Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 31(9): 1868-80, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23712654

RESUMO

A cardinal property of neural stem cells (NSCs) is their ability to adopt multiple fates upon differentiation. The epigenome is widely seen as a read-out of cellular potential and a manifestation of this can be seen in embryonic stem cells (ESCs), where promoters of many lineage-specific regulators are marked by a bivalent epigenetic signature comprising trimethylation of both lysine 4 and lysine 27 of histone H3 (H3K4me3 and H3K27me3, respectively). Bivalency has subsequently emerged as a powerful epigenetic indicator of stem cell potential. Here, we have interrogated the epigenome during differentiation of ESC-derived NSCs to immature GABAergic interneurons. We show that developmental transitions are accompanied by loss of bivalency at many promoters in line with their increasing developmental restriction from pluripotent ESC through multipotent NSC to committed GABAergic interneuron. At the NSC stage, the promoters of genes encoding many transcriptional regulators required for differentiation of multiple neuronal subtypes and neural crest appear to be bivalent, consistent with the broad developmental potential of NSCs. Upon differentiation to GABAergic neurons, all non-GABAergic promoters resolve to H3K27me3 monovalency, whereas GABAergic promoters resolve to H3K4me3 monovalency or retain bivalency. Importantly, many of these epigenetic changes occur before any corresponding changes in gene expression. Intriguingly, another group of gene promoters gain bivalency as NSCs differentiate toward neurons, the majority of which are associated with functions connected with maturation and establishment and maintenance of connectivity. These data show that bivalency provides a dynamic epigenetic signature of developmental potential in both NSCs and in early neurons.


Assuntos
Epigênese Genética , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Linhagem da Célula/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Camundongos , Dados de Sequência Molecular , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética
2.
Stem Cells ; 30(3): 425-34, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22162260

RESUMO

Neural differentiation of embryonic stem cells (ESCs) requires coordinated repression of the pluripotency regulatory program and reciprocal activation of the neurogenic regulatory program. Upon neural induction, ESCs rapidly repress expression of pluripotency genes followed by staged activation of neural progenitor and differentiated neuronal and glial genes. The transcriptional factors that underlie maintenance of pluripotency are partially characterized whereas those underlying neural induction are much less explored, and the factors that coordinate these two developmental programs are completely unknown. One transcription factor, REST (repressor element 1 silencing transcription factor), has been linked with terminal differentiation of neural progenitors and more recently, and controversially, with control of pluripotency. Here, we show that in the absence of REST, coordination of pluripotency and neural induction is lost and there is a resultant delay in repression of pluripotency genes and a precocious activation of both neural progenitor and differentiated neuronal and glial genes. Furthermore, we show that REST is not required for production of radial glia-like progenitors but is required for their subsequent maintenance and differentiation into neurons, oligodendrocytes, and astrocytes. We propose that REST acts as a regulatory hub that coordinates timely repression of pluripotency with neural induction and neural differentiation.


Assuntos
Células-Tronco Embrionárias/fisiologia , Neurogênese , Células-Tronco Pluripotentes/fisiologia , Proteínas Repressoras/fisiologia , Animais , Benzamidas/farmacologia , Diferenciação Celular , Dioxóis/farmacologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Células-Tronco Pluripotentes/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
3.
Nature ; 460(7255): 632-6, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19641596

RESUMO

Mutations in the presenilin genes are the main cause of familial Alzheimer's disease. Loss of presenilin activity and/or accumulation of amyloid-beta peptides have been proposed to mediate the pathogenesis of Alzheimer's disease by impairing synaptic function. However, the precise site and nature of the synaptic dysfunction remain unknown. Here we use a genetic approach to inactivate presenilins conditionally in either presynaptic (CA3) or postsynaptic (CA1) neurons of the hippocampal Schaeffer-collateral pathway. We show that long-term potentiation induced by theta-burst stimulation is decreased after presynaptic but not postsynaptic deletion of presenilins. Moreover, we found that presynaptic but not postsynaptic inactivation of presenilins alters short-term plasticity and synaptic facilitation. The probability of evoked glutamate release, measured with the open-channel NMDA (N-methyl-D-aspartate) receptor antagonist MK-801, is reduced by presynaptic inactivation of presenilins. Notably, depletion of endoplasmic reticulum Ca(2+) stores by thapsigargin, or blockade of Ca(2+) release from these stores by ryanodine receptor inhibitors, mimics and occludes the effects of presynaptic presenilin inactivation. Collectively, these results indicate a selective role for presenilins in the activity-dependent regulation of neurotransmitter release and long-term potentiation induction by modulation of intracellular Ca(2+) release in presynaptic terminals, and further suggest that presynaptic dysfunction might be an early pathogenic event leading to dementia and neurodegeneration in Alzheimer's disease.


Assuntos
Regulação da Expressão Gênica , Neurônios/metabolismo , Neurotransmissores/metabolismo , Presenilinas/genética , Presenilinas/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Ácido Glutâmico/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Terminações Pré-Sinápticas/metabolismo
4.
J Biol Chem ; 284(20): 13705-13713, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19289467

RESUMO

Presenilins are essential for synaptic function, memory formation, and neuronal survival. Previously, we reported that expression of cAMP response element-binding protein (CREB) target genes is reduced in the cerebral cortex of presenilin (PS) conditional double knock-out (cDKO) mice. To determine whether the reduced expression of the CREB target genes in these mutant mice is due to loss of presenilin directly or secondary to the impaired neuronal activity, we established a sensitive luciferase reporter system to assess direct transcriptional regulation in cultured cells. We first used immortalized PS-deficient mouse embryonic fibroblasts (MEFs), and found that both CREB-mediated transcription and Notch-mediated HES1 transcription are decreased. However, the ubiquitin-C promoter-mediated transcription is also reduced, and among these three reporters, transfection of exogenous PS1 can rescue only the Notch-mediated HES1 transcription. Further Northern analysis revealed transcriptional alterations of Creb, ubiquitin-C, and other housekeeping genes in PS-deficient MEFs, indicating transcriptional dysregulation in these cells. We then used the Cre/loxP system to develop a postnatal PS-deficient cortical neuronal culture. Surprisingly, in these PS-null neurons, CREB-mediated transcription is not significantly decreased, and levels of total and phosphorylated CREB proteins are unchanged as well. Notch-mediated HES1 transcription is markedly reduced, and this reduction can be rescued by exogenous PS1. Together, our findings suggest that CREB-mediated transcription is regulated indirectly by PS in the adult cerebral cortex, and that attenuation of CREB target gene expression in PS cDKO mice is likely due to reduced neuronal activity in these mutant brains.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Presenilinas/metabolismo , Regiões Promotoras Genéticas/fisiologia , Transcrição Gênica/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Presenilinas/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição HES-1 , Ubiquitina C/genética , Ubiquitina C/metabolismo
5.
Dev Neurosci ; 28(1-2): 102-17, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16508308

RESUMO

The Notch signaling pathway plays a variety of roles in cell fate decisions during development. Previous studies have shown that reduced Notch signaling results in premature differentiation of neural progenitor cells, while increased Notch activities promote apoptotic death of neural progenitor cells in the developing brain. Whether Notch signaling is involved in the specification of neuronal subtypes is unclear. Here we examine the role of Notch1 in the development of neuronal subtypes in the spinal cord using conditional knockout (cKO) mice lacking Notch1 specifically in neural progenitor cells. Notch1 inactivation results in accelerated neuronal differentiation in the ventral spinal cord and gradual disappearance of the ventral central canal. These changes are accompanied by reduced expression of Hes1 and Hes5 and increased expression of Mash1 and Neurogenin 1 and 2. Using markers (Nkx2.2, Nkx6.1, Olig2, Pax6 and Dbx1) for one or multiple progenitor cell types, we found reductions of all subtypes of progenitor cells in the ventral spinal cord of Notch1 cKO mice. Similarly, using markers (Islet1/2, Lim3, Sim1, Chox10, En1 and Evx1/2) specific for motor neurons and distinct classes of interneurons, we found increases in the number of V0-2 interneurons in the ventral spinal cord of Notch1 cKO mice. Specifically, the number of Lim3+/Chox10+ V2 interneurons is markedly increased while the number of Lim3+/Islet+motor neurons is decreased in the Notch1 cKO spinal cord, suggesting that V2 interneurons are generated at the expense of motor neurons in the absence of Notch1. These results provide support for a role of Notch1 in neuronal subtype specification in the ventral spinal cord.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Interneurônios/metabolismo , Neurônios Motores/metabolismo , Receptor Notch1/genética , Medula Espinal/embriologia , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Regulação para Baixo/fisiologia , Proteína Homeobox Nkx-2.2 , Interneurônios/citologia , Camundongos , Camundongos Knockout , Neurônios Motores/citologia , Proteínas do Tecido Nervoso/metabolismo , Medula Espinal/citologia , Células-Tronco/citologia
6.
Trends Pharmacol Sci ; 27(1): 33-40, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16337694

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disorder and is characterized by memory loss and other cognitive disabilities. Mutations in the presenilin genes are the major cause of familial AD. Analysis of conditional knockout mice has shown that inactivation of presenilins results in progressive memory impairment and age-dependent neurodegeneration, suggesting that reduced presenilin activity might represent an important pathogenic mechanism. Presenilins positively regulate the transcription of cAMP response element (CRE)-containing genes, some of which are known to be important for memory formation and neuronal survival. Phosphodiesterase 4 and histone deacetylase inhibitors, which can enhance CRE-dependent gene expression, have been shown to ameliorate memory deficits and neurodegeneration in animal models. Thus, modulation of CRE-dependent transcription might be beneficial for the treatment of dementia in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , AMP Cíclico/farmacologia , Proteínas de Membrana/fisiologia , Elementos de Resposta/fisiologia , Transcrição Gênica , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/fisiologia , Animais , Sobrevivência Celular , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Humanos , Proteínas de Membrana/genética , Memória , Mutação , Neurônios/fisiologia , Presenilina-1 , Presenilina-2 , Transdução de Sinais
7.
Mol Cell Biol ; 25(16): 7278-88, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055736

RESUMO

Neurexophilin 3 (Nxph3) is a specific ligand of synaptic alpha-neurexins that are essential for efficient neurotransmitter release. Previous biochemical work demonstrated that Nxph3 interacts with an extracellular domain of alpha-neurexins in a tight complex; however, no information is available on the localization or functional role of Nxph3 in the brain. Here, we generated lacZ reporter gene knock-in mice to investigate the distribution of Nxph3 at the single-cell level and Nxph3 knockout mice to examine its functional importance. Nxph3 expression was restricted mostly to subplate-derived neurons in cortical layer 6b, granule cells in the vestibulocerebellum, and Cajal-Retzius cells during development. Colabeling experiments demonstrated that neurons expressing Nxph3 do not belong to a uniform cell type. Morphological analyses and systematic behavioral testing of knockout mice revealed no anatomical defects but uncovered remarkable functional abnormalities in sensory information processing and motor coordination, evident by increased startle response, reduced prepulse inhibition, and poor rotarod performance. Since Nxph3-deficient mice behaved normally while performing a number of other tasks, our data suggest an important role for Nxph3 as a locally and temporally regulated neuropeptide-like molecule, presumably acting in a complex with alpha-neurexins in select neuronal circuits.


Assuntos
Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Glicoproteínas/biossíntese , Neuropeptídeos/metabolismo , Alelos , Análise de Variância , Animais , Comportamento Animal , Encéfalo/metabolismo , Células COS , Feminino , Genes Reporter , Glicoproteínas/metabolismo , Óperon Lac , Luz , Masculino , Aprendizagem em Labirinto , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Genéticos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neuropeptídeos/biossíntese , Fenótipo , Ratos , Receptores Pré-Sinápticos/metabolismo , Transmissão Sináptica , Fatores de Tempo
8.
Neurodegener Dis ; 2(1): 6-15, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16908998

RESUMO

Mutations in presenilins (PS) 1 and 2 are the major cause of familial Alzheimer's disease. Conditional inactivation of PS1 in the mouse postnatal forebrain leads to mild deficits in spatial learning and memory, whereas inactivation of both PS1 and PS2 results in severe memory and synaptic plasticity impairments, followed by progressive and substantial neurodegeneration. Here we investigate the effect of a familial Alzheimer's disease-linked PS1 missense mutation using knock-in (KI) mice, in which the wild-type PS1 allele is replaced with the M146V mutant allele. In the Morris water maze task, PS1 KI mice at 3 months of age exhibit reduced quadrant occupancy and platform crossing in the probe trial after 6 days of training, though their performance was normal in the probe trial after 12 days of training. By the age of 9 months, even after 12 days of training, PS1 homozygous KI mice still exhibit reduced platform crossing in the post-training probe trial. ELISA analysis revealed a selective increase in cortical levels of beta-amyloid 42 in PS1 KI mice, whereas production of beta-amyloid 40 was normal. Histological and quantitative real-time RT-PCR analyses showed normal gross hippocampal morphology and unaltered expression of three genes involved in inflammatory responses in PS1 KI mice. These results show hippocampal spatial memory impairments caused by the PS1 M146V mutation and age-related deterioration of the memory impairment, suggesting that PS1 KI mice are a valuable model system for the study of memory loss in AD.


Assuntos
Doença de Alzheimer/metabolismo , Predisposição Genética para Doença/genética , Hipocampo/metabolismo , Proteínas de Membrana/genética , Transtornos da Memória/metabolismo , Mutação/genética , Envelhecimento/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Encefalite/genética , Encefalite/metabolismo , Encefalite/fisiopatologia , Endopeptidases/química , Endopeptidases/genética , Endopeptidases/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1
9.
J Biol Chem ; 279(45): 46907-14, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15345711

RESUMO

Mutations in presenilins (PS) 1 and 2 are the major cause of familial Alzheimer's disease. Conditional double knock-out mice lacking both presenilins in the postnatal forebrain (PS cDKO mice) exhibit memory and synaptic plasticity impairments followed by progressive neurodegeneration in the cerebral cortex. Here we further investigate the molecular events that may underlie the observed phenotypes and identify additional neuropathological markers in the PS cDKO brain. Enzyme-linked immunosorbent assay analysis showed reduced levels of the toxic beta-amyloid (Abeta) peptides in the cerebral cortex of PS cDKO mice. Interestingly, the reduction in Abeta40 and Abeta42 peptides is similar in PS1 conditional knock-out and PS cDKO mice. We further examined the gene expression profile by oligonucleotide microarrays in the PS cDKO cerebral cortex and found that a high number of genes are differentially expressed, most notably a group of up-regulated inflammatory genes. Quantitative real-time reverse transcription PCR and Western analyses confirmed the elevated levels of glial fibrillary acidic protein, complement component C1q, and cathepsin S, up-regulation of which has been associated with inflammatory responses in various neurodegenerative processes. Immunohistochemical analysis revealed that the increase in complement component C1q is confined to the hippocampal formation, whereas glial fibrillary acidic protein and cathepsin S are up-regulated throughout the entire neocortex and hippocampus. In addition, strong microglial activation occurs in the hippocampus and the deeper cortical layers of PS cDKO mice. These results indicate that the memory impairment and neurodegeneration in PS cDKO mice are not caused by Abeta accumulation and that loss of PS function leads to differential up-regulation of inflammatory markers in the cerebral cortex.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Inflamação/genética , Proteínas de Membrana/genética , Animais , Western Blotting , Encéfalo/metabolismo , Catepsinas/metabolismo , Córtex Cerebral/metabolismo , Complemento C1q/metabolismo , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Imuno-Histoquímica , Antígenos Comuns de Leucócito/biossíntese , Camundongos , Camundongos Knockout , Doenças Neurodegenerativas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/química , Fenótipo , Presenilina-1 , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
10.
Neuron ; 42(1): 23-36, 2004 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15066262

RESUMO

Mutations in presenilins are the major cause of familial Alzheimer's disease, but the pathogenic mechanism by which presenilin mutations cause memory loss and neurodegeneration remains unclear. Here we demonstrate that conditional double knockout mice lacking both presenilins in the postnatal forebrain exhibit impairments in hippocampal memory and synaptic plasticity. These deficits are associated with specific reductions in NMDA receptor-mediated responses and synaptic levels of NMDA receptors and alphaCaMKII. Furthermore, loss of presenilins causes reduced expression of CBP and CREB/CBP target genes, such as c-fos and BDNF. With increasing age, mutant mice develop striking neurodegeneration of the cerebral cortex and worsening impairments of memory and synaptic function. Neurodegeneration is accompanied by increased levels of the Cdk5 activator p25 and hyperphosphorylated tau. These results define essential roles and molecular targets of presenilins in synaptic plasticity, learning and memory, and neuronal survival in the adult cerebral cortex.


Assuntos
Proteínas de Ligação a DNA , Proteínas de Membrana/fisiologia , Transtornos da Memória/metabolismo , Doenças Neurodegenerativas/metabolismo , Plasticidade Neuronal/fisiologia , Fator 1 Ativador da Transcrição , Fatores Etários , Animais , Anticorpos Monoclonais/metabolismo , Aprendizagem por Associação/fisiologia , Comportamento Animal , Western Blotting/métodos , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/farmacologia , Medo/fisiologia , Regulação da Expressão Gênica , Produtos do Gene gag/metabolismo , Imuno-Histoquímica/métodos , Técnicas In Vitro , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Potenciais da Membrana/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , N-Metilaspartato/farmacologia , Técnicas de Patch-Clamp , Testes de Precipitina/métodos , Presenilina-1 , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/biossíntese , Tempo de Reação/genética , Tempo de Reação/fisiologia , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Virais/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana
11.
Neuromolecular Med ; 6(1): 13-30, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15781974

RESUMO

Studies using genetic manipulations have proven invaluable in the research of neurological disorders. In the forefront of these approaches is the knockout technology that engineers a targeted gene mutation in mice resulting in inactivation of gene expression. In many cases, important roles of a particular gene in embryonic development have precluded the in vivo study of its function in the adult brain, which is usually the most relevant experimental context for the study of neurological disorders. The conditional knockout technology has provided a tool to overcome this restriction and has been used successfully to generate viable mouse models with gene inactivation patterns in certain regions or cell types of the postnatal brain. This review first describes the methodology of gene targeting in mice, detailing the aspects of designing a targeting vector, introducing it into embryonic stem cells in culture and screening for correct recombination events, and generating chimeric and null mutant mice from the positive clones. It then discusses the special issues and considerations for the generation of conditional knockout mice, including a section about approaches for inducible gene inactivation in the brain and some of their applications. An overview of gene-targeted mouse models that have been used in the study of several neurological disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, seizure disorders, and schizophrenia, is also presented. The importance of the results obtained by these models for the understanding of the pathogenic mechanism underlying the disorders is discussed.


Assuntos
Marcação de Genes/métodos , Doenças do Sistema Nervoso/genética , Animais , Modelos Animais de Doenças , Deleção de Genes , Humanos , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA