Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neural Dev ; 18(1): 5, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37684687

RESUMO

BACKGROUND: E-proteins encoded by Tcf3, Tcf4, and Tcf12 are class I basic helix-loop-helix (bHLH) transcription factors (TFs) that are thought to be widely expressed during development. However, their function in the developing brain, specifically in the telencephalon remains an active area of research. Our study examines for the first time if combined loss of two E-proteins (Tcf3 and Tcf12) influence distinct cell fates and oligodendrocyte development in the mouse telencephalon. METHODS: We generated Tcf3/12 double conditional knockouts (dcKOs) using Olig2Cre/+ or Olig1Cre/+ to overcome compensatory mechanisms between E-proteins and to understand the specific requirement for Tcf3 and Tcf12 in the ventral telencephalon and during oligodendrogenesis. We utilized a combination of in situ hybridization, immunohistochemistry, and immunofluorescence to address development of the telencephalon and oligodendrogenesis at embryonic and postnatal stages in Tcf3/12 dcKOs. RESULTS: We show that the E-proteins Tcf3 and Tcf12 are expressed in progenitors of the embryonic telencephalon and throughout the oligodendrocyte lineage in the postnatal brain. Tcf3/12 dcKOs showed transient defects in progenitor cells with an enlarged medial ganglionic eminence (MGE) region which correlated with reduced generation of embryonic oligodendrocyte progenitor cells (OPCs) and increased expression of MGE interneuron genes. Postnatal Tcf3/12 dcKOs showed a recovery of OPCs but displayed a sustained reduction in mature oligodendrocytes (OLs). Interestingly, Tcf4 remained expressed in the dcKOs suggesting that it cannot compensate for the loss of Tcf3 and Tcf12. Generation of Tcf3/12 dcKOs with Olig1Cre/+ avoided the MGE morphology defect caused by Olig2Cre/+ but dcKOs still exhibited reduced embryonic OPCs and subsequent reduction in postnatal OLs. CONCLUSION: Our data reveal that Tcf3 and Tcf12 play a role in controlling OPC versus cortical interneuron cell fate decisions in MGE progenitors in addition to playing roles in the generation of embryonic OPCs and differentiation of postnatal OLs in the oligodendrocyte lineage.


Assuntos
Encéfalo , Fatores de Transcrição , Animais , Camundongos , Fatores de Transcrição/genética , Diferenciação Celular , Células-Tronco Embrionárias , Histeria , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética
2.
J Neurosci ; 42(16): 3344-3364, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35273083

RESUMO

The projection neurons of the striatum, the principal nucleus of the basal ganglia, belong to one of the following two major pathways: the striatopallidal (indirect) pathway or the striatonigral (direct) pathway. Striatonigral axons project long distances and encounter ascending tracts (thalamocortical) while coursing alongside descending tracts (corticofugal) as they extend through the internal capsule and cerebral peduncle. These observations suggest that striatal circuitry may help to guide their trajectories. To investigate the developmental contributions of striatonigral axons to internal capsule formation, we have made use of Sox8-EGFP (striatal direct pathway) and Fezf2-TdTomato (corticofugal pathway) BAC transgenic reporter mice in combination with immunohistochemical markers to trace these axonal pathways throughout development. We show that striatonigral axons pioneer the internal capsule and cerebral peduncle and are temporally and spatially well positioned to provide guidance for corticofugal and thalamocortical axons. Using Isl1 conditional knock-out (cKO) mice, which exhibit disrupted striatonigral axon outgrowth, we observe both corticofugal and thalamocortical axon defects with either ventral forebrain- or telencephalon-specific Isl1 inactivation, despite Isl1 not being expressed in either cortical or thalamic projection neurons. Striatonigral axon defects can thus disrupt internal capsule formation. Our genome-wide transcriptomic analysis in Isl1 cKOs reveals changes in gene expression relevant to cell adhesion, growth cone dynamics, and extracellular matrix composition, suggesting potential mechanisms by which the striatonigral pathway exerts this guidance role. Together, our data support a novel pioneering role for the striatal direct pathway in the correct assembly of the ascending and descending axon tracts within the internal capsule and cerebral peduncle.SIGNIFICANCE STATEMENT The basal ganglia are a group of subcortical nuclei with established roles in the coordination of voluntary motor programs, aspects of cognition, and the selection of appropriate social behaviors. Hence, disruptions in basal ganglia connectivity have been implicated in the motor, cognitive, and social dysfunction characterizing common neurodevelopmental disorders such as attention-deficit/hyperactivity disorder, autism spectrum disorder, obsessive-compulsive disorder, and tic disorder. Here, we identified a novel role for the striatonigral (direct) pathway in pioneering the internal capsule and cerebral peduncle, and in guiding axons extending to and from the cortex. Our findings suggest that the abnormal development of basal ganglia circuits can drive secondary internal capsule defects and thereby may contribute to the pathology of these disorders.


Assuntos
Transtorno do Espectro Autista , Pedúnculo Cerebral , Animais , Transtorno do Espectro Autista/metabolismo , Axônios/fisiologia , Córtex Cerebral/metabolismo , Cápsula Interna , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Vias Neurais/fisiologia , Tálamo
3.
Front Cell Dev Biol ; 9: 673995, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34222248

RESUMO

The leucine zipper-like transcriptional regulator 1 (Lztr1) is a BTB-Kelch domain protein involved in RAS/MAPK pathway regulation. Mutations in LZTR1 are associated with cancers and Noonan syndrome, the most common RASopathy. The expression and function of Lztr1 in the developing brain remains poorly understood. Here we show that Lztr1 is expressed in distinct regions of the telencephalon, the most anterior region of the forebrain. Lztr1 expression was robust in the cortex, amygdala, hippocampus, and oligodendrocytes in the white matter. To gain insight into the impact of Lztr1 deficiency, we generated a conditional knockout (cKO) restricted to the telencephalon using Foxg1 IREScre/+. Lztr1 cKOs are viable to postnatal stages and show reduced Lztr1 expression in the telencephalon. Interestingly, Lztr1 cKOs exhibit an increase in MAPK pathway activation in white matter regions and subsequently show an altered expression of stage-specific markers in the oligodendrocyte lineage with increased oligodendrocyte progenitor cells (OPCs) and decreased markers of oligodendrocyte differentiation. Moreover, Lztr1 cKOs also exhibit an increased expression of the astrocyte marker GFAP. These results highlight the generation of a new mouse model to study Lztr1 deficiency in the brain and reveal a novel role for Lztr1 in normal oligodendrocyte and astrocyte development in the telencephalon.

4.
Dev Biol ; 476: 137-147, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33775695

RESUMO

The MAPK pathway is a major growth signal that has been implicated during the development of progenitors, neurons, and glia in the embryonic brain. Here, we show that the MAPK pathway plays an important role in the generation of distinct cell types from progenitors in the ventral telencephalon. Our data reveal that phospho-p44/42 (called p-ERK1/2) and the ETS transcription factor Etv5, both downstream effectors in the MAPK pathway, show a regional bias in expression during ventral telencephalic development, with enriched expression in the dorsal region of the LGE and ventral region of the MGE at E13.5 and E15.5. Interestingly, expression of both factors becomes more uniform in ventricular zone (VZ) progenitors by E18.5. To gain insight into the role of MAPK activity during progenitor cell development, we used a cre inducible constitutively active MEK1 allele (RosaMEK1DD/+) in combination with a ventral telencephalon enriched cre (Gsx2e-cre) or a dorsal telencephalon enriched cre (Emx1cre/+). Sustained MEK/MAPK activity in the ventral telencephalon (Gsx2e-cre; RosaMEK1DD/+) expanded dorsal lateral ganglionic eminence (dLGE) enriched genes (Gsx2 and Sp8) and oligodendrocyte progenitor cell (OPC) markers (Olig2, Pdgfrα, and Sox10), and also reduced markers in the ventral (v) LGE domain (Isl1 and Foxp1). Activation of MEK/MAPK activity in the dorsal telencephalon (Emx1cre/+; RosaMEK1DD/+) did not initially activate the expression of dLGE or OPC genes at E15.5 but ectopic expression of Gsx2 and OPC markers were observed at E18.5. These results support the idea that MAPK activity as readout by p-ERK1/2 and Etv5 expression is enriched in distinct subdomains of ventral telencephalic progenitors during development. In addition, sustained activation of the MEK/MAPK pathway in the ventral or dorsal telencephalon influences dLGE and OPC identity from progenitors.


Assuntos
Diferenciação Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Telencéfalo/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Gânglios/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , MAP Quinase Quinase 1/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Fatores de Transcrição SOXE/genética , Telencéfalo/embriologia , Telencéfalo/fisiologia , Fatores de Transcrição/metabolismo
5.
Dev Biol ; 442(1): 115-126, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29990475

RESUMO

The homeobox gene Gsx2 has previously been shown to inhibit oligodendroglial specification in dorsal lateral ganglionic eminence (dLGE) progenitors of the ventral telencephalon. The precocious specification of oligodendrocyte progenitor cells (OPCs) observed in Gsx2 mutants, however, is transient and begins to normalize by late stages of embryogenesis. Interestingly, this normalization correlates with the expansion of Gsx1, a close homolog of Gsx2, in a subset of progenitors in the Gsx2 mutant LGE. Here, we interrogated the mechanisms underlying oligodendroglial specification in Gsx2 mutants in relation to Gsx1. We found that Gsx1/2 double mutant embryos exhibit a more robust expansion of Olig2+ cells (i.e. OPCs) in the subventricular zone (SVZ) of the dLGE than Gsx2 mutants. Moreover, misexpression of Gsx1 throughout telencephalic VZ progenitors from E15 and onward resulted in a significant reduction of cortical OPCs. These results demonstrate redundant roles of Gsx1 and Gsx2 in suppressing early OPC specification in LGE VZ progenitors. However, Gsx1/2 mutants did not show a significant increase in adjacent cortical OPCs at later stages compared to Gsx2 mutants. This is likely due to reduced proliferation of OPCs within the SVZ of the Gsx1/2 double mutant LGE, suggesting a novel role for Gsx1 in expansion of migrating OPCs in the ventral telencephalon. We further investigated the glial specification mechanisms downstream of Gsx2 by generating Olig2/Gsx2 double mutants. Consistent with the known essential role for Olig2 in OPC specification, ectopic production of cortical OPCs observed in Gsx2 mutants disappeared in Olig2/Gsx2 double mutants. These mutants, however, maintained the expanded expression of gliogenic markers Zbtb20 and Bcan in the VZ of the LGE similarly to Gsx2 single mutants, suggesting that Gsx2 suppresses gliogenesis via Olig2-dependent and -independent mechanisms.


Assuntos
Proteínas de Homeodomínio/genética , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula , Embrião de Mamíferos/metabolismo , Gânglios/metabolismo , Gânglios/fisiologia , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neuroglia/metabolismo , Neuroglia/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Telencéfalo/metabolismo , Fatores de Transcrição
6.
PLoS Genet ; 14(3): e1007227, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29538372

RESUMO

Esophageal cancer occurs as either squamous cell carcinoma (ESCC) or adenocarcinoma. ESCCs comprise almost 90% of cases worldwide, and recur with a less than 15% five-year survival rate despite available treatments. The identification of new ESCC drivers and therapeutic targets is critical for improving outcomes. Here we report that expression of the human DEK oncogene is strongly upregulated in esophageal SCC based on data in the cancer genome atlas (TCGA). DEK is a chromatin-associated protein with important roles in several nuclear processes including gene transcription, epigenetics, and DNA repair. Our previous data have utilized a murine knockout model to demonstrate that Dek expression is required for oral and esophageal SCC growth. Also, DEK overexpression in human keratinocytes, the cell of origin for SCC, was sufficient to cause hyperplasia in 3D organotypic raft cultures that mimic human skin, thus linking high DEK expression in keratinocytes to oncogenic phenotypes. However, the role of DEK over-expression in ESCC development remains unknown in human cells or genetic mouse models. To define the consequences of Dek overexpression in vivo, we generated and validated a tetracycline responsive Dek transgenic mouse model referred to as Bi-L-Dek. Dek overexpression was induced in the basal keratinocytes of stratified squamous epithelium by crossing Bi-L-Dek mice to keratin 5 tetracycline transactivator (K5-tTA) mice. Conditional transgene expression was validated in the resulting Bi-L-Dek_K5-tTA mice and was suppressed with doxycycline treatment in the tetracycline-off system. The mice were subjected to an established HNSCC and esophageal carcinogenesis protocol using the chemical carcinogen 4-nitroquinoline 1-oxide (4NQO). Dek overexpression stimulated gross esophageal tumor development, when compared to doxycycline treated control mice. Furthermore, high Dek expression caused a trend toward esophageal hyperplasia in 4NQO treated mice. Taken together, these data demonstrate that Dek overexpression in the cell of origin for SCC is sufficient to promote esophageal SCC development in vivo.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proteínas de Ligação a DNA/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Proteínas Oncogênicas/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Proteínas de Ligação a DNA/metabolismo , Epitélio/patologia , Neoplasias Esofágicas/induzido quimicamente , Carcinoma de Células Escamosas do Esôfago , Regulação Neoplásica da Expressão Gênica , Queratinócitos/patologia , Camundongos Transgênicos , Proteínas Oncogênicas/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Tetraciclina/farmacologia , Língua/efeitos dos fármacos , Língua/patologia , Transgenes
7.
Dev Dyn ; 247(1): 222-228, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28744915

RESUMO

BACKGROUND: The lateral ganglionic eminence (LGE) in the ventral telencephalon is a diverse progenitor domain subdivided by distinct gene expression into a dorsal region (dLGE) that gives rise to olfactory bulb and amygdalar interneurons and a ventral region (vLGE) that gives rise to striatal projection neurons. The homeobox gene, Gsx2, is an enriched marker of the LGE and is expressed in a high dorsal to low ventral gradient in the ventricular zone (VZ) as development proceeds. Aside from Gsx2, markers restricted to the VZ in the dLGE and/or vLGE remain largely unknown. RESULTS: Here, we show that the gene and protein expression of Glucocorticoid-induced transcript 1 (Glcci1) has a similar dorsal to ventral gradient of expression in the VZ as Gsx2. We found that Glcci1 gene and protein expression are reduced in Gsx2 mutants, and are increased in the cortex after early and late Gsx2 misexpression. Moreover, Glcci1 expressing cells are restricted to a subpopulation of Gsx2 positive cells on the basal side of the VZ and co-express Ascl1, but not the subventricular zone dLGE marker, Sp8. CONCLUSIONS: These findings suggest that Glcci1 is a new marker of a subpopulation of LGE VZ progenitor cells in the Gsx2 lineage. Developmental Dynamics 247:222-228, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Neurais/metabolismo , Receptores de Glucocorticoides/metabolismo , Telencéfalo/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Receptores de Glucocorticoides/genética , Telencéfalo/embriologia
8.
J Neurosci ; 34(10): 3767-78, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24599474

RESUMO

The protein tyrosine phosphatase Shp2 (PTPN11) is crucial for normal brain development and has been implicated in dorsal telencephalic neuronal and astroglia cell fate decisions. However, its roles in the ventral telencephalon and during oligodendrogenesis in the telencephalon remain largely unknown. Shp2 gain-of-function (GOF) mutations are observed in Noonan syndrome, a type of RASopathy associated with multiple phenotypes, including cardiovascular, craniofacial, and neurocognitive abnormalities. To gain insight into requirements for Shp2 (LOF) and the impact of abnormal Shp2 GOF mutations, we used a Shp2 conditional mutant allele (LOF) and a cre inducible Shp2-Q79R GOF transgenic mouse in combination with Olig2(cre/+) mice to target embryonic ventral telencephalic progenitors and the oligodendrocyte lineage. In the absence of Shp2 (LOF), neuronal cell types originating from progenitors in the ventral telencephalon were generated, but oligodendrocyte progenitor cell (OPC) generation was severely impaired. Late embryonic and postnatal Shp2 cKOs showed defects in the generation of OPCs throughout the telencephalon and subsequent reductions in white matter myelination. Conversely, transgenic expression of the Shp2 GOF Noonan syndrome mutation resulted in elevated OPC numbers in the embryo and postnatal brain. Interestingly, expression of this mutation negatively influenced myelination as mice displayed abnormal myelination and fewer myelinated axons in the white matter despite elevated OPC numbers. Increased proliferating OPCs and elevated MAPK activity were also observed during oligodendrogenesis after expression of Shp2 GOF mutation. These results support the notion that appropriate Shp2 activity levels control the number as well as the differentiation of oligodendrocytes during development.


Assuntos
Fibras Nervosas Mielinizadas/enzimologia , Oligodendroglia/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/biossíntese , Células-Tronco/enzimologia , Telencéfalo/embriologia , Telencéfalo/enzimologia , Animais , Diferenciação Celular/fisiologia , Camundongos , Camundongos Transgênicos , Telencéfalo/citologia
9.
Proc Natl Acad Sci U S A ; 110(42): E4026-35, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24082127

RESUMO

The mammalian striatum controls the output of the basal ganglia via two distinct efferent pathways, the direct (i.e., striatonigral) and the indirect (i.e., striatopallidal) pathways. The LIM homeodomain transcription factor Islet1 (Isl1) is expressed in a subpopulation of striatal progenitors; however, its specific role in striatal development remains unknown. Our genetic fate-mapping results show that Isl1-expressing progenitors give rise to striatal neurons belonging to the striatonigral pathway. Conditional inactivation of Isl1 in the telencephalon resulted in a smaller striatum with fewer striatonigral neurons and reduced projections to the substantia nigra. Additionally, conditional inactivation in the ventral forebrain (including both the telencephalon and diencephalon) revealed a unique role for Isl1 in diencephalic cells bordering the internal capsule for the normal development of the striatonigral pathway involving PlexinD1-Semaphorin 3e (Sema3e) signaling. Finally, Isl1 conditional mutants displayed a hyperlocomotion phenotype, and their locomotor response to psychostimulants was significantly blunted, indicating that the alterations in basal ganglia circuitry contribute to these mutant behaviors.


Assuntos
Corpo Estriado/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Substância Negra/embriologia , Fatores de Transcrição/metabolismo , Animais , Comportamento Animal/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Corpo Estriado/citologia , Proteínas do Citoesqueleto , Glicoproteínas/genética , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Homeodomínio LIM/genética , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Semaforinas , Substância Negra/citologia , Fatores de Transcrição/genética
10.
J Neurosci ; 30(20): 6944-53, 2010 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-20484636

RESUMO

We have taken a genetic-based fate-mapping approach to determine the specific contributions of telencephalic progenitors to the structures that comprise the amygdalar fear circuit including the central (CA), lateral (LA), and basolateral (BLA) amygdala. Our data indicate that progenitors in the ventral pallium (VP) contribute projection neurons to the LA and BLA but not the CA. Rather, the CA appears to derive, at least in part, from progenitors located in the ventral lateral ganglionic eminence (vLGE). Diverse groups of interneurons populate these amygdalar nuclei, and as predicted our data support the notion that they originate from subpallial progenitors. A rather specific population of amygdalar interneurons, the intercalated cells (ITCs), is known to play a fundamental role in fear-related behaviors. However, no information on their specific origin has, as yet, been provided. Our findings suggest that the ITCs arise from the dorsal lateral ganglionic eminence (dLGE) and migrate in the lateral migratory stream to populate the paracapsular regions as well as the main intercalated mass of the amygdala (IA). Germ-line Gsx2 mutants are known to exhibit an expansion of the VP into the LGE and a concomitant reduction in the dLGE and vLGE. Accordingly, Gsx2 conditional mutants display a significantly enlarged LA and a significant reduction in ITCs both within the paracapsular regions and the IA. Additional support for a dLGE origin of the ITCs was obtained in conditional mutants of the dLGE gene Sp8. Thus, our findings indicate diverse origins for the neuronal components that comprise the amygdalar fear circuit.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/crescimento & desenvolvimento , Medo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/fisiologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal , Mapeamento Encefálico , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/fisiologia , Neurônios/classificação , Fatores de Transcrição/genética
11.
Neuron ; 63(4): 451-65, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19709628

RESUMO

The homeobox gene Gsx2 (formerly Gsh2) is known to be required for striatal and olfactory bulb neurogenesis; however, its specific role in the specification of these two neuronal subtypes remains unclear. To address this, we have employed a temporally regulated gain-of-function approach in transgenic mice and found that misexpression of Gsx2 at early stages of telencephalic neurogenesis favors the specification of striatal projection neuron identity over that of olfactory bulb interneurons. In contrast, delayed activation of the Gsx2 transgene until later stages exclusively promotes olfactory bulb interneuron identity. In a complementary approach, we have conditionally inactivated Gsx2 in a temporally progressive manner. Unlike germline Gsx2 mutants, which exhibit severe alterations in both striatal and olfactory bulb neurogenesis at birth, the conditional mutants exhibited defects restricted to olfactory bulb interneurons. These results demonstrate that Gsx2 specifies striatal projection neuron and olfactory bulb interneuron identity at distinct time points during telencephalic neurogenesis.


Assuntos
Corpo Estriado/patologia , Genes Homeobox/fisiologia , Proteínas de Homeodomínio/genética , Interneurônios/patologia , Neurogênese/genética , Bulbo Olfatório/patologia , Animais , Corpo Estriado/embriologia , Feminino , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Bulbo Olfatório/embriologia , Gravidez , Fatores de Tempo
12.
Stress ; 9(3): 153-63, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17060049

RESUMO

We examined the ontogeny of the corticosterone response to (+)-methamphetamine in neonatal rats. In experiment-1, animals were injected with 10 mg/kg of (+)-methamphetamine or saline and plasma corticosterone levels were examined in separate groups 30 or 105 min later on postnatal day (P) 1, 3, 5, 7, 9, 11, 13, 15, 17, or 19. The adrenal response to methamphetamine was best described by a U-shaped function with the nadir of corticosterone release occurring between P7 and P13. Experiment-2 was similar except that the effect of four consecutive days of exposure to (+)-methamphetamine (four times daily at 2 h intervals with 10 mg/kg) was assessed with a single final dose early on the fifth day (i.e. P1-5, 3-7, 5-9, 7-11, 9-13, 11-15, 13-17, 15-19). The 30 min corticosterone response after multiple methamphetamine doses was augmented compared to single exposures, with the exception of the two earliest dosing intervals ending on P5 and P7, where the responses were lower. In addition, at 105 min, the levels of corticosterone were attenuated relative to a single drug administration. With the exception of animals receiving methamphetamine from P15 to P19, thymus weights were unaffected. The data demonstrate that (+)-methamphetamine is a robust activator of corticosterone release in developing animals and this release is extensively modified by age and previous drug exposure.


Assuntos
Glândulas Suprarrenais/metabolismo , Corticosterona/metabolismo , Metanfetamina/farmacologia , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Peso Corporal , Corticosterona/sangue , Feminino , Cinética , Tamanho da Ninhada de Vivíparos , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
13.
J Neurochem ; 98(5): 1369-78, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16923155

RESUMO

We have previously shown that neonatal administration of (+/-)3,4-methylenedioxymethamphetamine and (+)fenfluramine produce deficits in spatial and path integration learning, whereas (+)methamphetamine causes deficits in spatial learning. Conversely, cocaine and (+/-)methylphenidate have no effect on either form of learning following neonatal administration. The purpose of the present study was to determine whether corticosterone and/or monoamine levels were changed following subcutaneous administration of 10 mg/kg (+)methamphetamine, (+/-)3,4-methylenedioxymethamphetamine, (+)fenfluramine, (+/-)methylphenidate or cocaine every 2 h (total of four injections) on postnatal day 11. Twenty-four hours after the first dose, plasma, striatum and hippocampus were collected. Corticosterone levels were increased in methamphetamine-, fenfluramine-, methylenedioxymethamphetamine- and methylphenidate-treated rats relative to levels in saline-treated rats, whereas cocaine-treated rats were unaffected. In the striatum and hippocampus, serotonin and 5-hydroxyindolacetic acid were reduced in animals treated with methylenedioxymethamphetamine or fenfluramine, compared with levels in saline controls. Dopamine levels were not changed by any of the drugs, although 3,4-dihydroxyphenylacetic acid was decreased following methylenedioxymethamphetamine or methamphetamine. Minimal effects were seen in neurotransmitter levels following injection of cocaine or methylphenidate. These data suggest that drugs that affect corticosterone and hippocampal serotonin are associated with both spatial learning and path integration deficits, and those that affect corticosterone and 3,4-dihydroxyphenylacetic acid are associated with spatial learning deficits only.


Assuntos
Monoaminas Biogênicas/metabolismo , Química Encefálica/efeitos dos fármacos , Corticosterona/sangue , Inibidores da Captação de Neurotransmissores/administração & dosagem , 3,4-Metilenodioxianfetamina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão/métodos , Cocaína/farmacologia , Corpo Estriado/química , Corpo Estriado/efeitos dos fármacos , Eletroquímica/métodos , Feminino , Fenfluramina/farmacologia , Hipocampo/química , Hipocampo/efeitos dos fármacos , Técnicas Imunoenzimáticas/métodos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Metanfetamina/farmacologia , Metilfenidato/farmacologia , Gravidez , Ratos , Ratos Sprague-Dawley
14.
Eur J Neurosci ; 22(6): 1265-76, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16190882

RESUMO

Npas3 is a member of the bHLH-PAS superfamily of transcription factors that is expressed broadly in the developing neuroepithelium. To study the function of this gene, mice deficient in Npas3 were generated and characterized. Npas3-/- mice were growth-retarded and exhibited developmental brain abnormalities that included a reduction in size of the anterior hippocampus, hypoplasia of the corpus callosum and enlargement of the ventricles. A number of behavioural abnormalities were identified in Npas3-/- mice including locomotor hyperactivity, subtle gait defects, impairment of prepulse inhibition of acoustic startle, deficit in recognition memory and altered anxiety-related responses. Characterization of neurosignaling pathways using several pharmacological agents revealed dysfunctional glutamate, dopamine and serotonin neurotransmitter signaling. Consistent with these findings, we identified a significant alteration in cortical PSD-95 expression, a PDZ-containing protein that has been shown to be involved in postsynaptic signal transduction. Together, our observations indicate an important role for Npas3 in controlling normal brain development and neurosignaling pathways.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Comportamento Animal/fisiologia , Sistema Nervoso/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Encéfalo/anormalidades , Proteína 4 Homóloga a Disks-Large , Marcha Atáxica/genética , Marcha Atáxica/fisiopatologia , Guanilato Quinases , Peptídeos e Proteínas de Sinalização Intracelular/genética , Imageamento por Ressonância Magnética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Atividade Motora/genética , Atividade Motora/fisiologia , Neurotransmissores/metabolismo , Equilíbrio Postural/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Brain Res ; 1039(1-2): 97-107, 2005 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-15781051

RESUMO

We have previously shown that +/-3,4-methylenedioxymethamphetamine (MDMA) treatment from P11 to P20 in rats produces deficits in cognitive ability when these animals are tested in adulthood. The purpose of this experiment was to explore the neuroendocrine and neurochemical changes produced by MDMA treatment on P11. We examined monoamines in the hippocampus and striatum and the serotonin transporter in the hippocampus as well as pituitary and adrenal output following administration of MDMA (10 mg/kg, 4 times) on postnatal day 11. Significant depletions in serotonin were evident in the hippocampus 1 h and in the striatum 24 h after the first dose and remained reduced 78 h later. No changes in serotonin transporter were observed following MDMA treatment, although females had lower levels than males. No changes in dopamine were detected. The metabolites of serotonin and dopamine had different profiles than the parent compounds after MDMA administration. Plasmatic ACTH was elevated immediately following MDMA and remained elevated for at least 1 h after the last dose and returned to baseline by 24 h. Corticosterone was increased after the first dose and remained increased for at least 24 h, and returned to baseline by 30 h. The decreases in serotonin in regions important for learning and memory in conjunction with elevated levels of corticosterone during a period of stress hyporesponsiveness suggest that these initial responses to MDMA may contribute to the long-term learning and memory deficits following neonatal MDMA exposure.


Assuntos
Corpo Estriado/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Serotoninérgicos/toxicidade , Hormônio Adrenocorticotrópico/sangue , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Corpo Estriado/metabolismo , Corticosterona/sangue , Dopamina/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Glicoproteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina , Fatores Sexuais , Timo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...