Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
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.
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
4.
Development ; 142(20): 3601-11, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26400094

RESUMO

The ability to induce targeted mutations in somatic cells in developing organisms and then track the fates of those cells is a powerful approach both for studying neural development and for modeling human disease. The CRISPR/Cas9 system allows for such targeted mutagenesis, and we therefore tested it in combination with a piggyBac transposase lineage labeling system to track the development of neocortical neural progenitors with targeted mutations in genes linked to neurodevelopmental disruptions and tumor formation. We show that sgRNAs designed to target PTEN successfully decreased PTEN expression, and led to neuronal hypertrophy and altered neuronal excitability. Targeting NF1, by contrast, caused increased astrocytogenesis at the expense of neurogenesis, and combined targeting of three tumor suppressors (PTEN, NF1 and P53) resulted in formation of glioblastoma tumors. Our results demonstrate that CRISPR/Cas9 combined with piggyBac transposase lineage labeling can produce unique models of neurodevelopmental disruption and tumors caused by somatic mutation in neural progenitors.


Assuntos
Astrócitos/citologia , Sistemas CRISPR-Cas/genética , Cromossomos Artificiais Bacterianos , Células-Tronco/citologia , Transposases/genética , Animais , Encéfalo/metabolismo , Neoplasias Encefálicas/patologia , Linhagem da Célula , Eletroporação , Feminino , Glioblastoma/patologia , Humanos , Mutagênese , Mutação , Neurofibromina 1/metabolismo , Neurônios/citologia , PTEN Fosfo-Hidrolase/metabolismo , Gravidez , Prenhez , Ratos , Ratos Wistar , Proteína Supressora de Tumor p53/metabolismo
5.
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
6.
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
7.
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
8.
Development ; 139(13): 2299-307, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22627280

RESUMO

NG2-expressing cells (NG2 cells or polydendrocytes) generate oligodendrocytes throughout the CNS and a subpopulation of protoplasmic astrocytes in the gray matter of the ventral forebrain. The mechanisms that regulate their oligodendrocyte or astrocyte fate and the degree to which they exhibit lineage plasticity in vivo have remained unclear. The basic helix-loop-helix transcription factor Olig2 is required for oligodendrocyte specification and differentiation. We have found that Olig2 expression is spontaneously downregulated in NG2 cells in the normal embryonic ventral forebrain as they differentiate into astrocytes. To further examine the role of Olig2 in NG2 cell fate determination, we used genetic fate mapping of NG2 cells in constitutive and tamoxifen-inducible Olig2 conditional knockout mice in which Olig2 was deleted specifically in NG2 cells. Constitutive deletion of Olig2 in NG2 cells in the neocortex and corpus callosum but not in ventral forebrain caused them to convert their fate into astrocytes, with a concomitant severe reduction in the number of oligodendrocytes and myelin. Deletion of Olig2 in NG2 cells in perinatal mice also resulted in astrocyte generation from neocortical NG2 cells. These observations indicate that the developmental fate of NG2 cells can be switched by altering a single transcription factor Olig2.


Assuntos
Astrócitos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Animais , Antígenos/análise , Diferenciação Celular , Linhagem da Célula , Corpo Caloso/crescimento & desenvolvimento , Corpo Caloso/metabolismo , Regulação para Baixo , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neocórtex/crescimento & desenvolvimento , Neocórtex/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Proteoglicanas/análise
9.
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
10.
Proc Natl Acad Sci U S A ; 108(40): 16789-94, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21921234

RESUMO

Neurogenesis requires mechanisms that coordinate early cell-fate decisions, migration, and terminal differentiation. Here, we show that the transcriptional repressor, repressor element 1 silencing transcription factor (REST), regulates radial migration and the timing of neural progenitor differentiation during neocortical development, and that the regulation is contingent upon differential REST levels. Specifically, a sustained presence of REST blocks migration and greatly delays--but does not prevent--neuronal differentiation, resulting in a subcortical band heterotopia-like phenotype, reminiscent of loss of doublecortin. We further show that doublecortin is a direct gene target of REST, and that its overexpression rescues, at least in part, the aberrant phenotype caused by persistent presence of REST. Our studies support the view that the targeted down-regulation of REST to low levels in neural progenitors, and its subsequent disappearance during neurogenesis, is critical for timing the spatiotemporal transition of neural progenitor cells to neurons.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Neocórtex/embriologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Proteínas Repressoras/metabolismo , Animais , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Proteínas Correpressoras , Primers do DNA/genética , DNA Complementar/genética , Proteínas do Domínio Duplacortina , Eletroporação , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/metabolismo , Proteínas Repressoras/genética
11.
Stem Cells ; 30(9): 1852-62, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22753232

RESUMO

Human embryonic stem cell-derived neuronal progenitors (hNPs) provide a potential source for cellular replacement following neurodegenerative diseases. One of the greatest challenges for future neuron replacement therapies will be to control extensive cell proliferation and stimulate cell migration of transplanted cells. The doublecortin (DCX) gene encodes the protein DCX, a microtubule-associated protein essential for the migration of neurons in the human brain. In this study, we tested whether increasing the expression of DCX in hNPs would favorably alter their proliferation and migration. Migration and proliferation of hNPs was compared between hNPs expressing a bicistronic DCX/IRES-GFP transgene and those expressing a green fluorescent protein (GFP) transgene introduced by piggyBac-mediated transposition. The DCX-transfected hNPs showed a significant decrease in their proliferation and migrated significantly further on two different substrates, Matrigel and brain slices. Additionally, a dense network of nestin-positive (+) and vimentin+ fibers were found to extend from neurospheres transplanted onto brain slices, and this fiber growth was increased from neurospheres containing DCX-transfected hNPs. In summary, our results show that increased DCX expression inhibits proliferation and promotes migration of hNPs.


Assuntos
Movimento Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Neurônios/metabolismo , Neuropeptídeos/biossíntese , Aminoácidos , Animais , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Células-Tronco Embrionárias/citologia , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neuropeptídeos/genética , Prosencéfalo/citologia , Transfecção , Transgenes
12.
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
13.
Neurobiol Dis ; 38(2): 173-80, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19616627

RESUMO

Reading Disability (RD) is a significant impairment in reading accuracy, speed and/or comprehension despite adequate intelligence and educational opportunity. RD affects 5-12% of readers, has a well-established genetic risk, and is of unknown neurobiological cause or causes. In this review we discuss recent findings that revealed neuroanatomic anomalies in RD, studies that identified 3 candidate genes (KIAA0319, DYX1C1, and DCDC2), and compelling evidence that potentially link the function of candidate genes to the neuroanatomic anomalies. A hypothesis has emerged in which impaired neuronal migration is a cellular neurobiological antecedent to RD. We critically evaluate the evidence for this hypothesis, highlight missing evidence, and outline future research efforts that will be required to develop a more complete cellular neurobiology of RD.


Assuntos
Córtex Cerebral/fisiopatologia , Dislexia/genética , Dislexia/fisiopatologia , Neurônios/fisiologia , Movimento Celular/genética , Proteínas do Citoesqueleto , Predisposição Genética para Doença , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética
14.
Cereb Cortex ; 19 Suppl 1: i120-5, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19395528

RESUMO

In utero electroporation (IUE) has become a method of choice for rapid gain and loss of function studies in embryonic cerebral cortex. In this review we highlight some of the proven and recent advances in IUE technology that make it applicable to an increasingly wide array of experiments requiring spatial and temporal control of gene expression. Recently, cell-type-specific promoters and tamoxifen-gated cre-recombinase have been shown to work effectively with IUE. Experiments can now be designed and carried out to test whether and which cell-type-specific mechanisms operate within defined periods of neuronal migration and maturation. We have recently adapted this conditional expression approach to implement conditional rescue experiments. In conditional rescue, expression of an RNA interference (RNAi) target is restored by tamoxifen-induced cre-mediated recombination. An initial disruption in migration, and resultant malformation, caused by DCX RNAi was reversed by delayed re-expression of Dcx. In the future, combinations of spatially directed, cell-type-specific, and tamoxifen-gated transgene expression can be used to address the complex mechanisms likely to operate during development of cerebral cortex.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/fisiologia , Eletroporação/métodos , Engenharia Genética/métodos , Neurônios/fisiologia , Células-Tronco/fisiologia , Transfecção/métodos , Animais , Movimento Celular/fisiologia , Córtex Cerebral/citologia , Neurônios/citologia , Células-Tronco/citologia
15.
Nat Neurosci ; 9(10): 1213-7, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17001339

RESUMO

All four genes thus far linked to developmental dyslexia participate in brain development, and abnormalities in brain development are increasingly reported in dyslexia. Comparable abnormalities induced in young rodent brains cause auditory and cognitive deficits, underscoring the potential relevance of these brain changes to dyslexia. Our perspective on dyslexia is that some of the brain changes cause phonological processing abnormalities as well as auditory processing abnormalities; the latter, we speculate, resolve in a proportion of individuals during development, but contribute early on to the phonological disorder in dyslexia. Thus, we propose a tentative pathway between a genetic effect, developmental brain changes, and perceptual and cognitive deficits associated with dyslexia.


Assuntos
Comportamento , Dislexia/genética , Dislexia/fisiopatologia , Animais , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Dislexia/patologia , Humanos , Modelos Moleculares
16.
Epileptic Disord ; 11(3): 206-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19740719

RESUMO

Epilepsy-associated glioneuronal malformations (malformations of cortical development [MCD]) include focal cortical dysplasias (FCD) and highly differentiated glioneuronal tumors, most frequently gangliogliomas. The neuropathological findings are variable but suggest aberrant proliferation, migration, and differentiation of neural precursor cells as essential pathogenetic elements. Recent advances in animal models for MCDs allow new insights in the molecular pathogenesis of these epilepsy-associated lesions. Novel approaches, presented here, comprise RNA interference strategies to generate and study experimental models of subcortical band heterotopia and study functional aspects of aberrantly shaped and positioned neurons. Exciting analyses address impaired NMDA receptor expression in FCD animal models compared to human FCDs and excitatory imbalances in MCD animal models such as lissencephaly gene ablated mice as well as in utero irradiated rats. An improved understanding of relevant pathomechanisms will advance the development of targeted treatment strategies for epilepsy-associated malformations.


Assuntos
Córtex Cerebral/anormalidades , Córtex Cerebral/crescimento & desenvolvimento , Epilepsia/patologia , Animais , Córtex Cerebral/patologia , Epilepsia/congênito , Humanos , Camundongos , Neuroglia/patologia , Neurônios/patologia , Interferência de RNA , Ratos , Especificidade da Espécie
17.
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
18.
Neuron ; 41(2): 175-7, 2004 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-14741096

RESUMO

Doublecortin (DCX) is a microtubule-associated protein that interacts with and regulates the microtubule cytoskeleton and is required for neuronal migration in the cortex. Two papers in this issue of Neuron (Schaar et al. and Tanaka et al.) demonstrate a role for phosphorylation in the regulation of Doublecortin. Together with recent results showing that Doublecortin may play a role regulating the morphology of migrating neurons, these findings provide new insight into the mechanisms governing neuronal migration.


Assuntos
Proteínas Associadas aos Microtúbulos/fisiologia , Neuropeptídeos/fisiologia , Serina/fisiologia , Movimento Celular , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Citoesqueleto/fisiologia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Humanos , Microtúbulos/fisiologia , Neurônios/fisiologia
19.
J Neurosci ; 27(52): 14459-69, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18160654

RESUMO

Physiological processing of the beta-amyloid precursor protein (APP) generates amyloid beta-protein, which can assemble into oligomers that mediate synaptic failure in Alzheimer's disease. Two decades of research have led to human trials of compounds that chronically target this processing, and yet the normal function of APP in vivo remains unclear. We used the method of in utero electroporation of shRNA constructs into the developing cortex to acutely knock down APP in rodents. This approach revealed that neuronal precursor cells in embryonic cortex require APP to migrate correctly into the nascent cortical plate. cDNAs encoding human APP or its homologues, amyloid precursor-like protein 1 (APLP1) or APLP2, fully rescued the shRNA-mediated migration defect. Analysis of an array of mutations and deletions in APP revealed that both the extracellular and cytoplasmic domains of APP are required for efficient rescue. Whereas knock-down of APP inhibited cortical plate entry, overexpression of APP caused accelerated migration of cells past the cortical plate boundary, confirming that normal APP levels are required for correct neuronal migration. In addition, we found that Disabled-1 (Dab1), an adaptor protein with a well established role in cortical cell migration, acts downstream of APP for this function in cortical plate entry. We conclude that full-length APP functions as an important factor for proper migration of neuronal precursors into the cortical plate during the development of the mammalian brain.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/fisiologia , Interferência de RNA/fisiologia , Animais , Células COS , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Chlorocebus aethiops , Eletroporação/métodos , Embrião de Mamíferos , Feminino , Proteínas de Fluorescência Verde/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Gravidez , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Transfecção/métodos , Útero/fisiologia
20.
Trends Neurosci ; 29(7): 407-413, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16713637

RESUMO

The genetic basis is now known for several disorders of neuronal migration in the developing cerebral cortex. Identification of the cellular processes mediated by the implicated genes is revealing crucial stages of neuronal migration and has the potential to reveal common cellular causes of neuronal migration disorders. We hypothesize that a newly recognized morphological stage of neuronal migration, the multipolar stage, is vulnerable and is disrupted in several disorders of neocortical development. The multipolar stage occurs as bipolar progenitor cells become radially migrating neurons. Several studies using in utero electroporation and RNAi have revealed that transition out of the multipolar stage depends on the function of filamin A, LIS1 and DCX. Mutations in the genes encoding these proteins in humans cause distinct neuronal migration disorders, including periventricular nodular heterotopia, subcortical band heterotopia and lissencephaly. The multipolar stage therefore seems to be a critical point of migration control and a vulnerable target for disruption of neocortical development. This review is part of the INMED/TINS special issue "Nature and nurture in brain development and neurological disorders", based on presentations at the annual INMED/TINS symposium (http://inmednet.com/).


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/anormalidades , Córtex Cerebral/fisiologia , Malformações do Sistema Nervoso/patologia , Neurônios/fisiologia , Animais , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Malformações do Sistema Nervoso/etiologia , Malformações do Sistema Nervoso/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA