Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34948008

RESUMO

Recent studies have begun to reveal surprising levels of cell diversity in the human brain, both in adults and during development. Distinctive cellular phenotypes point to complex molecular profiles, cellular hierarchies and signaling pathways in neural stem cells, progenitor cells, neuronal and glial cells. Several recent reports have suggested that neural stem and progenitor cell types found in the developing and adult brain share several properties and phenotypes with cells from brain primary tumors, such as gliomas. This transcriptomic crosstalk may help us to better understand the cell hierarchies and signaling pathways in both gliomas and the normal brain, and, by clarifying the phenotypes of cells at the origin of the tumor, to therapeutically address their most relevant signaling pathways.


Assuntos
Neoplasias Encefálicas/genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Glioma/genética , Células-Tronco Neurais/química , Neoplasias Encefálicas/patologia , Comunicação Celular , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/patologia , Fenótipo , Transdução de Sinais , Telencéfalo/química , Telencéfalo/citologia , Telencéfalo/patologia
2.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-33525421

RESUMO

The considerable post-traumatic brain recovery in fishes makes them a useful model for studying the mechanisms that provide reparative neurogenesis, which is poorly represented in mammals. After a mechanical injury to the telencephalon in adult fish, lost neurons are actively replaced due to the proliferative activity of neuroepithelial cells and radial glia in the neurogenic periventricular zone. However, it is not enough clear which signaling mechanisms are involved in the activation of adult neural stem cells (aNSC) after the injury (reactive proliferation) and in the production of new neurons (regenerative neurogenesis) from progenitor cells (NPC). In juvenile Pacific salmon, the predominant type of NSCs in the telencephalon are neuroepithelial cells corresponding to embryonic NSCs. Expression of glutamine synthetase (GS), a NSC molecular marker, was detected in the neuroepithelial cells of the pallium and subpallium of juvenile chum salmon, Oncorhynchus keta. At 3 days after a traumatic brain injury (TBI) in juvenile chum salmon, the GS expression was detected in the radial glia corresponding to aNSC in the pallium and subpallium. The maximum density of distribution of GS+ radial glia was found in the dorsal pallial region. Hydrogen sulfide (H2S) is a proneurogenic factor that reduces oxidative stress and excitotoxicity effects, along with the increased GS production in the brain cells of juvenile chum salmon. In the fish brain, H2S producing by cystathionine ß-synthase in neurogenic zones may be involved in maintaining the microenvironment that provides optimal conditions for the functioning of neurogenic niches during constitutive neurogenesis. After injury, H2S can determine cell survivability, providing a neuroprotective effect in the area of injury and reducing the process of glutamate excitotoxicity, acting as a signaling molecule involved in changing the neurogenic environment, which leads to the reactivation of neurogenic niches and cell regeneration programs. The results of studies on the control of the expression of regulatory Sonic Hedgehog genes (Shh) and the transcription factors Paired Box2 (Pax2) regulated by them are still insufficient. A comparative analysis of Pax2 expression in the telencephalon of intact chum salmon showed the presence of constitutive patterns of Pax2 expression in neurogenic areas and non-neurogenic parenchymal zones of the pallium and subpallium. After mechanical injury, the patterns of Pax2 expression changed, and the amount of Pax2+ decreased (p < 0.05) in lateral (Dl), medial (Dm) zones of the pallium, and the lateral zone (Vl) of the subpallium compared to the control. We believe that the decrease in the expression of Pax2 may be caused by the inhibitory effect of the Pax6 transcription factor, whose expression in the juvenile salmon brain increases upon injury.


Assuntos
Lesões Encefálicas/genética , Regeneração do Cérebro/genética , Cistationina beta-Sintase/genética , Proteínas de Peixes/genética , Glutamato-Amônia Ligase/genética , Fator de Transcrição PAX2/genética , Telencéfalo/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Diferenciação Celular , Proliferação de Células , Cistationina beta-Sintase/metabolismo , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Glutamato-Amônia Ligase/metabolismo , Ácido Glutâmico/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Sulfeto de Hidrogênio/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Neurogênese/genética , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Oncorhynchus keta , Fator de Transcrição PAX2/metabolismo , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , Telencéfalo/lesões , Telencéfalo/patologia
3.
Cell Death Dis ; 12(2): 151, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542214

RESUMO

Reactive oxygen species (ROS) stress has been demonstrated as potentially critical for induction and maintenance of cellular senescence, and been considered as a contributing factor in aging and in various neurological disorders including Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). In response to low-level ROS stress, the expression of Δ133p53, a human p53 isoform, is upregulated to promote cell survival and protect cells from senescence by enhancing the expression of antioxidant genes. In normal conditions, the basal expression of Δ133p53 prevents human fibroblasts, T lymphocytes, and astrocytes from replicative senescence. It has been also found that brain tissues from AD and ALS patients showed decreased Δ133p53 expression. However, it is uncharacterized if Δ133p53 plays a role in brain aging. Here, we report that zebrafish Δ113p53, an ortholog of human Δ133p53, mainly expressed in some of the radial glial cells along the telencephalon ventricular zone in a full-length p53-dependent manner. EDU-labeling and cell lineage tracing showed that Δ113p53-positive cells underwent cell proliferation to contribute to the neuron renewal process. Importantly, Δ113p53M/M mutant telencephalon possessed less proliferation cells and more senescent cells compared to wild-type (WT) zebrafish telencephalon since 9-months old, which was associated with decreased antioxidant genes expression and increased level of ROS in the mutant telencephalon. More interestingly, unlike the mutant fish at 5-months old with cognition ability, Δ113p53M/M zebrafish, but not WT zebrafish, lost their learning and memory ability at 19-months old. The results demonstrate that Δ113p53 protects the brain from aging by its antioxidant function. Our finding provides evidence at the organism level to show that depletion of Δ113p53/Δ133p53 may result in long-term ROS stress, and finally lead to age-related diseases, such as AD and ALS in humans.


Assuntos
Envelhecimento/metabolismo , Proliferação de Células , Senescência Celular , Estresse Oxidativo , Telencéfalo/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Fatores Etários , Envelhecimento/genética , Envelhecimento/patologia , Animais , Animais Geneticamente Modificados , Antioxidantes/metabolismo , Linhagem da Célula , Mutação com Perda de Função , Neurogênese , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Isoformas de Proteínas , Espécies Reativas de Oxigênio/metabolismo , Telencéfalo/patologia , Proteína Supressora de Tumor p53/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
4.
Nucleic Acids Res ; 47(1): 168-183, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30329130

RESUMO

Cortical development is controlled by transcriptional programs, which are orchestrated by transcription factors. Yet, stable inheritance of spatio-temporal activity of factors influencing cell fate and localization in different layers is only partly understood. Here we find that deletion of Dot1l in the murine telencephalon leads to cortical layering defects, indicating DOT1L activity and chromatin methylation at H3K79 impact on the cell cycle, and influence transcriptional programs conferring upper layer identity in early progenitors. Specifically, DOT1L prevents premature differentiation by increasing expression of genes that regulate asymmetric cell division (Vangl2, Cenpj). Loss of DOT1L results in reduced numbers of progenitors expressing genes including SoxB1 gene family members. Loss of DOT1L also leads to altered cortical distribution of deep layer neurons that express either TBR1, CTIP2 or SOX5, and less activation of transcriptional programs that are characteristic for upper layer neurons (Satb2, Pou3f3, Cux2, SoxC family members). Data from three different mouse models suggest that DOT1L balances transcriptional programs necessary for proper neuronal composition and distribution in the six cortical layers. Furthermore, because loss of DOT1L in the pre-neurogenic phase of development impairs specifically generation of SATB2-expressing upper layer neurons, our data suggest that DOT1L primes upper layer identity in cortical progenitors.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz/genética , Metiltransferases/genética , Neurogênese/genética , Neurônios/metabolismo , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Divisão Celular/genética , Proliferação de Células/genética , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Cromatina/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase , Metilação , Camundongos , Neurônios/patologia , Proteínas Repressoras/genética , Fatores de Transcrição SOXD/genética , Proteínas com Domínio T , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/metabolismo , Telencéfalo/patologia , Proteínas Supressoras de Tumor/genética
5.
Cell Rep ; 21(13): 3754-3766, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29281825

RESUMO

Focal cortical dysplasia (FCD) and hemimegalencephaly (HME) are epileptogenic neurodevelopmental malformations caused by mutations in mTOR pathway genes. Deep sequencing of these genes in FCD/HME brain tissue identified an etiology in 27 of 66 cases (41%). Radiographically indistinguishable lesions are caused by somatic activating mutations in AKT3, MTOR, and PIK3CA and germline loss-of-function mutations in DEPDC5, NPRL2, and TSC1/2, including TSC2 mutations in isolated HME demonstrating a "two-hit" model. Mutations in the same gene cause a disease continuum from FCD to HME to bilateral brain overgrowth, reflecting the progenitor cell and developmental time when the mutation occurred. Single-cell sequencing demonstrated mTOR activation in neurons in all lesions. Conditional Pik3ca activation in the mouse cortex showed that mTOR activation in excitatory neurons and glia, but not interneurons, is sufficient for abnormal cortical overgrowth. These data suggest that mTOR activation in dorsal telencephalic progenitors, in some cases specifically the excitatory neuron lineage, causes cortical dysplasia.


Assuntos
Malformações do Desenvolvimento Cortical/genética , Mutação/genética , Transdução de Sinais , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Telencéfalo/patologia , Animais , Linhagem da Célula , Classe I de Fosfatidilinositol 3-Quinases/genética , Hemimegalencefalia/genética , Hemimegalencefalia/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/patologia
6.
Dis Model Mech ; 10(1): 15-28, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27935819

RESUMO

Somatic mutations activating MAPK and PI3K signalling play a pivotal role in both tumours and brain developmental disorders. We developed a zebrafish model of brain tumours based on somatic expression of oncogenes that activate MAPK and PI3K signalling in neural progenitor cells and found that HRASV12 was the most effective in inducing both heterotopia and invasive tumours. Tumours, but not heterotopias, require persistent activation of phospho (p)-ERK and express a gene signature similar to the mesenchymal glioblastoma subtype, with a strong YAP component. Application of an eight-gene signature to human brain tumours establishes that YAP activation distinguishes between mesenchymal glioblastoma and low grade glioma in a wide The Cancer Genome Atlas (TCGA) sample set including gliomas and glioblastomas (GBMs). This suggests that the activation of YAP might be an important event in brain tumour development, promoting malignant versus benign brain lesions. Indeed, co-expression of dominant-active YAP (YAPS5A) and HRASV12 abolishes the development of heterotopias and leads to the sole development of aggressive tumours. Thus, we have developed a model proving that neurodevelopmental disorders and brain tumours might originate from the same activation of oncogenes through somatic mutations, and established that YAP activation is a hallmark of malignant brain tumours.


Assuntos
Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Aminoacil-tRNA Sintetases/genética , Animais , Neoplasias Encefálicas/genética , Carcinogênese/genética , Carcinogênese/patologia , Proliferação de Células , Sobrevivência Celular , Células Clonais , Modelos Animais de Doenças , Elementos Facilitadores Genéticos/genética , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Genes ras , Glioblastoma/genética , Glioblastoma/patologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Mesoderma/patologia , Células-Tronco Neurais/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Telencéfalo/patologia , Proteínas de Sinalização YAP , Proteínas de Peixe-Zebra/genética
7.
J Neuropathol Exp Neurol ; 74(7): 653-71, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26079447

RESUMO

Fetal-onset hydrocephalus affects 1 to 3 per 1,000 live births. It is not only a disorder of cerebrospinal fluid dynamics but also a brain disorder that corrective surgery does not ameliorate. We hypothesized that cell junction abnormalities of neural stem cells (NSCs) lead to the inseparable phenomena of fetal-onset hydrocephalus and abnormal neurogenesis. We used bromodeoxyuridine labeling, immunocytochemistry, electron microscopy, and cell culture to study the telencephalon of hydrocephalic HTx rats and correlated our findings with those in human hydrocephalic and nonhydrocephalic human fetal brains (n = 12 each). Our results suggest that abnormal expression of the intercellular junction proteins N-cadherin and connexin-43 in NSC leads to 1) disruption of the ventricular and subventricular zones, loss of NSCs and neural progenitor cells; and 2) abnormalities in neurogenesis such as periventricular heterotopias and abnormal neuroblast migration. In HTx rats, the disrupted NSC and progenitor cells are shed into the cerebrospinal fluid and can be grown into neurospheres that display intercellular junction abnormalities similar to those of NSC of the disrupted ventricular zone; nevertheless, they maintain their potential for differentiating into neurons and glia. These NSCs can be used to investigate cellular and molecular mechanisms underlying this condition, thereby opening the avenue for stem cell therapy.


Assuntos
Hidrocefalia/patologia , Junções Intercelulares/patologia , Células-Tronco Neurais/patologia , Neurogênese/fisiologia , Obstrução do Fluxo Ventricular Externo/patologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular , Movimento Celular , Células Cultivadas , Embrião de Mamíferos , Feminino , Feto , Idade Gestacional , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Junções Intercelulares/ultraestrutura , Masculino , Microscopia Eletrônica , Células-Tronco Neurais/ultraestrutura , Ratos , Telencéfalo/embriologia , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/patologia , Telencéfalo/ultraestrutura
8.
Hum Mol Genet ; 24(9): 2578-93, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25631876

RESUMO

Primary cilia are complex subcellular structures that play key roles during embryogenesis by controlling the cellular response to several signaling pathways. Defects in the function and/or structure of primary cilia underlie a large number of human syndromes collectively referred to as ciliopathies. Often, ciliopathies are associated with mental retardation (MR) and malformation of the corpus callosum. However, the possibility of defects in other forebrain axon tracts, which could contribute to the cognitive disorders of these patients, has not been explored. Here, we investigate the formation of the corticothalamic/thalamocortical tracts in mice mutant for Rfx3, which regulates the expression of many genes involved in ciliogenesis and cilia function. Using DiI axon tracing and immunohistochemistry experiments, we show that some Rfx3(-/-) corticothalamic axons abnormally migrate toward the pial surface of the ventral telencephalon (VT). Some thalamocortical axons (TCAs) also fail to leave the diencephalon or abnormally project toward the amygdala. Moreover, the Rfx3(-/-) VT displays heterotopias containing attractive guidance cues and expressing the guidance molecules Slit1 and Netrin1. Finally, the abnormal projection of TCAs toward the amygdala is also present in mice carrying a mutation in the Inpp5e gene, which is mutated in Joubert Syndrome and which controls cilia signaling and stability. The presence of identical thalamocortical malformations in two independent ciliary mutants indicates a novel role for primary cilia in the formation of the corticothalamic/thalamocortical tracts by establishing the correct cellular environment necessary for its development.


Assuntos
Padronização Corporal/genética , Córtex Cerebral/metabolismo , Proteínas de Ligação a DNA/genética , Telencéfalo/metabolismo , Tálamo/metabolismo , Fatores de Transcrição/genética , Animais , Embrião de Mamíferos , Homozigoto , Imuno-Histoquímica , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais , Neurônios/metabolismo , Monoéster Fosfórico Hidrolases/genética , Fatores de Transcrição de Fator Regulador X , Telencéfalo/embriologia , Telencéfalo/patologia , Tálamo/embriologia , Tálamo/patologia , Proteína Gli3 com Dedos de Zinco
9.
J Neurosci ; 34(32): 10475-87, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100583

RESUMO

Radial glial cells (RGCs) in the ventricular neuroepithelium of the dorsal telencephalon are the progenitor cells for neocortical projection neurons and astrocytes. Here we show that the adherens junction proteins afadin and CDH2 are critical for the control of cell proliferation in the dorsal telencephalon and for the formation of its normal laminar structure. Inactivation of afadin or CDH2 in the dorsal telencephalon leads to a phenotype resembling subcortical band heterotopia, also known as "double cortex," a brain malformation in which heterotopic gray matter is interposed between zones of white matter. Adherens junctions between RGCs are disrupted in the mutants, progenitor cells are widely dispersed throughout the developing neocortex, and their proliferation is dramatically increased. Major subtypes of neocortical projection neurons are generated, but their integration into cell layers is disrupted. Our findings suggest that defects in adherens junctions components in mice massively affects progenitor cell proliferation and leads to a double cortex-like phenotype.


Assuntos
Caderinas/deficiência , Proliferação de Células , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/patologia , Proteínas dos Microfilamentos/deficiência , Telencéfalo/patologia , Fatores Etários , Animais , Caderinas/genética , Proteínas do Domínio Duplacortina , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/fisiologia , Telencéfalo/anormalidades , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Hum Mol Genet ; 23(23): 6177-90, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990151

RESUMO

Simultaneous generation of neural cells and that of the nutrient-supplying vasculature during brain development is called neurovascular coupling. We report on a transgenic mouse with impaired transforming growth factor ß (TGFß)-signalling in forebrain-derived neural cells using a Foxg1-cre knock-in to drive the conditional knock-out of the Tgfbr2. Although the expression of FOXG1 is assigned to neural progenitors and neurons of the telencephalon, Foxg1(cre/+);Tgfbr2(flox/flox) (Tgfbr2-cKO) mutants displayed intracerebral haemorrhage. Blood vessels exhibited an atypical, clustered appearance were less in number and displayed reduced branching. Vascular endothelial growth factor (VEGF) A, insulin-like growth factor (IGF) 1, IGF2, TGFß, inhibitor of DNA binding (ID) 1, thrombospondin (THBS) 2, and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) 1 were altered in either expression levels or tissue distribution. Accordingly, human umbilical vein endothelial cells (HUVEC) displayed branching defects after stimulation with conditioned medium (CM) that was derived from primary neural cultures of the ventral and dorsal telencephalon of Tgfbr2-cKO. Supplementing CM of Tgfbr2-cKO with VEGFA rescued these defects, but application of TGFß aggravated them. HUVEC showed reduced migration towards CM of mutants compared with controls. Supplementing the CM with growth factors VEGFA, fibroblast growth factor (FGF) 2 and IGF1 partially restored HUVEC migration. In contrast, TGFß supplementation further impaired migration of HUVEC. We observed differences along the dorso-ventral axis of the telencephalon with regard to the impact of these factors on the phenotype. Together these data establish a TGFBR2-dependent molecular crosstalk between neural and endothelial cells during brain vessel development. These findings will be useful to further elucidate neurovascular interaction in general and to understand pathologies of the blood vessel system such as intracerebral haemorrhages, hereditary haemorrhagic telangiectasia, Alzheimers disease, cerebral amyloid angiopathy or tumour biology.


Assuntos
Encéfalo/metabolismo , Neovascularização Fisiológica , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Movimento Celular , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Meios de Cultivo Condicionados , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurônios/patologia , Pericitos/metabolismo , Pericitos/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Via Secretória , Telencéfalo/irrigação sanguínea , Telencéfalo/metabolismo , Telencéfalo/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Genome Res ; 24(4): 592-603, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24642863

RESUMO

Genome-wide association studies (GWAS) identified the MEIS1 locus for Restless Legs Syndrome (RLS), but causal single nucleotide polymorphisms (SNPs) and their functional relevance remain unknown. This locus contains a large number of highly conserved noncoding regions (HCNRs) potentially functioning as cis-regulatory modules. We analyzed these HCNRs for allele-dependent enhancer activity in zebrafish and mice and found that the risk allele of the lead SNP rs12469063 reduces enhancer activity in the Meis1 expression domain of the murine embryonic ganglionic eminences (GE). CREB1 binds this enhancer and rs12469063 affects its binding in vitro. In addition, MEIS1 target genes suggest a role in the specification of neuronal progenitors in the GE, and heterozygous Meis1-deficient mice exhibit hyperactivity, resembling the RLS phenotype. Thus, in vivo and in vitro analysis of a common SNP with small effect size showed allele-dependent function in the prospective basal ganglia representing the first neurodevelopmental region implicated in RLS.


Assuntos
Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Neoplasias/genética , Síndrome das Pernas Inquietas/genética , Telencéfalo/crescimento & desenvolvimento , Alelos , Animais , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Íntrons , Camundongos , Proteína Meis1 , Polimorfismo de Nucleotídeo Único , Telencéfalo/patologia
12.
PLoS One ; 9(2): e86025, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24516524

RESUMO

Embryonic neural crest cells contribute to the development of the craniofacial mesenchyme, forebrain meninges and perivascular cells. In this study, we investigated the function of ß-catenin signaling in neural crest cells abutting the dorsal forebrain during development. In the absence of ß-catenin signaling, neural crest cells failed to expand in the interhemispheric region and produced ectopic smooth muscle cells instead of generating dermal and calvarial mesenchyme. In contrast, constitutive expression of stabilized ß-catenin in neural crest cells increased the number of mesenchymal lineage precursors suggesting that ß-catenin signaling is necessary for the expansion of neural crest-derived mesenchymal cells. Interestingly, the loss of neural crest-derived mesenchymal stem cells (MSCs) leads to failure of telencephalic midline invagination and causes ventricular system defects. This study shows that ß-catenin signaling is required for the switch of neural crest cells to MSCs and mediates the expansion of MSCs to drive the formation of mesenchymal structures of the head. Furthermore, loss of these structures causes striking defects in forebrain morphogenesis.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Telencéfalo/crescimento & desenvolvimento , Via de Sinalização Wnt , Animais , Linhagem da Célula , Proliferação de Células , Deleção de Genes , Meninges/citologia , Camundongos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Telencéfalo/patologia , beta Catenina/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-24114617

RESUMO

This study assessed spatial memory and orientation strategies in Chiloscyllium griseum. In the presence of visual landmarks, six sharks were trained in a fixed turn response. Group 1 started from two possible compartments approaching two goal locations, while group 2 started from and approached only one location, respectively. The learning criterion was reached within 9 ± 5.29 (group 1) and 8.3 ± 3.51 sessions (group 2). Transfer tests revealed that sharks had applied a direction strategy, possibly in combination with some form of place learning. Without visual cues, sharks relied solely on the former. To identify the underlying neural substrate(s), telencephalic were lesioned and performance compared before and after surgery. Ablation of the dorsal and medial pallia only had an effect on one shark (group 1), indicating that the acquisition and retention of previously gained knowledge were unaffected in the remaining four individuals. Nonetheless, the shark re-learned the task. In summary, C. griseum can utilize fixed turn responses to navigate to a goal; there is also some evidence for the use of external visual landmarks while orienting. Probably, strategies can be used alone or in combination. Neither the dorsal nor medial pallium seems to be responsible for the acquisition and processing of egocentric information.


Assuntos
Atividade Motora/fisiologia , Orientação/fisiologia , Tubarões/fisiologia , Telencéfalo/fisiologia , Animais , Sinais (Psicologia) , Feminino , Objetivos , Masculino , Aprendizagem em Labirinto/fisiologia , Testes Neuropsicológicos , Estimulação Luminosa , Prática Psicológica , Percepção Espacial/fisiologia , Análise e Desempenho de Tarefas , Telencéfalo/patologia , Transferência de Experiência/fisiologia , Percepção Visual/fisiologia
14.
Sci Transl Med ; 5(197): 197ra104, 2013 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-23926202

RESUMO

Consumption of certain substances during pregnancy can interfere with brain development, leading to deleterious long-term neurological and cognitive impairments in offspring. To test whether modulators of adenosine receptors affect neural development, we exposed mouse dams to a subtype-selective adenosine type 2A receptor (A2AR) antagonist or to caffeine, a naturally occurring adenosine receptor antagonist, during pregnancy and lactation. We observed delayed migration and insertion of γ-aminobutyric acid (GABA) neurons into the hippocampal circuitry during the first postnatal week in offspring of dams treated with the A2AR antagonist or caffeine. This was associated with increased neuronal network excitability and increased susceptibility to seizures in response to a seizure-inducing agent. Adult offspring of mouse dams exposed to A2AR antagonists during pregnancy and lactation displayed loss of hippocampal GABA neurons and some cognitive deficits. These results demonstrate that exposure to A2AR antagonists including caffeine during pregnancy and lactation in rodents may have adverse effects on the neural development of their offspring.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Cafeína/farmacologia , Feto/efeitos dos fármacos , Feto/embriologia , Antagonistas de Receptores Purinérgicos P1/farmacologia , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Movimento Celular/efeitos dos fármacos , Transtornos Cognitivos/patologia , Suscetibilidade a Doenças , Feminino , Feto/patologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Glutamatos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Haplorrinos/embriologia , Hipocampo/efeitos dos fármacos , Hipocampo/embriologia , Hipocampo/patologia , Camundongos , Rede Nervosa/efeitos dos fármacos , Gravidez , Ratos , Receptores A2 de Adenosina/metabolismo , Convulsões/embriologia , Convulsões/patologia , Telencéfalo/efeitos dos fármacos , Telencéfalo/embriologia , Telencéfalo/patologia
15.
J Comp Neurol ; 521(13): 3099-115, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23787922

RESUMO

The zebrafish has recently become a source of new data on the mechanisms of neural stem cell (NSC) maintenance and ongoing neurogenesis in adult brains. In this vertebrate, neurogenesis occurs at high levels in all ventricular regions of the brain, and brain injuries recover successfully, owing to the recruitment of radial glia, which function as NSCs. This new vertebrate model of adult neurogenesis is thus advancing our knowledge of the molecular cues in use for the activation of NSCs and fate of their progeny. Because the regenerative potential of somatic stem cells generally weakens with increasing age, it is important to assess the extent to which zebrafish NSC potential decreases or remains unaltered with age. We found that neurogenesis in the ventricular zone, in the olfactory bulb, and in a newly identified parenchymal zone of the telencephalon indeed declines as the fish ages and that oligodendrogenesis also declines. In the ventricular zone, the radial glial cell population remains largely unaltered morphologically but enters less frequently into the cell cycle and hence produces fewer neuroblasts. The neuroblasts themselves do not change their behavior with age and produce the same number of postmitotic neurons. Thus, decreased neurogenesis in the physiologically aging zebrafish brain is correlated with an increasing quiescence of radial glia. After injuries, radial glia in aged brains are reactivated, and the percentage of cell cycle entry is increased in the radial glia population. However, this reaction is far less pronounced than in younger animals, pointing to irreversible changes in aging zebrafish radial glia.


Assuntos
Envelhecimento , Lesões Encefálicas/patologia , Regeneração Nervosa/fisiologia , Células-Tronco Neurais/fisiologia , Neuroglia/fisiologia , Telencéfalo/patologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Bromodesoxiuridina/metabolismo , Contagem de Células , Modelos Animais de Doenças , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Proteína Semelhante a ELAV 3 , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
16.
Nat Neurosci ; 16(6): 692-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23644485

RESUMO

Impaired GABA-mediated neurotransmission has been implicated in many neurologic diseases, including epilepsy, intellectual disability and psychiatric disorders. We found that inhibitory neuron transplantation into the hippocampus of adult mice with confirmed epilepsy at the time of grafting markedly reduced the occurrence of electrographic seizures and restored behavioral deficits in spatial learning, hyperactivity and the aggressive response to handling. In the recipient brain, GABA progenitors migrated up to 1,500 µm from the injection site, expressed genes and proteins characteristic for interneurons, differentiated into functional inhibitory neurons and received excitatory synaptic input. In contrast with hippocampus, cell grafts into basolateral amygdala rescued the hyperactivity deficit, but did not alter seizure activity or other abnormal behaviors. Our results highlight a critical role for interneurons in epilepsy and suggest that interneuron cell transplantation is a powerful approach to halting seizures and rescuing accompanying deficits in severely epileptic mice.


Assuntos
Comportamento Animal , Epilepsia/fisiopatologia , Epilepsia/terapia , Neurônios GABAérgicos/transplante , Interneurônios/transplante , Transplante de Células-Tronco/métodos , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/psicologia , Epilepsia do Lobo Temporal/fisiopatologia , Epilepsia do Lobo Temporal/psicologia , Epilepsia do Lobo Temporal/terapia , Neurônios GABAérgicos/patologia , Camundongos , Telencéfalo/embriologia , Telencéfalo/patologia , Telencéfalo/transplante
17.
Morfologiia ; 142(4): 19-24, 2012.
Artigo em Russo | MEDLINE | ID: mdl-23236886

RESUMO

Nestin and Musashil (Msi-1) proteins are most often used for labeling of neural stem cells and progenitor cells in vivo, however it remains unclear if these markers really label the same cells. As a result of the study of structural characteristics and localization of nestin- and Msil-expressing cells it was found that these proteins were detected in non-identical cell populations in the brain of intact 15 rats. We failed to find cell groups demonstrating a coexpression of nestin and Msi-1. However, after ischemic lesion of the brain, which was caused in 38 rats by an endovascular occlusion of the left medial cerebral artery for 30 min with the following reperfusion for 48 hours, both markers were detected in cells of subventricular zone and in ependymocytes. These results indicate the changes in cytochemical patterns of the candidate stem cells.


Assuntos
Antígenos de Diferenciação/biossíntese , Isquemia Encefálica/metabolismo , Regulação da Expressão Gênica , Proteínas de Filamentos Intermediários/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Células-Tronco Neurais/metabolismo , Proteínas de Ligação a RNA/biossíntese , Telencéfalo/metabolismo , Animais , Isquemia Encefálica/patologia , Masculino , Nestina , Células-Tronco Neurais/patologia , Ratos , Ratos Wistar , Telencéfalo/patologia
18.
Am J Pathol ; 181(2): 626-41, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22658483

RESUMO

Mutations in chromosome-helicase-DNA-binding protein 7 (CHD7) are identified as the main cause for CHARGE syndrome (coloboma, heart anomaly, choanal atresia, retardation, genital and ear anomalies). Most patients (55% to 85%) with CHARGE syndrome display developmental defects in the central nervous system (CNS), of which pathology and molecular mechanisms remain unclear. In this study, we report a novel mutant mouse strain carrying a nonsense mutation, COA1, in exon4 of Chd7 gene. Chd7(COA1/+) mice phenocopied human CHARGE syndrome and displayed developmental defects in the telencephalic midline, including dilated third and lateral ventricles, reduced cerebral cortex, and corpus callosum crossing failure. Programed cell death in the telencephalic midline zone of Chd7(COA1/+) embryos was impaired, consistent with the incomplete telencephalic medial invagination in Chd7(COA1/+) embryos. Interestingly, expression of Bmp4, a signal well known to induce forebrain midline cell fate and apoptosis, was down-regulated and also expanded in the forebrain of Chd7(COA1/+) embryos. Furthermore, in vitro studies suggested that CHD7 may directly regulate Bmp4 expression by binding with an enhancer element downstream of the Bmp4 locus. These studies provide novel insight into pathogenesis of CNS anomalies in CHARGE syndrome.


Assuntos
Proteína Morfogenética Óssea 4/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Mutação/genética , Telencéfalo/embriologia , Telencéfalo/patologia , Animais , Apoptose/genética , Comportamento Animal , Proteína Morfogenética Óssea 4/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula/genética , Proliferação de Células , Cruzamentos Genéticos , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Elementos Facilitadores Genéticos/genética , Anormalidades do Olho/enzimologia , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Feminino , Genoma/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Neuroglia/patologia , Ligação Proteica , Transdução de Sinais/genética , Telencéfalo/metabolismo
19.
Cell Stem Cell ; 9(5): 447-62, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-22056141

RESUMO

Tuberous Sclerosis Complex (TSC) is a multisystem genetic disorder characterized by hamartomatous neurological lesions that exhibit abnormal cell proliferation and differentiation. Hyperactivation of mTOR pathway by mutations in either the Tsc1 or Tsc2 gene underlies TSC pathogenesis, but involvement of specific neural cell populations in the formation of TSC-associated neurological lesions remains unclear. We deleted Tsc1 in Emx1-expressing embryonic telencephalic neural stem cells (NSCs) and found that mutant mice faithfully recapitulated TSC neuropathological lesions, such as cortical lamination defects and subependymal nodules (SENs). These alterations were caused by enhanced generation of SVZ neural progeny, followed by their premature differentiation and impaired maturation during both embryonic and postnatal development. Notably, mTORC1-dependent Akt inhibition and STAT3 activation were involved in the reduced self-renewal and earlier neuronal and astroglial differentiation of mutant NSCs. Thus, finely tuned mTOR activation in embryonic NSCs may be critical to prevent development of TSC-associated brain lesions.


Assuntos
Células-Tronco Embrionárias/enzimologia , Células-Tronco Neurais/enzimologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/metabolismo , Esclerose Tuberosa/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Movimento Celular , Proliferação de Células , Ventrículos Cerebrais/metabolismo , Ventrículos Cerebrais/patologia , Ventrículos Cerebrais/ultraestrutura , Desenvolvimento Embrionário , Epilepsia/complicações , Epilepsia/patologia , Inativação Gênica , Marcação de Genes , Megalencefalia/complicações , Megalencefalia/patologia , Camundongos , Mutação/genética , Células Neuroepiteliais/metabolismo , Células Neuroepiteliais/patologia , Neurônios/metabolismo , Neurônios/patologia , Telencéfalo/crescimento & desenvolvimento , Telencéfalo/metabolismo , Telencéfalo/patologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA