Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Neurosci ; 25(8): 1049-1058, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35915179

RESUMO

Mammalian neocortical neurons span one of the most diverse cell type spectra of any tissue. Cortical neurons are born during embryonic development, and their maturation extends into postnatal life. The regulatory strategies underlying progressive neuronal development and maturation remain unclear. Here we present an integrated single-cell epigenomic and transcriptional analysis of individual mouse and marmoset cortical neuron classes, spanning both early postmitotic stages of identity acquisition and later stages of neuronal plasticity and circuit integration. We found that, in both species, the regulatory strategies controlling early and late stages of pan-neuronal development diverge. Early postmitotic neurons use more widely shared and evolutionarily conserved molecular regulatory programs. In contrast, programs active during later neuronal maturation are more brain- and neuron-specific and more evolutionarily divergent. Our work uncovers a temporal shift in regulatory choices during neuronal diversification and maturation in both mice and marmosets, which likely reflects unique evolutionary constraints on distinct events of neuronal development in the neocortex.


Assuntos
Neocórtex , Animais , Callithrix , Mamíferos , Camundongos , Neurogênese/fisiologia , Plasticidade Neuronal , Neurônios/fisiologia
2.
Nature ; 598(7879): 182-187, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34616069

RESUMO

Diverse types of glutamatergic pyramidal neurons mediate the myriad processing streams and output channels of the cerebral cortex1,2, yet all derive from neural progenitors of the embryonic dorsal telencephalon3,4. Here we establish genetic strategies and tools for dissecting and fate-mapping subpopulations of pyramidal neurons on the basis of their developmental and molecular programs. We leverage key transcription factors and effector genes to systematically target temporal patterning programs in progenitors and differentiation programs in postmitotic neurons. We generated over a dozen temporally inducible mouse Cre and Flp knock-in driver lines to enable the combinatorial targeting of major progenitor types and projection classes. Combinatorial strategies confer viral access to subsets of pyramidal neurons defined by developmental origin, marker expression, anatomical location and projection targets. These strategies establish an experimental framework for understanding the hierarchical organization and developmental trajectory of subpopulations of pyramidal neurons that assemble cortical processing networks and output channels.


Assuntos
Córtex Cerebral/citologia , Regulação da Expressão Gênica/genética , Ácido Glutâmico/metabolismo , Células Piramidais/citologia , Células Piramidais/metabolismo , Animais , Linhagem da Célula/genética , Córtex Cerebral/metabolismo , Masculino , Camundongos , Células Piramidais/classificação , Fatores de Transcrição/metabolismo
3.
Science ; 370(6520)2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33243861

RESUMO

The number of disease risk genes and loci identified through human genetic studies far outstrips the capacity to systematically study their functions. We applied a scalable genetic screening approach, in vivo Perturb-Seq, to functionally evaluate 35 autism spectrum disorder/neurodevelopmental delay (ASD/ND) de novo loss-of-function risk genes. Using CRISPR-Cas9, we introduced frameshift mutations in these risk genes in pools, within the developing mouse brain in utero, followed by single-cell RNA-sequencing of perturbed cells in the postnatal brain. We identified cell type-specific and evolutionarily conserved gene modules from both neuronal and glial cell classes. Recurrent gene modules and cell types are affected across this cohort of perturbations, representing key cellular effects across sets of ASD/ND risk genes. In vivo Perturb-Seq allows us to investigate how diverse mutations affect cell types and states in the developing organism.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/patologia , Encéfalo/anormalidades , Neuroglia/patologia , Neurônios/patologia , Animais , Anquirinas/genética , Anquirinas/metabolismo , Sistemas CRISPR-Cas , Proteínas de Ligação a DNA/genética , Mutação da Fase de Leitura , Perfilação da Expressão Gênica , Loci Gênicos , Humanos , Camundongos , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/genética , Risco , Fatores de Transcrição/genética
4.
Nat Commun ; 10(1): 5192, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729356

RESUMO

The extent of neocortical gyrification is an important determinant of a species' cognitive abilities, yet the mechanisms regulating cortical gyrification are poorly understood. We uncover long-range regulation of this process originating at the telencephalic dorsal midline, where levels of secreted Bmps are maintained by factors in both the neuroepithelium and the overlying mesenchyme. In the mouse, the combined loss of transcription factors Lmx1a and Lmx1b, selectively expressed in the midline neuroepithelium and the mesenchyme respectively, causes dorsal midline Bmp signaling to drop at early neural tube stages. This alters the spatial and temporal Wnt signaling profile of the dorsal midline cortical hem, which in turn causes gyrification of the distal neocortex. Our study uncovers early mesenchymal-neuroepithelial interactions that have long-range effects on neocortical gyrification and shows that lissencephaly in mice is actively maintained via redundant genetic regulation of dorsal midline development and signaling.


Assuntos
Mesoderma/embriologia , Neocórtex/embriologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/metabolismo , Células Neuroepiteliais/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
5.
J Biosci ; 43(1): 75-83, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29485116

RESUMO

In the developing central nervous system, transcription factors play a crucial role in the regulation of cell fate. Previously we demonstrated that LHX2 is a critical regulator of the neuron-glia cell fate switch in the developing mouse hippocampus. Here, we test LHX2 target gene Pax6 for a role in this process. We report that Pax6 overexpression is able to suppress the enhanced astrogliogenesis arising due to loss of functional LHX2. Furthermore, we show that like Lhx2, Pax6 is also able to suppress induced astrogliogenesis caused by overexpression of progliogenic factor Nfia. This demonstrates that overexpression of Pax6 can substitute for Lhx2 in the regulation of the neuronal versus glial cell fate in the developing hippocampus, and therefore, supports a role for PAX6 as a mediator of LHX2 function in this process.


Assuntos
Astrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/metabolismo , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição NFI/genética , Neurônios/metabolismo , Fator de Transcrição PAX6/genética , Fatores de Transcrição/genética , Animais , Astrócitos/citologia , Diferenciação Celular , Eletroporação , Embrião de Mamíferos , Feminino , Hipocampo/citologia , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/citologia , Fator de Transcrição PAX6/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
6.
Development ; 145(1)2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29229772

RESUMO

During forebrain development, a telencephalic organizer called the cortical hem is crucial for inducing hippocampal fate in adjacent cortical neuroepithelium. How the hem is restricted to its medial position is therefore a fundamental patterning issue. Here, we demonstrate that Foxg1-Lhx2 interactions are crucial for the formation of the hem. Loss of either gene causes a region of the cortical neuroepithelium to transform into hem. We show that FOXG1 regulates Lhx2 expression in the cortical primordium. In the absence of Foxg1, the presence of Lhx2 is sufficient to suppress hem fate, and hippocampal markers appear selectively in Lhx2-expressing regions. FOXG1 also restricts the temporal window in which loss of Lhx2 results in a transformation of cortical primordium into hem. Therefore, Foxg1 and Lhx2 form a genetic hierarchy in the spatiotemporal regulation of cortical hem specification and positioning, and together ensure the normal development of this hippocampal organizer.


Assuntos
Fatores de Transcrição Forkhead/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/embriologia , Proteínas com Homeodomínio LIM/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Telencéfalo/embriologia , Fatores de Transcrição/biossíntese , Animais , Fatores de Transcrição Forkhead/genética , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética
7.
Neural Dev ; 12(1): 19, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29141678

RESUMO

Patterning of the telencephalic neuroepithelium is a tightly regulated process controlled by transcription factors and signalling molecules. The cortical primordium is flanked by two signalling centres, the hem medially, and the antihem laterally. The hem induces the formation of the hippocampus in adjacent neuroepithelium. Therefore, the position of the hem defines the position of the hippocampus in the brain. The antihem is positioned at the boundary between the dorsal and ventral telencephalon and proposed to provide patterning cues during development. LIM-homeodomain (LIM-HD) transcription factor LHX2 suppresses both hem and antihem fate in the cortical neuroepithelium. Upon loss of Lhx2, medial cortical neuroepithelium is transformed into hem, whereas lateral cortical neuroepithelium is transformed into antihem. Here, we show that transcription factor PAX6, known to regulate patterning of the lateral telencephalon, restricts this tissue from transforming into hem upon loss of Lhx2. When Lhx2 and Pax6 are both deleted, the cortical hem expands to occupy almost the complete extent of the cortical primordium, indicating that both factors act to suppress hem fate in the lateral telencephalon. Furthermore, the shift in the pallial-subpallial boundary and absence of the antihem, observed in the Pax6 mutant, are both restored in the Lhx2; Pax6 double mutant. Together, these results not only reveal a novel function for LHX2 in regulating dorsoventral patterning in the telencephalon, but also identify PAX6 as a fundamental regulator of where the hem can form, and therefore implicate this molecule as a determinant of hippocampal positioning.


Assuntos
Proteínas com Homeodomínio LIM/deficiência , Neurogênese/fisiologia , Fator de Transcrição PAX6/deficiência , Telencéfalo/embriologia , Fatores de Transcrição/deficiência , Animais , Camundongos , Camundongos Knockout
8.
J Neurosci ; 37(46): 11245-11254, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29025924

RESUMO

Regulation of the neuron-glia cell-fate switch is a critical step in the development of the CNS. Previously, we demonstrated that Lhx2 is a necessary and sufficient regulator of this process in the mouse hippocampal primordium, such that Lhx2 overexpression promotes neurogenesis and suppresses gliogenesis, whereas loss of Lhx2 has the opposite effect. We tested a series of transcription factors for their ability to mimic Lhx2 overexpression and suppress baseline gliogenesis, and also to compensate for loss of Lhx2 and suppress the resulting enhanced level of gliogenesis in the hippocampus. Here, we demonstrate a novel function of Dmrt5/Dmrta2 as a neurogenic factor in the developing hippocampus. We show that Dmrt5, as well as known neurogenic factors Neurog2 and Pax6, can each not only mimic Lhx2 overexpression, but also can compensate for loss of Lhx2 to different extents. We further uncover a reciprocal regulatory relationship between Dmrt5 and Lhx2, such that each can compensate for loss of the other. Dmrt5 and Lhx2 also have opposing regulatory control on Pax6 and Neurog2, indicating a complex bidirectionally regulated network that controls the neuron-glia cell-fate switch.SIGNIFICANCE STATEMENT We identify Dmrt5 as a novel regulator of the neuron-glia cell-fate switch in the developing hippocampus. We demonstrate Dmrt5 to be neurogenic, and reciprocally regulated by Lhx2: loss of either factor promotes gliogenesis; overexpression of either factor suppresses gliogenesis and promotes neurogenesis; each can substitute for loss of the other. Furthermore, each factor has opposing effects on established neurogenic genes Neurog2 and Pax6 Dmrt5 is known to suppress their expression, and we show that Lhx2 is required to maintain it. Our study reveals a complex regulatory network with bidirectional control of a fundamental feature of CNS development, the control of the production of neurons versus astroglia in the developing hippocampus.Finally, we confirm that Lhx2 binds a highly conserved putative enhancer of Dmrt5, suggesting an evolutionarily conserved regulatory relationship between these factors. Our findings uncover a complex network that involves Lhx2, Dmrt5, Neurog2, and Pax6, and that ensures the appropriate amount and timing of neurogenesis and gliogenesis in the developing hippocampus.


Assuntos
Hipocampo/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Neurogênese/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Hipocampo/citologia , Hipocampo/embriologia , Masculino , Camundongos , Camundongos Transgênicos , Gravidez
9.
J Neurosci ; 37(37): 9037-9053, 2017 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-28821643

RESUMO

Cell type-specific changes in neuronal excitability have been proposed to contribute to the selective degeneration of corticospinal neurons in amyotrophic lateral sclerosis (ALS) and to neocortical hyperexcitability, a prominent feature of both inherited and sporadic variants of the disease, but the mechanisms underlying selective loss of specific cell types in ALS are not known. We analyzed the physiological properties of distinct classes of cortical neurons in the motor cortex of hSOD1G93A mice of both sexes and found that they all exhibit increases in intrinsic excitability that depend on disease stage. Targeted recordings and in vivo calcium imaging further revealed that neurons adapt their functional properties to normalize cortical excitability as the disease progresses. Although different neuron classes all exhibited increases in intrinsic excitability, transcriptional profiling indicated that the molecular mechanisms underlying these changes are cell type specific. The increases in excitability in both excitatory and inhibitory cortical neurons show that selective dysfunction of neuronal cell types cannot account for the specific vulnerability of corticospinal motor neurons in ALS. Furthermore, the stage-dependent alterations in neuronal function highlight the ability of cortical circuits to adapt as disease progresses. These findings show that both disease stage and cell type must be considered when developing therapeutic strategies for treating ALS.SIGNIFICANCE STATEMENT It is not known why certain classes of neurons preferentially die in different neurodegenerative diseases. It has been proposed that the enhanced excitability of affected neurons is a major contributor to their selective loss. We show using a mouse model of amyotrophic lateral sclerosis (ALS), a disease in which corticospinal neurons exhibit selective vulnerability, that changes in excitability are not restricted to this neuronal class and that excitability does not increase monotonically with disease progression. Moreover, although all neuronal cell types tested exhibited abnormal functional properties, analysis of their gene expression demonstrated cell type-specific responses to the ALS-causing mutation. These findings suggest that therapies for ALS may need to be tailored for different cell types and stages of disease.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Excitabilidade Cortical , Neurônios Motores , Neocórtex/fisiopatologia , Rede Nervosa/fisiopatologia , Neurônios , Tratos Piramidais/fisiopatologia , Adaptação Fisiológica , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal
10.
Cell Rep ; 19(2): 335-350, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402856

RESUMO

Autism spectrum disorder (ASD) is a heterogeneous disease, but genetically defined models can provide an entry point to studying the molecular underpinnings of this disorder. We generated germline mutant mice with loss-of-function mutations in Chd8, a de novo mutation strongly associated with ASD, and demonstrate that these mice display hallmark ASD behaviors, macrocephaly, and craniofacial abnormalities similar to patient phenotypes. Chd8+/- mice display a broad, brain-region-specific dysregulation of major regulatory and cellular processes, most notably histone and chromatin modification, mRNA and protein processing, Wnt signaling, and cell-cycle regulation. We also find altered synaptic physiology in medium spiny neurons of the nucleus accumbens. Perturbation of Chd8 in adult mice recapitulates improved acquired motor learning behavior found in Chd8+/- animals, suggesting a role for CHD8 in adult striatal circuits. These results support a mechanism linking chromatin modification to striatal dysfunction and the molecular pathology of ASD.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Ligação a DNA/genética , Megalencefalia/genética , Animais , Transtorno do Espectro Autista/patologia , Cromatina/genética , Corpo Estriado/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Mutação em Linhagem Germinativa , Histonas/genética , Humanos , Megalencefalia/patologia , Camundongos , Via de Sinalização Wnt/genética
11.
J Neurosci ; 37(1): 194-203, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28053041

RESUMO

In the developing cerebral cortex, sequential transcriptional programs take neuroepithelial cells from proliferating progenitors to differentiated neurons with unique molecular identities. The regulatory changes that occur in the chromatin of the progenitors are not well understood. During deep layer neurogenesis, we show that transcription factor LHX2 binds to distal regulatory elements of Fezf2 and Sox11, critical determinants of neuron subtype identity in the mouse neocortex. We demonstrate that LHX2 binds to the nucleosome remodeling and histone deacetylase histone remodeling complex subunits LSD1, HDAC2, and RBBP4, which are proximal regulators of the epigenetic state of chromatin. When LHX2 is absent, active histone marks at the Fezf2 and Sox11 loci are increased. Loss of LHX2 produces an increase, and overexpression of LHX2 causes a decrease, in layer 5 Fezf2 and CTIP2-expressing neurons. Our results provide mechanistic insight into how LHX2 acts as a necessary and sufficient regulator of genes that control cortical neuronal subtype identity. SIGNIFICANCE STATEMENT: The functional complexity of the cerebral cortex arises from an array of distinct neuronal subtypes with unique connectivity patterns that are produced from common progenitors. This study reveals that transcription factor LHX2 regulates the numbers of specific cortical output neuron subtypes by controlling the genes that are required to produce them. Loss or increase in LHX2 during neurogenesis is sufficient to increase or decrease, respectively, a particular subcerebrally projecting population. Mechanistically, LHX2 interacts with chromatin modifying protein complexes to edit the chromatin landscape of its targets Fezf2 and Sox11, which regulates their expression and consequently the identities of the neurons produced. Thus, LHX2 is a key component of the control network for producing neurons that will participate in cortical circuitry.


Assuntos
Córtex Cerebral/citologia , Proteínas de Ligação a DNA/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Fatores de Transcrição SOXC/metabolismo , Fatores de Transcrição/metabolismo , Animais , Córtex Cerebral/diagnóstico por imagem , Cromatina/genética , Epigênese Genética , Feminino , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Masculino , Camundongos , Nucleossomos/metabolismo , Gravidez
12.
Trends Neurosci ; 38(2): 117-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25529141

RESUMO

The mammalian cerebral cortex is responsible for the highest levels of associative, cognitive and motor functions. In the central nervous system (CNS) the cortex stands as a prime example of extreme neuronal diversity, broadly classified into excitatory projection neurons (PNs) and inhibitory interneurons (INs). We review here recent progress made in understanding the strategies and mechanisms that shape PN diversity during embryogenesis, and discuss how PN classes may be maintained, postnatally, for the life of the organism. In addition, we consider the intriguing possibility that PNs may be amenable to directed reprogramming of their class-specific features to allow enhanced cortical plasticity in the adult.


Assuntos
Córtex Cerebral/fisiologia , Rede Nervosa/fisiologia , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Animais , Humanos , Interneurônios/fisiologia
13.
Proc Natl Acad Sci U S A ; 110(50): E4913-21, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24262147

RESUMO

LIM homeodomain transcription factors are critical regulators of early development in multiple systems but have yet to be examined for a role in circuit formation. The LIM homeobox gene Lhx2 is expressed in cortical progenitors during development and also in the superficial layers of the neocortex in maturity. However, analysis of Lhx2 function at later stages of cortical development has been hampered by severe phenotypes associated with early loss of function. We identified a particular Cre-recombinase line that acts in the cortical primordium after its specification is complete, permitting an analysis of Lhx2 function in neocortical lamination, regionalization, and circuit formation by selective elimination of Lhx2 in the dorsal telencephalon. We report a profound disruption of cortical neuroanatomical and molecular features upon loss of Lhx2 in the cortex from embryonic day 11.5. A unique feature of cortical circuitry, the somatosensory barrels, is undetectable, and molecular patterning of cortical regions appears disrupted. Surprisingly, thalamocortical afferents innervate the mutant cortex with apparently normal regional specificity. Electrophysiological recordings reveal a loss of responses evoked by stimulation of individual whiskers, but responses to simultaneous stimulation of multiple whiskers were present, suggesting that thalamic afferents are unable to organize the neurocircuitry for barrel formation because of a cortex-specific requirement of Lhx2. We report that Lhx2 is required for the expression of transcription factor paired box gene 6, axon guidance molecule Ephrin A5, and the receptor NMDA receptor 1. These genes may mediate Lhx2 function in the formation of specialized neurocircuitry necessary for neocortical function.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Córtex Somatossensorial/embriologia , Fatores de Transcrição/metabolismo , Animais , Imunoprecipitação da Cromatina , Efrina-A5/metabolismo , Potenciais Evocados/fisiologia , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Integrases , Proteínas com Homeodomínio LIM/deficiência , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Vias Neurais/embriologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Repressoras/metabolismo , Córtex Somatossensorial/metabolismo , Fatores de Transcrição/deficiência
14.
F1000Res ; 2: 205, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-25110573

RESUMO

The LIM-homeodomain (LIM-HD) family of transcription factors is well known for its functions during several developmental processes including cell fate specification, cell migration and axon guidance, and its members play fundamental roles in hippocampal development. The hippocampus is a structure that displays striking activity dependent plasticity.  We examined whether LIM-HD genes and their co-factors are regulated during kainic acid induced seizure in the adult rat hippocampus as well as in early postnatal rats, when the hippocampal circuitry is not fully developed.  We report a distinct and field-specific regulation of LIM-HD genes Lhx1, Lhx2, and Lhx9, LIM-only gene Lmo4, and cofactor Clim1a in the adult hippocampus after seizure induction. In contrast none of these genes displayed altered levels upon induction of seizure in postnatal animals.  Our results provide evidence of temporal and spatial seizure mediated regulation of LIM-HD family members and suggest that LIM-HD gene function may be involved in activity dependent plasticity in the adult hippocampus.

15.
Proc Natl Acad Sci U S A ; 108(27): E265-74, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690374

RESUMO

The sequential production of neurons and astrocytes from neuroepithelial precursors is a fundamental feature of central nervous system development. We report that LIM-homeodomain (LIM-HD) transcription factor Lhx2 regulates this transition in the developing hippocampus. Disrupting Lhx2 function in the embryonic hippocampus by in utero electroporation and in organotypic slice culture caused the premature production of astrocytes at stages when neurons are normally generated. Lhx2 function is therefore necessary to suppress astrogliogenesis during the neurogenic period. Furthermore, Lhx2 overexpression was sufficient to suppress astrogliogenesis and prolong the neurogenic period. We provide evidence that Lhx2 overexpression can counteract the instructive astrogliogenic effect of Notch activation. Lhx2 overexpression was also able to override and suppress the activation of the GFAP promoter by Nfia, a Notch-regulated transcription factor that is required for gliogenesis. Thus, Lhx2 appears to act as a "brake" on Notch/Nfia-mediated astrogliogenesis. This critical role for Lhx2 is spatially restricted to the hippocampus, because loss of Lhx2 function in the neocortex did not result in premature astrogliogenesis at the expense of neurogenesis. Our results therefore place Lhx2 as a central regulator of the neuron-glia cell fate decision in the hippocampus and reveal a striking regional specificity of this fundamental function within the dorsal telencephalon.


Assuntos
Hipocampo/embriologia , Proteínas de Homeodomínio/fisiologia , Neurogênese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição NFI/fisiologia , Neocórtex/citologia , Neocórtex/embriologia , Neocórtex/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Fenótipo , Gravidez , Receptores Notch/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
16.
Science ; 319(5861): 304-9, 2008 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-18202285

RESUMO

The earliest step in creating the cerebral cortex is the specification of neuroepithelium to a cortical fate. Using mouse genetic mosaics and timed inactivations, we demonstrated that Lhx2 acts as a classic selector gene and essential intrinsic determinant of cortical identity. Lhx2 selector activity is restricted to an early critical period when stem cells comprise the cortical neuroepithelium, where it acts cell-autonomously to specify cortical identity and suppress alternative fates in a spatially dependent manner. Laterally, Lhx2 null cells adopt antihem identity, whereas medially they become cortical hem cells, which can induce and organize ectopic hippocampal fields. In addition to providing functional evidence for Lhx2 selector activity, these findings show that the cortical hem is a hippocampal organizer.


Assuntos
Córtex Cerebral/embriologia , Hipocampo/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Organizadores Embrionários/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Agregação Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Quimera , Giro Denteado/citologia , Giro Denteado/embriologia , Giro Denteado/metabolismo , Indução Embrionária , Células-Tronco Embrionárias/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/citologia , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Knockout , Mutação , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Organizadores Embrionários/embriologia , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Células Piramidais/citologia , Células Piramidais/embriologia , Recombinação Genética , Telencéfalo/citologia , Telencéfalo/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...