Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 544: 128-137, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38447690

RESUMO

In Robo3cKO mice, midline crossing defects of the trigeminothalamic projections from the trigeminal principal sensory nucleus result in bilateral whisker maps in the somatosensory thalamus and consequently in the face representation area of the primary somatosensory (S1) cortex (Renier et al., 2017; Tsytsarev et al., 2017). We investigated whether this bilateral sensory representation in the whisker-barrel cortex is also reflected in the downstream projections from the S1 to the primary motor (M1) cortex. To label these projections, we injected anterograde viral axonal tracer in S1 cortex. Corticocortical projections from the S1 distribute to similar areas across the ipsilateral hemisphere in control and Robo3cKO mice. Namely, in both genotypes they extend to the M1, premotor/prefrontal cortex (PMPF), secondary somatosensory (S2) cortex. Next, we performed voltage-sensitive dye imaging (VSDi) in the left hemisphere following ipsilateral and contralateral single whisker stimulation. While controls showed only activation in the contralateral whisker barrel cortex and M1 cortex, the Robo3cKO mouse left hemisphere was activated bilaterally in both the barrel cortex and the M1 cortex. We conclude that the midline crossing defect of the trigeminothalamic projections leads to bilateral whisker representations not only in the thalamus and the S1 cortex but also downstream from the S1, in the M1 cortex.


Assuntos
Córtex Motor , Córtex Somatossensorial , Camundongos , Animais , Córtex Somatossensorial/fisiologia , Vibrissas/fisiologia , Córtex Motor/fisiologia , Tálamo/diagnóstico por imagem , Núcleos do Trigêmeo
2.
Neuroscience ; 512: 85-98, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36549605

RESUMO

In Alzheimer's disease and related dementias, amyloid beta (Aß) and amyloid plaques can disrupt long-term synaptic plasticity, learning and memory and cognitive function. Plaque accumulation can disrupt corticocortical circuitry leading to abnormalities in sensory, motor, and cognitive processing. In this study, using 5xFAD (five Familial Alzheimer's Disease - FAD - mutations) mice, we evaluated amyloid plaque formation in different cortical areas, and whether differential amyloid accumulation across cortical fields correlates with changes in dendritic complexity of layer 3 corticocortical projection neurons and functional responses in the primary somatosensory cortex following whisker stimulation. We focused on three cortical areas: the primary somatosensory cortex (S1), the primary motor cortex (M1), and the prefrontal cortex (PFC including the anterior cingulate, prelimbic, and infralimbic subdivisions). We found that Aß and amyloid plaque accumulation is not uniform across 5xFAD cortical areas, while there is no expression in littermate controls. We also found that there are differential layer 3 pyramidal cell dendritic complexity changes across the three areas in 5xFAD mice, compared to same age controls, with no apparent relation to differential amyloid accumulation. We used voltage-sensitive dye imaging (VSDi) to visualize neural activity in S1, M1 and PFC following whisker activation. Control mice show normal physiological responses in all three cortical areas, whereas 5xFAD mice only display physiological responses in S1. Taken together our results show that 5xFAD mutation affects the overall dendritic morphology of layer 3 pyramidal cells across sensory-motor and association cortex irrespective of the density and distribution of the Aß amyloid proteins. Corticocortical circuitry between the sensory and motor/association areas is most likely disrupted in 5xFAD mice as cortical responses to whisker stimulation are altered.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Camundongos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Placa Amiloide
3.
Neuroscience ; 494: 140-151, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35598701

RESUMO

In Robo3R3-5cKO mouse brain, rhombomere 3-derived trigeminal principal nucleus (PrV) neurons project bilaterally to the somatosensory thalamus. As a consequence, whisker-specific neural modules (barreloids and barrels) representing whiskers on both sides of the face develop in the sensory thalamus and the primary somatosensory cortex. We examined the morphological complexity of layer 4 barrel cells, their postsynaptic partners in layer 3, and functional specificity of layer 3 pyramidal cells. Layer 4 spiny stellate cells form much smaller barrels and their dendritic fields are more focalized and less complex compared to controls, while layer 3 pyramidal cells did not show notable differences. Using in vivo 2-photon imaging of a genetically encoded fluorescent [Ca2+] sensor, we visualized neural activity in the normal and Robo3R3-5cKO barrel cortex in response to ipsi- and contralateral single whisker stimulation. Layer 3 neurons in control animals responded only to their contralateral whiskers, while in the mutant cortex layer 3 pyramidal neurons showed both ipsi- and contralateral whisker responses. These results indicate that bilateral whisker map inputs stimulate different but neighboring groups of layer 3 neurons which normally relay contralateral whisker-specific information to other cortical areas.


Assuntos
Córtex Somatossensorial , Vibrissas , Animais , Camundongos , Neurônios/fisiologia , Células Piramidais/fisiologia , Córtex Somatossensorial/fisiologia , Tálamo , Vibrissas/fisiologia
4.
Biol Open ; 10(6)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34100899

RESUMO

Genetic studies have linked FAT1 (FAT atypical cadherin 1) with autism spectrum disorder (ASD); however, the role that FAT1 plays in ASD remains unknown. In mice, the function of Fat1 has been primarily implicated in embryonic nervous system development with less known about its role in postnatal development. We show for the first time that FAT1 protein is expressed in mouse postnatal brains and is enriched in the cerebellum, where it localizes to granule neurons and Golgi cells in the granule layer, as well as inhibitory neurons in the molecular layer. Furthermore, subcellular characterization revealed FAT1 localization in neurites and soma of granule neurons, as well as being present in the synaptic plasma membrane and postsynaptic densities. Interestingly, FAT1 expression was decreased in induced pluripotent stem cell (iPSC)-derived neural precursor cells (NPCs) from individuals with ASD. These findings suggest a novel role for FAT1 in postnatal development and may be particularly important for cerebellum function. As the cerebellum is one of the vulnerable brain regions in ASD, our study warrants further investigation of FAT1 in the disease etiology.


Assuntos
Transtorno Autístico/etiologia , Caderinas/genética , Suscetibilidade a Doenças , Animais , Transtorno Autístico/metabolismo , Biomarcadores , Caderinas/metabolismo , Cerebelo/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipocampo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Interneurônios/metabolismo , Camundongos , Neuritos/metabolismo , Transporte Proteico , Transcriptoma
5.
Front Behav Neurosci ; 12: 261, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30483073

RESUMO

Altered neuronal connectivity has been implicated in the pathophysiology of Autism Spectrum Disorder (ASD). SLIT/ROBO signaling plays an important role in developmental processes of neuronal connectivity, including axon guidance, neuronal migration, and axonal and dendritic branching. Genetic evidence supports that SLIT3, one of the genes encoding SLITs, is associated with ASD. Yet the causal link between SLIT3 mutation and autism symptoms has not been examined. Here we assessed ASD-associated behaviors in Slit3 knockout (KO) mice. Our data showed that Slit3-KO mice exhibited reduced marble burying behaviors but normal social behaviors. In addition, Slit3-KO mice displayed hypolocomotion in the open field test and impaired motor coordination in the rotarod test. Anxiety-like behaviors were mainly observed in female KO mice assessed by three types of behavioral tests, namely, the open field test, elevated plus maze test, and light/dark box test. No differences were observed between KO and wildtype mice in recognition memory in the novel object recognition test or depression-like behavior in the tail suspension test. Taken together, loss of Slit3 may result in disrupted neural circuits related to motor function and increased anxiety-like states, which are co-occurring symptoms in ASD.

6.
Cell Transplant ; 25(6): 1085-99, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26395573

RESUMO

Stem cell therapy is under active investigation for traumatic brain injury (TBI). Noninvasive stem cell delivery is the preferred method, but retention of stem cells at the site of injury in TBI has proven challenging and impacts effectiveness. To investigate the effects of applying a magnetic field on cell homing and retention, we delivered human neuroprogenitor cells (hNPCs) labeled with a superparamagnetic nanoparticle into post-TBI animals in the presence of a static magnetic field. We have previously devised a method of loading hNPCs with ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles Molday ION Rhodamine B (MIRB™). Labeling of hNPCs (MIRB-hNPCs) does not affect hNPC viability, proliferation, or differentiation. The 0.6 tesla (T) permanent magnet was placed ∼4 mm above the injured parietal cortex prior to intracarotid injection of 4 × 10(4) MIRB-hNPCs. Fluorescence imaging, Perls' Prussian blue histochemistry, immunocytochemistry with SC121, a human-specific antibody, and T2-weighted magnetic resonance imaging ex vivo revealed there was increased homing and retention of MIRB-hNPCs in the injured cortex as compared to the control group in which MIRB-hNPCs were injected in the absence of a static magnetic field. Fluoro-Jade C staining and immunolabeling with specific markers confirmed the viability status of MIRB-hNPCs posttransplantation. These results show that increased homing and retention of MIRB-hNPCs post-TBI by applying a static magnetic field is a promising technique to deliver cells into the CNS for treatment of neurological injuries and neurodegenerative diseases.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Magnetismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Animais , Lesões Encefálicas Traumáticas/patologia , Morte Celular , Humanos , Inflamação/patologia , Campos Magnéticos , Imageamento por Ressonância Magnética , Masculino , Necrose , Células-Tronco Neurais/metabolismo , Ratos Sprague-Dawley , Rodaminas/metabolismo
7.
Cereb Cortex ; 24(1): 261-79, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23042739

RESUMO

Neural progenitor cells have the ability to give rise to neurons and glia in the embryonic, postnatal and adult brain. During development, the program regulating whether these cells divide and self-renew or exit the cell cycle and differentiate is tightly controlled, and imbalances to the normal trajectory of this process can lead to severe functional consequences. However, our understanding of the molecular regulation of these fundamental events remains limited. Moreover, processes underpinning development of the postnatal neurogenic niches within the cortex remain poorly defined. Here, we demonstrate that Nuclear factor one X (NFIX) is expressed by neural progenitor cells within the embryonic hippocampus, and that progenitor cell differentiation is delayed within Nfix(-/-) mice. Moreover, we reveal that the morphology of the dentate gyrus in postnatal Nfix(-/-) mice is abnormal, with fewer subgranular zone neural progenitor cells being generated in the absence of this transcription factor. Mechanistically, we demonstrate that the progenitor cell maintenance factor Sry-related HMG box 9 (SOX9) is upregulated in the hippocampus of Nfix(-/-) mice and demonstrate that NFIX can repress Sox9 promoter-driven transcription. Collectively, our findings demonstrate that NFIX plays a central role in hippocampal morphogenesis, regulating the formation of neuronal and glial populations within this structure.


Assuntos
Diferenciação Celular/fisiologia , Hipocampo/embriologia , Fatores de Transcrição NFI/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Contagem de Células , Corantes , Biologia Computacional , Giro Denteado/embriologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Eletroporação , Feminino , Hematoxilina , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/metabolismo , Inclusão em Parafina , Gravidez , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real
8.
Int J Nanomedicine ; 8: 4593-600, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24348036

RESUMO

Ultrasmall superparamagnetic iron-oxide particles (USPIOs) loaded into stem cells have been suggested as a way to track stem cell transplantation with magnetic resonance imaging, but the labeling, and post-labeling proliferation, viability, differentiation, and retention of USPIOs within the stem cells have yet to be determined for each type of stem cell and for each type of USPIO. Molday ION Rhodamine B™ (BioPAL, Worcester, MA, USA) (MIRB) has been shown to be a USPIO labeling agent for mesenchymal stem cells, glial progenitor cells, and stem cell lines. In this study, we have evaluated MIRB labeling in human neuroprogenitor cells and found that human neuroprogenitor cells are effectively labeled with MIRB without use of transfection reagents. Viability, proliferation, and differentiation properties are unchanged between MIRB-labeled neuroprogenitors cells and unlabeled cells. Moreover, MIRB-labeled human neuroprogenitor cells can be frozen, thawed, and replated without loss of MIRB or even without loss of their intrinsic biology. Overall, those results show that MIRB has advantageous properties that can be used for cell-based therapy.


Assuntos
Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Corantes/toxicidade , Nanopartículas de Magnetita/toxicidade , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células Cultivadas , Corantes/química , Corantes/farmacocinética , Humanos , Nanopartículas de Magnetita/química , Células-Tronco Neurais/química , Células-Tronco Neurais/metabolismo , Fenótipo , Rodaminas/química , Rodaminas/farmacocinética , Rodaminas/toxicidade , Coloração e Rotulagem
9.
J Neurosci ; 33(7): 2916-26, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23407950

RESUMO

Evidence for coexpression of two or more classic neurotransmitters in neurons has increased, but less is known about cotransmission. Ventral tegmental area (VTA) neurons corelease dopamine (DA), the excitatory transmitter glutamate, and the inhibitory transmitter GABA onto target cells in the striatum. Olfactory bulb (OB) short axon cells (SACs) form interglomerular connections and coexpress markers for DA and GABA. Using an optogenetic approach, we provide evidence that mouse OB SACs release both GABA and DA onto external tufted cells (ETCs) in other glomeruli. Optical activation of channelrhodopsin specifically expressed in DAergic SACs produced a GABA(A) receptor-mediated monosynaptic inhibitory response, followed by DA-D(1)-like receptor-mediated excitatory response in ETCs. The GABA(A) receptor-mediated hyperpolarization activates I(h) current in ETCs; synaptically released DA increases I(h), which enhances postinhibitory rebound spiking. Thus, the opposing actions of synaptically released GABA and DA are functionally integrated by I(h) to generate an inhibition-to-excitation "switch" in ETCs. Consistent with the established role of I(h) in ETC burst firing, we show that endogenous DA release increases ETC spontaneous bursting frequency. ETCs transmit sensory signals to mitral/tufted output neurons and drive intraglomerular inhibition to shape glomerulus output to downstream olfactory networks. GABA and DA cotransmission from SACs to ETCs may play a key role in regulating output coding across the glomerular array.


Assuntos
Axônios/metabolismo , Dopamina/metabolismo , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Axônios/fisiologia , Channelrhodopsins , Estimulação Elétrica , Glutamato Descarboxilase/metabolismo , Humanos , Imuno-Histoquímica , Sistema Justaglomerular/citologia , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/fisiologia , Bulbo Olfatório/citologia , Técnicas de Patch-Clamp , Estimulação Luminosa , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Transmissão Sináptica/fisiologia , Tirosina 3-Mono-Oxigenase/genética
10.
J Comp Neurol ; 520(14): 3135-49, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22886731

RESUMO

The nuclear factor one (NFI) family of transcription factors consists of four members in vertebrates, NFIA, NFIB, NFIC, and NFIX, which share a highly conserved N-terminal DNA-binding domain. NFI genes are widely expressed in the developing mouse brain, and mouse mutants lacking NFIA, NFIB, or NFIX exhibit developmental deficits in several areas, including the cortex, hippocampus, pons, and cerebellum. Here we analyzed the expression of NFIA and NFIB in the developing and adult olfactory bulb (OB), rostral migratory stream (RMS), and subventricular zone (SVZ). We found that NFIA and NFIB are expressed within these regions during embryonic and postnatal development and in the adult. Immunohistochemical analysis using cell-type-specific markers revealed that migrating neuroblasts in the adult brain express NFI transcription factors, as do astrocytes within the RMS and progenitor cells within the SVZ. Moreover, astrocytes within the OB express NFIA, whereas mitral cells within the OB express NFIB. Taken together these data show that NFIA and NFIB are expressed in both the developing and the adult OB and in the RMS and SVZ, indicative of a regulatory role for these transcription factors in the development of this facet of the olfactory system.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Transcrição NFI/genética , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Fatores Etários , Animais , Especificidade de Anticorpos , Movimento Celular/fisiologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Ventrículos Laterais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Transcrição NFI/imunologia , Fatores de Transcrição NFI/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Bulbo Olfatório/embriologia , Bulbo Olfatório/crescimento & desenvolvimento , Gravidez
11.
J Neurosci ; 32(17): 6000-13, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539860

RESUMO

The extracellular levels of excitatory amino acids are kept low by the action of the glutamate transporters. Glutamate/aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) are the most abundant subtypes and are essential for the functioning of the mammalian CNS, but the contribution of the EAAC1 subtype in the clearance of synaptic glutamate has remained controversial, because the density of this transporter in different tissues has not been determined. We used purified EAAC1 protein as a standard during immunoblotting to measure the concentration of EAAC1 in different CNS regions. The highest EAAC1 levels were found in the young adult rat hippocampus. Here, the concentration of EAAC1 was ∼0.013 mg/g tissue (∼130 molecules µm⁻³), 100 times lower than that of GLT-1. Unlike GLT-1 expression, which increases in parallel with circuit formation, only minor changes in the concentration of EAAC1 were observed from E18 to adulthood. In hippocampal slices, photolysis of MNI-D-aspartate (4-methoxy-7-nitroindolinyl-D-aspartate) failed to elicit EAAC1-mediated transporter currents in CA1 pyramidal neurons, and D-aspartate uptake was not detected electron microscopically in spines. Using EAAC1 knock-out mice as negative controls to establish antibody specificity, we show that these relatively small amounts of EAAC1 protein are widely distributed in somata and dendrites of all hippocampal neurons. These findings raise new questions about how so few transporters can influence the activation of NMDA receptors at excitatory synapses.


Assuntos
Sistema Nervoso Central/citologia , Transportador 3 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/metabolismo , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Ácido Aspártico/farmacologia , Sistema Nervoso Central/anatomia & histologia , Ácido D-Aspártico/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Inibidores Enzimáticos/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Transportador 2 de Aminoácido Excitatório/deficiência , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/deficiência , Transportador 3 de Aminoácido Excitatório/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/metabolismo , Técnicas In Vitro , Rim/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica , Proteína Básica da Mielina/metabolismo , Neurônios/efeitos dos fármacos , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Proteolipídeos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Frações Subcelulares/metabolismo , Sinaptofisina/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato
12.
J Mol Histol ; 43(2): 215-21, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22389027

RESUMO

Olfactory bulb interneurons are continuously generated in the subventricular zone (SVZ) and migrate along the rostral migratory stream (RMS) into the olfactory bulb (OB) where the majority becomes local GABAergic interneurons. We previously showed that SVZ-derived progenitor cells expressed glutamic acid decarboxylase 65 kDa (GAD65) very early in the migratory pathway. However, only approximately half of OB GABAergic interneurons use GAD65, an equal number express the 67 kDa GAD enzyme. To investigate the differentiation of these GABAergic interneurons we examined their migration in a transgenic mouse expressing green fluorescent protein (GFP) under the control of the GAD67 promoter. In adult, GFP was expressed by a subpopulation of migratory cells in the SVZ and along the RMS. Using Doublecortin (DCX) as a marker of migrating neuroblasts and bromodeoxyuridine (BrdU) incorporation, we show that these GAD67-GFP neurons co-express DCX and incorporate BrdU indicating they are newly born migratory neuroblasts. This is similar to GAD65 transgene expression, and in contrast to dopaminergic interneuron transgene expression which occurs only after cells reach the olfactory bulb. Although the GAD65/67 transgenes are expressed early in migration, there is minimal protein production in the cells prior to reaching the OB. These results suggest that migrating SVZ-derived neuroblasts acquire GABAergic identity prior to reaching their final location in the olfactory bulb.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Glutamato Descarboxilase/genética , Interneurônios/metabolismo , Bulbo Olfatório/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Movimento Celular , Dopamina/metabolismo , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interneurônios/citologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Bulbo Olfatório/citologia , Regiões Promotoras Genéticas , Células-Tronco/citologia , Transgenes
13.
J Comp Neurol ; 519(17): 3532-48, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21800304

RESUMO

Development of the cerebellum involves the coordinated proliferation, differentiation, maturation, and integration of cells from multiple neuronal and glial lineages. In rodent models, much of this occurs in the early postnatal period. However, our understanding of the molecular mechanisms that regulate this phase of cerebellar development remains incomplete. Here, we address the role of the transcription factor nuclear factor one X (NFIX), in postnatal development of the cerebellum. NFIX is expressed by progenitor cells within the external granular layer and by cerebellar granule neurons within the internal granule layer. Using NFIX⁻/⁻ mice, we demonstrate that the development of cerebellar granule neurons and Purkinje cells within the postnatal cerebellum is delayed in the absence of this transcription factor. Furthermore, the differentiation of mature glia within the cerebellum, such as Bergmann glia, is also significantly delayed in the absence of NFIX. Collectively, the expression pattern of NFIX, coupled with the delays in the differentiation of multiple cell populations of the developing cerebellum in NFIX⁻/⁻ mice, suggest a central role for NFIX in the regulation of cerebellar development, highlighting the importance of this gene for the maturation of this key structure.


Assuntos
Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Fatores de Transcrição NFI/fisiologia , Neurogênese/fisiologia , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Grânulos Citoplasmáticos/química , Grânulos Citoplasmáticos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Cereb Cortex ; 19 Suppl 1: i11-21, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19357391

RESUMO

Pioneer axons from the cingulate cortex initiate corpus callosum (CC) development, yet nothing is known about the molecules that regulate their guidance. We demonstrate that neuropilin 1 (Npn1) plays an integral role in the development of the CC. Npn1 is localized to axons of cingulate neurons as they cross the midline, and multiple class 3 semaphorins (Semas) are expressed around the developing CC, implicating these guidance molecules in the regulation of Npn1-expressing axons emanating from the cingulate cortex. Furthermore, axons from the cingulate cortex display guidance errors in Npn1(Sema-) mice, a knockin mouse line in which Npn1 is unable to bind Semas. Analysis of mice deficient in the transcription factor Emx2 demonstrated that the cingulate cortex of these mice was significantly reduced in comparison to wild-type controls at E17 and that the CC was absent in rostral sections. Expression of Npn1 was absent in rostral sections of Emx2 mutants, suggesting that Npn1-expressing cingulate pioneers are required for CC formation. These data highlight a central role for Npn1 in the development of projections from the cingulate cortex and further illustrate the importance of these pioneer axons in the formation of the CC.


Assuntos
Axônios/fisiologia , Corpo Caloso/embriologia , Corpo Caloso/metabolismo , Giro do Cíngulo/fisiologia , Neuropilina-1/metabolismo , Transdução de Sinais/fisiologia , Animais , Corpo Caloso/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais
15.
J Comp Neurol ; 513(1): 98-112, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19107796

RESUMO

Transcription factors of the Nuclear Factor I (Nfi) family are important for the development of specific neuronal and glial populations in the nervous system. One such population, the neurons of the basilar pontine nuclei, expresses high levels of Nfi proteins, and the pontine nuclei are greatly reduced in mice lacking a functional Nfib gene. Pontine neurons, along with other precerebellar neurons that populate the hindbrain, arise from precursors in the lower rhombic lip and migrate anteroventrally to reach their final location. Using immunohistochemistry, we find that NFI-B expression is specific for mossy fiber populations of the precerebellar system. Analysis of the Nfib(-/-) hindbrain indicates that the development of the basilar pontine nuclei is delayed, with pontine neurons migrating 1-2 days later than in control animals, and that significantly fewer pontine neurons are produced. While the mossy fiber nuclei of the caudal medulla do form, they also exhibit a developmental delay. Nfia and Nfix null mice exhibit no apparent pontine phenotype, implying specificity in the action of NFI family members. Collectively, these data demonstrate that Nfib plays an important role in the generation of precerebellar mossy fiber neurons, and may do so at least in part by regulating neurogenesis.


Assuntos
Córtex Cerebelar , Fatores de Transcrição NFI/metabolismo , Vias Neurais , Ponte , Animais , Córtex Cerebelar/anormalidades , Córtex Cerebelar/anatomia & histologia , Córtex Cerebelar/embriologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFI/genética , Vias Neurais/anormalidades , Vias Neurais/anatomia & histologia , Vias Neurais/embriologia , Neurônios/citologia , Neurônios/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Fenótipo , Ponte/anormalidades , Ponte/anatomia & histologia , Ponte/embriologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Rombencéfalo/anormalidades , Rombencéfalo/anatomia & histologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo
16.
BMC Dev Biol ; 8: 52, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18477394

RESUMO

BACKGROUND: The Nuclear Factor I (NFI) multi-gene family encodes site-specific transcription factors essential for the development of a number of organ systems. We showed previously that Nfia-deficient mice exhibit agenesis of the corpus callosum and other forebrain defects; Nfib-deficient mice have defects in lung maturation and show callosal agenesis and forebrain defects resembling those seen in Nfia-deficient animals, while Nfic-deficient mice have defects in tooth root formation. Recently the Nfix gene has been disrupted and these studies indicated that there were largely uncharacterized defects in brain and skeletal development in Nfix-deficient mice. RESULTS: Here we show that disruption of Nfix by Cre-recombinase mediated excision of the 2nd exon results in defects in brain development that differ from those seen in Nfia and Nfib KO mice. In particular, complete callosal agenesis is not seen in Nfix-/- mice but rather there appears to be an overabundance of aberrant Pax6- and doublecortin-positive cells in the lateral ventricles of Nfix-/- mice, increased brain weight, expansion of the cingulate cortex and entire brain along the dorsal ventral axis, and aberrant formation of the hippocampus. On standard lab chow Nfix-/- animals show a decreased growth rate from ~P8 to P14, lose weight from ~P14 to P22 and die at ~P22. If their food is supplemented with a soft dough chow from P10, Nfix-/- animals show a lag in weight gain from P8 to P20 but then increase their growth rate. A fraction of the animals survive to adulthood and are fertile. The weight loss correlates with delayed eye and ear canal opening and suggests a delay in the development of several epithelial structures in Nfix-/- animals. CONCLUSION: These data show that Nfix is essential for normal brain development and may be required for neural stem cell homeostasis. The delays seen in eye and ear opening and the brain morphology defects appear independent of the nutritional deprivation, as rescue of perinatal lethality with soft dough does not eliminate these defects.


Assuntos
Encéfalo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento , Fatores de Transcrição NFI/genética , Animais , Western Blotting , Córtex Cerebral/crescimento & desenvolvimento , Ventrículos Cerebrais/crescimento & desenvolvimento , Embrião de Mamíferos , Células-Tronco Embrionárias , Feminino , Marcação de Genes , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Reação em Cadeia da Polimerase
17.
J Comp Neurol ; 508(3): 385-401, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18335562

RESUMO

Three members of the Nuclear Factor I (Nfi) gene family of transcription factors; Nfia, Nfib, and Nfix are highly expressed in the developing mouse brain. Nfia and Nfib knockout mice display profound defects in the development of midline glial populations and the development of forebrain commissures (das Neves et al. [1999] Proc Natl Acad Sci U S A 96:11946-11951; Shu et al. [2003] J Neurosci 23:203-212; Steele-Perkins et al. [2005] Mol Cell Biol 25:685-698). These findings suggest that Nfi genes may regulate the substrate over which the commissural axons grow to reach targets in the contralateral hemisphere. However, these genes are also expressed in the cerebral cortex and, thus, it is important to assess whether this expression correlates with a cell-autonomous role in cortical development. Here we detail the protein expression of NFIA and NFIB during embryonic and postnatal mouse forebrain development. We find that both NFIA and NFIB are expressed in the deep cortical layers and subplate prenatally and display dynamic expression patterns postnatally. Both genes are also highly expressed in the developing hippocampus and in the diencephalon. We also find that principally neither NFIA nor NFIB are expressed in callosally projecting neurons postnatally, emphasizing the role for midline glial cell populations in commissure formation. However, a large proportion of laterally projecting neurons express both NFIA and NFIB, indicating a possible cell-autonomous role for these transcription factors in corticospinal neuron development. Collectively, these data suggest that, in addition to regulating the formation of axon guidance substrates, these genes also have cell-autonomous roles in cortical development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Transcrição NFI/metabolismo , Prosencéfalo , Animais , Animais Recém-Nascidos , Células Cultivadas , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFI/genética , Vias Neurais/citologia , Vias Neurais/embriologia , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo
18.
Mol Cell Neurosci ; 37(4): 719-30, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18272390

RESUMO

Axonal projections from the retina to the brain are regulated by molecules including the Slit family of ligands [Thompson, H., Barker, D., Camand, O., Erskine, L., 2006a. Slits contribute to the guidance of retinal ganglion cell axons in the mammalian optic tract. Dev. Biol. 296, 476-484, Thompson, H., Camand, O., Barker, D., Erskine, L., 2006b. Slit proteins regulate distinct aspects of retinal ganglion cell axon guidance within dorsal and ventral retina. J. Neurosci. 26, 8082-8091]. However, the roles of Slit receptors in mammals, (termed Robos), have not been investigated in visual system development. Here we examined Robo1 and 2 mutant mice and found that Robos regulate the correct targeting of retinal ganglion cell (RGC) axons along the entire visual projection. We noted aberrant projections of RGC axons into the cerebral cortex, an area not normally targeted by RGC axons. The optic chiasm was expanded along the rostro-caudal axis (similar to Slit mutant mice, Plump, A.S., Erskine, L., Sabatier, C., Brose, K., Epstein, C.J., Goodman, C.S., Mason, C.A., Tessier-Lavigne, M., 2002. Slit1 and Slit2 cooperate to prevent premature midline crossing of retinal axons in the mouse visual system. Neuron 33, 219-232), with ectopic crossing points, and some axons projecting caudally toward the corticospinal tract. Further, we found that axons exuberantly projected into the diencephalon. These defects were more pronounced in Robo2 than Robo1 knockout animals, implicating Robo2 as the predominant Robo receptor in visual system development.


Assuntos
Axônios/fisiologia , Encéfalo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores Imunológicos/fisiologia , Células Ganglionares da Retina/fisiologia , Vias Visuais/citologia , Vias Visuais/fisiologia , Animais , Encéfalo/citologia , Encéfalo/embriologia , Movimento Celular/genética , Feminino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Gravidez , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Células Ganglionares da Retina/citologia , Vias Visuais/embriologia , Proteínas Roundabout
19.
J Neurosci ; 27(39): 10345-9, 2007 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-17898206

RESUMO

In many cases of callosal dysgenesis in both human patients and mouse models, misguided fibers from the cortex form abnormal bilateral, barrel-shaped structures known as Probst bundles. Because little is known about how axons are arranged within these anomalous fiber bundles, understanding this arrangement may provide structural and molecular insights into how axons behave when they are misguided in vivo. Previous studies described these bundles as longitudinal swirls of axons that fail to cross the midline (Ozaki et al., 1987). However, recent studies on human acallosal patients using diffusion tensor magnetic resonance imaging (DTMRI) technology suggest that axons project in an anteroposterior direction within the Probst bundle (Lee et al., 2004; Tovar-Moll et al., 2007). This led us to ask the question, is DTMRI an accurate method for analyzing axonal tracts in regions of high axon overlap and disorganization, or is our current perception of axon arrangement within these bundles inaccurate? Using DTMRI, immunohistochemistry, and carbocyanine dye tract-tracing studies, we analyzed the Probst bundles in both Netrin1 and deleted in colorectal cancer (DCC) mutant mice. Our findings indicate that DTMRI can accurately demonstrate fiber tract orientation and morphology where axons are in ordered arrays such as in the dorsal part of the bundle. In ventral areas, where the axons are disorganized, no coordinated diffusion is apparent via DTMRI. In these regions, a higher-resolution approach such as tract tracing is required. We conclude that in DCC and Netrin1 mutant mice, guidance mechanisms remain in the dorsal part of the tract but are lost ventrally.


Assuntos
Agenesia do Corpo Caloso , Axônios/fisiologia , Imagem de Difusão por Ressonância Magnética , Malformações do Sistema Nervoso/fisiopatologia , Animais , Receptor DCC , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos , Fatores de Crescimento Neural/genética , Netrina-1 , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética
20.
Dev Neurosci ; 29(1-2): 143-58, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17148957

RESUMO

Hippocampal interneurons arise in the ventral forebrain and migrate dorsally in response to cues, including hepatocyte growth factor/scatter factor which signals via its receptor MET. Examination of the hippocampus in adult mice in which MET had been inactivated in the embryonic proliferative zones showed an increase in parvalbumin-expressing cells in the dentate gyrus, but a loss of these cells in the CA3 region. An overall loss of calretinin-expressing cells was seen throughout the hippocampus. A similar CA3 deficit of parvalbumin and calretinin cells was observed when MET was eliminated only in postmitotic cells. These data suggest that MET is required for the proper hippocampal development, and embryonic perturbations lead to long-term anatomical defects with possible learning and memory dysfunction.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Hipocampo/anormalidades , Hipocampo/metabolismo , Interneurônios/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Animais , Apoptose/genética , Calbindina 2 , Contagem de Células , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/citologia , Interneurônios/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/genética , Vias Neurais/anormalidades , Vias Neurais/citologia , Vias Neurais/metabolismo , Parvalbuminas/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Transdução de Sinais/genética , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...