sound and impaired speech sound discrimination. In the current study, rats were subjected in utero to RNA interference targeting of the gene Dcdc2 or a scrambled sequence. Primary auditory cortex (A1) responses were acquired from 11 rats (5 with Dcdc2 RNAi; DC-) before any behavioral training. A separate group of 8 rats (3 DC-) were trained on a variety of speech sound discrimination tasks, and auditory cortex responses were acquired following training. Dcdc2 RNAi nearly eliminated the ability of rats to identify specific speech sounds from a continuous train of speech sounds but did not impair performance during discrimination of isolated speech sounds. The neural responses to speech sounds in A1 were not degraded as a function of presentation rate before training. These results suggest that A1 is not directly involved in the impaired speech discrimination caused by Dcdc2 RNAi. This result contrasts earlier results using Kiaa0319 RNAi and suggests that different dyslexia genes may cause different deficits in the speech processing circuitry, which may explain differential responses to therapy. SIGNIFICANCE STATEMENT: Although dyslexia is diagnosed through reading difficulty, there is a great deal of variation in the phenotypes of these individuals. The underlying neural and genetic mechanisms causing these differences are still widely debated. In the current study, we demonstrate that suppression of a candidate-dyslexia gene causes deficits on tasks of rapid stimulus processing. These animals also exhibited abnormal neural plasticity after training, which may be a mechanism for why some children with dyslexia do not respond to intervention. These results are in stark contrast to our previous work with a different candidate gene, which caused a different set of deficits. Our results shed some light on possible neural and genetic mechanisms causing heterogeneity in the dyslexic population.


Assuntos
Estimulação Acústica/métodos , Dislexia/genética , Proteínas Associadas aos Microtúbulos/genética , Som , Percepção da Fala/fisiologia , Animais , Córtex Auditivo/fisiologia , Percepção Auditiva , Feminino , Masculino , Plasticidade Neuronal/genética , Interferência de RNA , Ratos , Percepção da Fala/genética , Percepção da Fala/efeitos da radiação
8.
Cereb Cortex ; 26(9): 3705-3718, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26250775

RESUMO

Variants in DCDC2 have been associated with reading disability in humans, and targeted mutation of Dcdc2 in mice causes impairments in both learning and sensory processing. In this study, we sought to determine whether Dcdc2 mutation affects functional synaptic circuitry in neocortex. We found mutation in Dcdc2 resulted in elevated spontaneous and evoked glutamate release from neurons in somatosensory cortex. The probability of release was decreased to wild-type level by acute application of N-methyl-d-aspartate receptor (NMDAR) antagonists when postsynaptic NMDARs were blocked by intracellular MK-801, and could not be explained by elevated ambient glutamate, suggesting altered, nonpostsynaptic NMDAR activation in the mutants. In addition, we determined that the increased excitatory transmission was present at layer 4-layer 4 but not thalamocortical connections in Dcdc2 mutants, and larger evoked synaptic release appeared to enhance the NMDAR-mediated effect. These results demonstrate an NMDAR activation-gated, increased functional excitatory connectivity between layer 4 lateral connections in somatosensory neocortex of the mutants, providing support for potential changes in cortical connectivity and activation resulting from mutation of dyslexia candidate gene Dcdc2.


Assuntos
Ácido Glutâmico/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neocórtex/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Animais , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Mutação , Neurotransmissores/metabolismo , Córtex Somatossensorial/fisiologia , Regulação para Cima/fisiologia
9.
PLoS One ; 10(4): e0122286, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25850004

RESUMO

Understanding single-neuron computations and encoding performed by spike-generation mechanisms of cortical neurons is one of the central challenges for cell electrophysiology and computational neuroscience. An established paradigm to study spike encoding in controlled conditions in vitro uses intracellular injection of a mixture of signals with fluctuating currents that mimic in vivo-like background activity. However this technique has two serious limitations: it uses current injection, while synaptic activation leads to changes of conductance, and current injection is technically most feasible in the soma, while the vast majority of synaptic inputs are located on the dendrites. Recent progress in optogenetics provides an opportunity to circumvent these limitations. Transgenic expression of light-activated ionic channels, such as Channelrhodopsin2 (ChR2), allows induction of controlled conductance changes even in thin distant dendrites. Here we show that photostimulation provides a useful extension of the tools to study neuronal encoding, but it has its own limitations. Optically induced fluctuating currents have a low cutoff (~70 Hz), thus limiting the dynamic range of frequency response of cortical neurons. This leads to severe underestimation of the ability of neurons to phase-lock their firing to high frequency components of the input. This limitation could be worked around by using short (2 ms) light stimuli which produce membrane potential responses resembling EPSPs by their fast onset and prolonged decay kinetics. We show that combining application of short light stimuli to different parts of dendritic tree for mimicking distant EPSCs with somatic injection of fluctuating current that mimics fluctuations of membrane potential in vivo, allowed us to study fast encoding of artificial EPSPs photoinduced at different distances from the soma. We conclude that dendritic photostimulation of ChR2 with short light pulses provides a powerful tool to investigate population encoding of simulated synaptic potentials generated in dendrites at different distances from the soma.


Assuntos
Neurônios/citologia , Optogenética/métodos , Animais , Encéfalo/citologia , Channelrhodopsins , Dendritos/metabolismo , Dendritos/efeitos da radiação , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Cinética , Luz , Camundongos , Neurônios/metabolismo , Neurônios/efeitos da radiação
10.
Am J Hum Genet ; 96(1): 81-92, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25557784

RESUMO

Nephronophthisis-related ciliopathies (NPHP-RC) are recessive diseases characterized by renal dysplasia or degeneration. We here identify mutations of DCDC2 as causing a renal-hepatic ciliopathy. DCDC2 localizes to the ciliary axoneme and to mitotic spindle fibers in a cell-cycle-dependent manner. Knockdown of Dcdc2 in IMCD3 cells disrupts ciliogenesis, which is rescued by wild-type (WT) human DCDC2, but not by constructs that reflect human mutations. We show that DCDC2 interacts with DVL and DCDC2 overexpression inhibits ß-catenin-dependent Wnt signaling in an effect additive to Wnt inhibitors. Mutations detected in human NPHP-RC lack these effects. A Wnt inhibitor likewise restores ciliogenesis in 3D IMCD3 cultures, emphasizing the importance of Wnt signaling for renal tubulogenesis. Knockdown of dcdc2 in zebrafish recapitulates NPHP-RC phenotypes, including renal cysts and hydrocephalus, which is rescued by a Wnt inhibitor and by WT, but not by mutant, DCDC2. We thus demonstrate a central role of Wnt signaling in the pathogenesis of NPHP-RC, suggesting an avenue for potential treatment of NPHP-RC.


Assuntos
Doenças Renais Císticas/genética , Proteínas Associadas aos Microtúbulos/genética , Via de Sinalização Wnt/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Cílios/genética , Cílios/patologia , Biologia Computacional , Proteínas Desgrenhadas , Éxons , Células HEK293 , Humanos , Rim/patologia , Camundongos , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Células NIH 3T3 , Fenótipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Peixe-Zebra/genética , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo
11.
J Comp Neurol ; 523(9): 1359-78, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25565602

RESUMO

We studied the effect of clonal overexpression of neuroligin 3 (NL3) or neuroligin 2 (NL2) in the adult rat cerebral cortex following in utero electroporation (IUEP) at embryonic stage E14. Overexpression of NL3 leads to a large increase in vesicular gamma-aminobutyric acid (GABA) transporter (vGAT) and glutamic acid decarboxylase (GAD)65 in the GABAergic contacts that the overexpressing neurons receive. Overexpression of NL2 produced a similar effect but to a lesser extent. In contrast, overexpression of NL3 or NL2 after IUEP does not affect vesicular glutamate transporter 1 (vGlut1) in the glutamatergic contacts that the NL3 or NL2-overexpressing neurons receive. The NL3 or NL2-overexpressing neurons do not show increased innervation by parvalbumin-containing GABAergic terminals or increased parvalbumin in the same terminals that show increased vGAT. These results indicate that the observed increase in vGAT and GAD65 is not due to increased GABAergic innervation but to increased expression of vGAT and GAD65 in the GABAergic contacts that NL3 or NL2-overexpressing neurons receive. The majority of bright vGAT puncta contacting the NL3-overexpressing neurons have no gephyrin juxtaposed to them, indicating that many of these contacts are nonsynaptic. This contrasts with the majority of the NL2-overexpressing neurons, which show plenty of synaptic gephyrin clusters juxtaposed to vGAT. Besides having an effect on GABAergic contacts, overexpression of NL3 interferes with the neuronal radial migration, in the cerebral cortex, of the neurons overexpressing NL3.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular/fisiologia , Córtex Cerebral/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Adjuvantes Imunológicos , Animais , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Eletroporação , Glutamato Descarboxilase/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Parvalbuminas/metabolismo , Ratos Sprague-Dawley , Ratos Wistar , Sinapses/metabolismo , Transfecção , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
12.
PLoS One ; 9(11): e113873, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25419571

RESUMO

ArcLight, a genetically encoded fluorescent protein voltage probe with a large ΔF/ΔV, is a fusion between the voltage sensing domain of the Ciona instestinalis voltage sensitive phosphatase and super ecliptic pHluorin carrying a single mutation (A227D in the fluorescent protein). Without this mutation the probe produces only a very small change in fluorescence in response to voltage deflections (∼ 1%). The large signal afforded by this mutation allows optical detection of action potentials and sub-threshold electrical events in single-trials in vitro and in vivo. However, it is unclear how this single mutation produces a probe with such a large modulation of its fluorescence output with changes in membrane potential. In this study, we identified which residues in super ecliptic pHluorin (vs eGFP) are critical for the ArcLight response, as a similarly constructed probe based on eGFP also exhibits large response amplitude if it carries these critical residues. We found that D147 is responsible for determining the pH sensitivity of the fluorescent protein used in these probes but by itself does not result in a voltage probe with a large signal. We also provide evidence that the voltage dependent signal of ArcLight is not simply sensing environmental pH changes. A two-photon polarization microscopy study showed that ArcLight's response to changes in membrane potential includes a reorientation of the super ecliptic pHluorin. We also explored different changes including modification of linker length, deletion of non-essential amino acids in the super ecliptic pHluorin, adding a farnesylation site, using tandem fluorescent proteins and other pH sensitive fluorescent proteins.


Assuntos
Potenciais de Ação , Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/química , Proteínas Luminescentes/química , Proteínas Recombinantes de Fusão/química , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Células Cultivadas , Fluorescência , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Cinética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Potenciais da Membrana , Microscopia Confocal , Mutação de Sentido Incorreto , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Prenilação , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Fluorescência
13.
Nat Neurosci ; 17(10): 1322-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25151262

RESUMO

Lineage progression and diversification is regulated by the coordinated action of unique sets of transcription factors. Oligodendrocytes (OL) and astrocytes (AS) comprise the glial sub-lineages in the CNS, and the manner in which their associated regulatory factors orchestrate lineage diversification during development and disease remains an open question. Sox10 and NFIA are key transcriptional regulators of gliogenesis associated with OL and AS. We found that NFIA inhibited Sox10 induction of OL differentiation through direct association and antagonism of its function. Conversely, we found that Sox10 antagonized NFIA function and suppressed AS differentiation in mouse and chick systems. Using this developmental paradigm as a model for glioma, we found that this relationship similarly regulated the generation of glioma subtypes. Our results describe the antagonistic relationship between Sox10 and NFIA that regulates the balance of OL and AS fate during development and demonstrate for the first time, to the best of our knowledge, that the transcriptional processes governing glial sub-lineage diversification oversee the generation of glioma subtypes.


Assuntos
Glioma/classificação , Glioma/metabolismo , Neuroglia/metabolismo , Fatores de Transcrição SOXE/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Galinha , Imunoprecipitação da Cromatina , Eletroporação , Embrião de Mamíferos , Glioma/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/efeitos dos fármacos , Fator de Transcrição 2 de Oligodendrócitos , Fatores de Transcrição SOXE/genética , Transfecção
14.
Cold Spring Harb Protoc ; 2014(7): 741-9, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24987137

RESUMO

In utero electroporation (IUE) is an effective transfection method for delivering plasmid DNA into neural progenitor cells and neurons of mammalian neocortex in vivo. Although IUE is effective at delivering multiple DNA plasmids into populations of cells, unfortunately plasmids delivered into neural progenitor cells remain largely episomal and often get inactivated or lost after cell division. This results in a form of "birthdate" labeling in which only the cell types that do not undergo a second cell division continue to express the transfected plasmids. This limits the application of IUE with standard plasmids and precludes its use in experiments where manipulating or labeling the complete cell lineage of a progenitor is desired. To circumvent this episomal loss of plasmid in IUE, we have used a binary piggyBac transposon system to induce nonviral genomic integration of transgenes. These transgenes do not appear to inactivate after cell division, and this results in stable somatic cellular transgenesis of neurons and glia. Like standard IUE, the system can be used with multiple combinations of plasmids to achieve multicolor labeling and both loss-of-function and gain-of-function manipulations. In this protocol, we describe the method for delivering a binary piggyBac transposon plasmid system by IUE.


Assuntos
Cromossomos Artificiais Bacterianos/metabolismo , Elementos de DNA Transponíveis/genética , Eletroporação , Técnicas de Transferência de Genes , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Animais , Cromossomos Artificiais Bacterianos/genética , Feminino , Gravidez , Ratos
15.
PLoS One ; 9(5): e98439, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24871331

RESUMO

In utero RNAi of the dyslexia-associated gene Kiaa0319 in rats (KIA-) degrades cortical responses to speech sounds and increases trial-by-trial variability in onset latency. We tested the hypothesis that KIA- rats would be impaired at speech sound discrimination. KIA- rats needed twice as much training in quiet conditions to perform at control levels and remained impaired at several speech tasks. Focused training using truncated speech sounds was able to normalize speech discrimination in quiet and background noise conditions. Training also normalized trial-by-trial neural variability and temporal phase locking. Cortical activity from speech trained KIA- rats was sufficient to accurately discriminate between similar consonant sounds. These results provide the first direct evidence that assumed reduced expression of the dyslexia-associated gene KIAA0319 can cause phoneme processing impairments similar to those seen in dyslexia and that intensive behavioral therapy can eliminate these impairments.


Assuntos
Moléculas de Adesão Celular/deficiência , Discriminação Psicológica/fisiologia , Dislexia/genética , Plasticidade Neuronal/genética , Percepção da Fala/genética , Animais , Moléculas de Adesão Celular/genética , Feminino , Masculino , Plasticidade Neuronal/fisiologia , Fonética , Interferência de RNA , Ratos , Ratos Wistar , Percepção da Fala/fisiologia , Fatores de Tempo
16.
J Neurosci ; 34(15): 5311-21, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24719109

RESUMO

KCNQ2 and KCNQ3 potassium channels have emerged as central regulators of pyramidal neuron excitability and spiking behavior. However, despite an abundance of evidence demonstrating that KCNQ2/3 heteromers underlie critical potassium conductances, it is unknown whether KCNQ2, KCNQ3, or both are obligatory for maintaining normal pyramidal neuron excitability. Here, we demonstrate that conditional deletion of Kcnq2 from cerebral cortical pyramidal neurons in mice results in abnormal electrocorticogram activity and early death, whereas similar deletion of Kcnq3 does not. At the cellular level, Kcnq2-null, but not Kcnq3-null, CA1 pyramidal neurons show increased excitability manifested as a decreased medium afterhyperpolarization and a longer-lasting afterdepolarization. As a result, these Kcnq2-deficient neurons are hyperexcitable, responding to current injections with an increased number and frequency of action potentials. Biochemically, the Kcnq2 deficiency secondarily results in a substantial loss of KCNQ3 and KCNQ5 protein levels, whereas loss of Kcnq3 only leads to a modest reduction of other KCNQ channels. Consistent with this finding, KCNQ allosteric activators can still markedly dampen neuronal excitability in Kcnq3-null pyramidal neurons, but have only weak effects in Kcnq2-null pyramidal neurons. Together, our data reveal the indispensable function of KCNQ2 channels at both the cellular and systems levels, and demonstrate that pyramidal neurons have near normal excitability in the absence of KCNQ3 channels.


Assuntos
Potenciais de Ação , Epilepsia/genética , Deleção de Genes , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células Piramidais/fisiologia , Animais , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Células Piramidais/metabolismo
17.
Mol Cancer Res ; 12(5): 742-53, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24501428

RESUMO

UNLABELLED: The etiology of central nervous system (CNS) tumor heterogeneity is unclear. To clarify this issue, a novel animal model was developed of glioma and atypical teratoid/rhabdoid-like tumor (ATRT) produced in rats by nonviral cellular transgenesis initiated in utero. This model system affords the opportunity for directed oncogene expression, clonal labeling, and addition of tumor-modifying transgenes. By directing HRasV12 and AKT transgene expression in different cell populations with promoters that are active ubiquitously (CAG promoter), astrocyte-selective (glial fibrillary acidic protein promoter), or oligodendrocyte-selective (myelin basic protein promoter) we generated glioblastoma multiforme and anaplastic oligoastrocytoma, respectively. Importantly, the glioblastoma multiforme and anaplastic oligoastrocytoma tumors were distinguishable at both the cellular and molecular level. Furthermore, proneural basic helix-loop-helix (bHLH) transcription factors, Ngn2 (NEUROG2) or NeuroD1, were expressed along with HRasV12 and AKT in neocortical radial glia, leading to the formation of highly lethal ATRT like tumors. This study establishes a unique model in which determinants of CNS tumor diversity can be parsed out and reveals that both mutation and expression of neurogenic bHLH transcription factors contribute to CNS tumor diversity. IMPLICATIONS: A novel CNS tumor model reveals that oncogenic events occurring in disparate cell types and/or molecular contexts lead to different tumor types; these findings shed light on the sources of brain tumor heterogeneity.


Assuntos
Neoplasias Encefálicas/genética , Modelos Animais de Doenças , Glioblastoma/genética , Animais , Neoplasias Encefálicas/patologia , Células Cultivadas , Feminino , Glioblastoma/patologia , Camundongos , Gravidez , Ratos , Ratos Wistar
18.
Cereb Cortex ; 24(2): 508-20, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23118195

RESUMO

Progenitors within the neocortical ventricular zone (VZ) first generate pyramidal neurons and then astrocytes. We applied novel piggyBac transposase lineage tracking methods to fate-map progenitor populations positive for Nestin or glutamate and aspartate transpoter (GLAST) promoter activities in the rat neocortex. GLAST+ and Nestin+ progenitors at embryonic day 13 (E13) produce lineages containing similar rations of neurons and astrocytes. By E15, the GLAST+ progenitor population diverges significantly to produce lineages with 5-10-fold more astrocytes relative to neurons than generated by the Nestin+ population. To determine when birth-dated progeny within GLAST+ and Nestin+ populations diverge, we used a Cre/loxP fate-mapping system in which plasmids are lost after a cell division. By E18, birth-dated progeny of GLAST+ progenitors give rise to 2-3-fold more neocortical astrocytes than do Nestin+ progenitors. Finally, we used a multicolor clonal labeling method to show that the GLAST+ population labeled at E15 generates astrocyte progenitors that produce larger, spatially restricted, clonal clusters than the Nestin+ population. This study provides in vivo evidence that by mid-corticogenesis (E15), VZ progenitor populations have significantly diversified in terms of their potential to generate astrocytes and neurons.


Assuntos
Astrócitos/fisiologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Neocórtex/embriologia , Neocórtex/fisiologia , Nestina/metabolismo , Células-Tronco Neurais/fisiologia , Animais , Linhagem da Célula/fisiologia , Células Cultivadas , Eletroporação , Células HEK293 , Humanos , Integrases/genética , Integrases/metabolismo , Neurogênese/fisiologia , Neurônios/fisiologia , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Transposases/genética , Transposases/metabolismo
19.
Nat Genet ; 45(9): 995-1003, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23872636

RESUMO

DYX1C1 has been associated with dyslexia and neuronal migration in the developing neocortex. Unexpectedly, we found that deleting exons 2-4 of Dyx1c1 in mice caused a phenotype resembling primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease, laterality defects and male infertility. This phenotype was confirmed independently in mice with a Dyx1c1 c.T2A start-codon mutation recovered from an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Morpholinos targeting dyx1c1 in zebrafish also caused laterality and ciliary motility defects. In humans, we identified recessive loss-of-function DYX1C1 mutations in 12 individuals with PCD. Ultrastructural and immunofluorescence analyses of DYX1C1-mutant motile cilia in mice and humans showed disruptions of outer and inner dynein arms (ODAs and IDAs, respectively). DYX1C1 localizes to the cytoplasm of respiratory epithelial cells, its interactome is enriched for molecular chaperones, and it interacts with the cytoplasmic ODA and IDA assembly factor DNAAF2 (KTU). Thus, we propose that DYX1C1 is a newly identified dynein axonemal assembly factor (DNAAF4).


Assuntos
Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo , Cílios/genética , Cílios/metabolismo , Proteínas do Tecido Nervoso/genética , Animais , Cílios/ultraestrutura , Modelos Animais de Doenças , Epêndima/metabolismo , Epêndima/patologia , Técnicas de Silenciamento de Genes , Ordem dos Genes , Marcação de Genes , Humanos , Espaço Intracelular/metabolismo , Síndrome de Kartagener/genética , Síndrome de Kartagener/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Ligação Proteica , Transporte Proteico , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA