Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genes Cells ; 20(9): 706-19, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26243725

RESUMO

In addition to its well-established role during immune system function, NF-κB regulates cell survival and synaptic plasticity in the mature nervous system. Here, we show that during mouse brain development, NF-κB activity is present in the neocortical ventricular and subventricular zones (VZ and SVZ), where it regulates proliferative pool maintenance. Activation of NF-κB signaling, by expression of p65 or an activated form of the IκB kinase complex subunit IKK2, inhibited neuronal differentiation and promoted retention of progenitors in the VZ and SVZ. In contrast, blockade of the pathway with dominant negative forms of IKK2 and IκBα promoted neuronal differentiation both in vivo and in vitro. Furthermore, by modulating both the NF-κB and Notch pathways, we show that in the absence of canonical Notch activity, after knockdown of the pathway effector CBF1, NF-κB signaling promoted Tbr2 expression and intermediate neural progenitor fate. Interestingly, however, activation of NF-κB in vivo, with canonical Notch signaling intact, promoted expression of the radial glial marker Pax6. This work identifies NF-κB signaling as a regulator of neocortical neurogenesis and suggests that the pathway plays roles in both the VZ and SVZ.


Assuntos
NF-kappa B/metabolismo , Neocórtex/crescimento & desenvolvimento , Neurogênese , Transdução de Sinais , Animais , Proteínas do Olho/metabolismo , Feminino , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Neocórtex/citologia , Células-Tronco Neurais/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo
2.
Hippocampus ; 25(5): 670-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25515406

RESUMO

Activation of the Notch pathway in neurons is essential for learning and memory in various species from invertebrates to mammals. However, it remains unclear how Notch signaling regulates neuronal plasticity, and whether the transcriptional regulator and canonical pathway effector RBP-J plays a role. Here, we report that conditional disruption of RBP-J in the postnatal hippocampus leads to defects in long-term potentiation, long-term depression, and in learning and memory. Using gene expression profiling and chromatin immunoprecipitation, we identified two GABA transporters, GAT2 and BGT1, as putative Notch/RBP-J pathway targets, which may function downstream of RBP-J to limit the accumulation of GABA in the Schaffer collateral pathway. Our results reveal an essential role for canonical Notch/RBP-J signaling in hippocampal synaptic plasticity and suggest that role, at least in part, is mediated by the regulation of GABAergic signaling.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Hipocampo/fisiologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Plasticidade Neuronal/fisiologia , Receptores Notch/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/fisiologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Camundongos Knockout , Transdução de Sinais , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos , Ácido gama-Aminobutírico/metabolismo
4.
Cancer Cell ; 21(5): 626-641, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22624713

RESUMO

Basal-like breast cancers (BLBC) express a luminal progenitor gene signature. Notch receptor signaling promotes luminal cell fate specification in the mammary gland, while suppressing stem cell self-renewal. Here we show that deletion of Lfng, a sugar transferase that prevents Notch activation by Jagged ligands, enhances stem/progenitor cell proliferation. Mammary-specific deletion of Lfng induces basal-like and claudin-low tumors with accumulation of Notch intracellular domain fragments, increased expression of proliferation-associated Notch targets, amplification of the Met/Caveolin locus, and elevated Met and Igf-1R signaling. Human BL breast tumors, commonly associated with JAGGED expression, elevated MET signaling, and CAVEOLIN accumulation, express low levels of LFNG. Thus, reduced LFNG expression facilitates JAG/NOTCH luminal progenitor signaling and cooperates with MET/CAVEOLIN basal-type signaling to promote BLBC.


Assuntos
Neoplasias da Mama/enzimologia , Caveolinas/metabolismo , Transformação Celular Neoplásica/metabolismo , Glicosiltransferases/metabolismo , Glândulas Mamárias Animais/enzimologia , Neoplasias Mamárias Experimentais/enzimologia , Células-Tronco Neoplásicas/enzimologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Caveolinas/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Cultivadas , Claudinas/metabolismo , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Glicosiltransferases/deficiência , Glicosiltransferases/genética , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/transplante , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/transplante , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-met/genética , Receptor IGF Tipo 1/metabolismo , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais
5.
Dev Cell ; 22(4): 707-20, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22445366

RESUMO

Regulation of self-renewal and differentiation of neural stem cells is still poorly understood. Here we investigate the role of a developmentally expressed protein, Botch, which blocks Notch, in neocortical development. Downregulation of Botch in vivo leads to cellular retention in the ventricular and subventricular zones, whereas overexpression of Botch drives neural stem cells into the intermediate zone and cortical plate. In vitro neurosphere and differentiation assays indicate that Botch regulates neurogenesis by promoting neuronal differentiation. Botch prevents cell surface presentation of Notch by inhibiting the S1 furin-like cleavage of Notch, maintaining Notch in the immature full-length form. Understanding the function of Botch expands our knowledge regarding both the regulation of Notch signaling and the complex signaling mediating neuronal development.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular , Embrião de Mamíferos/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fármacos Neuroprotetores/metabolismo , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo , Animais , Western Blotting , Proteínas de Transporte/genética , Membrana Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Complexo de Golgi/metabolismo , Humanos , Imunoprecipitação , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética , Transdução de Sinais , gama-Glutamilciclotransferase
6.
Science ; 334(6063): 1706-10, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22116031

RESUMO

B-type lamins, the major components of the nuclear lamina, are believed to be essential for cell proliferation and survival. We found that mouse embryonic stem cells (ESCs) do not need any lamins for self-renewal and pluripotency. Although genome-wide lamin-B binding profiles correlate with reduced gene expression, such binding is not directly required for gene silencing in ESCs or trophectoderm cells. However, B-type lamins are required for proper organogenesis. Defects in spindle orientation in neural progenitor cells and migration of neurons probably cause brain disorganizations found in lamin-B null mice. Thus, our studies not only disprove several prevailing views of lamin-Bs but also establish a foundation for redefining the function of the nuclear lamina in the context of tissue building and homeostasis.


Assuntos
Células-Tronco Embrionárias/fisiologia , Lamina Tipo B/fisiologia , Organogênese , Animais , Tamanho Corporal , Encéfalo/citologia , Encéfalo/embriologia , Ciclo Celular , Diferenciação Celular , Movimento Celular , Células Cultivadas , Cromatina/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Masculino , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Neurônios/citologia , Lâmina Nuclear/fisiologia , Tamanho do Órgão , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Regiões Promotoras Genéticas , Fuso Acromático/fisiologia , Fuso Acromático/ultraestrutura , Transcrição Gênica , Trofoblastos/citologia
7.
Genes Dev ; 25(13): 1399-411, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21724832

RESUMO

Blood vessel networks are typically formed by angiogenesis, a process in which new vessels form by sprouting of endothelial cells from pre-existing vessels. This process is initiated by vascular endothelial growth factor (VEGF)-mediated tip cell selection and subsequent angiogenic sprouting. Surprisingly, we found that VEGF directly controls the expression of Plexin-D1, the receptor for the traditional repulsive axon guidance cue, semaphorin 3E (Sema3E). Sema3E-Plexin-D1 signaling then negatively regulates the activity of the VEGF-induced Delta-like 4 (Dll4)-Notch signaling pathway, which controls the cell fate decision between tip and stalk cells. Using the mouse retina as a model system, we show that Plexin-D1 is selectively expressed in endothelial cells at the front of actively sprouting blood vessels and its expression is tightly controlled by VEGF secreted by surrounding tissues. Therefore, although the Sema3E secreted by retinal neurons is evenly distributed throughout the retina, Sema3E-Plexin-D1 signaling is spatially controlled by VEGF through its regulation of Plexin-D1. Moreover, we show that gain and loss of function of Sema3E and Plexin-D1 disrupts normal Dll4 expression, Notch activity, and tip/stalk cell distribution in the retinal vasculature. Finally, the retinal vasculature of mice lacking sema3E or plexin-D1 has an uneven growing front, a less-branched vascular network, and abnormal distribution of dll4-positive cells. Lowering Notch activity in the mutant mice can reverse this defect, solidifying the observation that Dll4-Notch signaling is regulated by Sema3E-Plexin-D1 and is required for its function in vivo. Together, these data reveal a novel role of Sema3E-Plexin-D1 function in modulating angiogenesis via a VEGF-induced feedback mechanism.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Retroalimentação Fisiológica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Células Endoteliais/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neovascularização Fisiológica/genética , Proteínas do Tecido Nervoso , Fenótipo , Receptores Notch/metabolismo , Retina/citologia , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Semaforinas/genética , Semaforinas/metabolismo
8.
Neuron ; 69(5): 840-55, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21382546

RESUMO

The Notch pathway is prominent among those known to regulate neural development in vertebrates. Notch receptor activation can inhibit neurogenesis, maintain neural progenitor character, and in some contexts promote gliogenesis and drive binary fate choices. Recently, a wave of exciting studies has emerged, which has both solidified previously held assertions and expanded our understanding of Notch function during neurogenesis and in the adult brain. These studies have examined pathway regulators and interactions, as well as pathway dynamics, with respect to both gene expression and cell-cell signaling. Here, focusing primarily on vertebrates, we review the current literature on Notch signaling in the nervous system, and highlight numerous recent studies that have generated interesting and unexpected advances.


Assuntos
Diferenciação Celular/fisiologia , Sistema Nervoso/metabolismo , Neurônios/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais
9.
Neuron ; 69(3): 437-44, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21315255

RESUMO

Notch signaling in the nervous system has been most studied in the context of cell fate specification. However, numerous studies have suggested that Notch also regulates neuronal morphology, synaptic plasticity, learning, and memory. Here we show that Notch1 and its ligand Jagged1 are present at the synapse, and that Notch signaling in neurons occurs in response to synaptic activity. In addition, neuronal Notch signaling is positively regulated by Arc/Arg3.1, an activity-induced gene required for synaptic plasticity. In Arc/Arg3.1 mutant neurons, the proteolytic activation of Notch1 is disrupted both in vivo and in vitro. Conditional deletion of Notch1 in the postnatal hippocampus disrupted both long-term potentiation (LTP) and long-term depression (LTD), and led to deficits in learning and short-term memory. Thus, Notch signaling is dynamically regulated in response to neuronal activity, Arc/Arg3.1 is a context-dependent Notch regulator, and Notch1 is required for the synaptic plasticity that contributes to memory formation.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Hipocampo/fisiologia , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Plasticidade Neuronal/fisiologia , Receptor Notch1/fisiologia , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes Neurológicos , Neurônios/fisiologia , Sinapses/fisiologia
10.
Cancer Res ; 71(3): 1115-25, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21245095

RESUMO

Although Notch signaling has been widely implicated in neoplastic growth, direct evidence for in vivo initiation of neoplasia by the pathway in murine models has been limited to tumors of lymphoid, breast, and choroid plexus cells. To examine tumorigenic potential in the eye and brain, we injected retroviruses encoding activated forms of Notch1, Notch2, or Notch3 into embryonic mice. Interestingly, the majority of animals infected with active Notch3 developed proliferative lesions comprised of pigmented ocular choroid cells, retinal and optic nerve glia, and lens epithelium. Notch3-induced lesions in the choroid, retina, and optic nerve were capable of invading adjacent tissues, suggesting that they were malignant tumors. Although Notch3 activation induced choroidal tumors in up to 67% of eyes, Notch1 or Notch2 activation never resulted in such tumors. Active forms of Notch1 and Notch2 did generate a few small proliferative glial nodules in the retina and optic nerve, whereas Notch3 was 10-fold more efficient at generating growths, many of which were large invasive gliomas. Expression of active Notch1/Notch3 chimeric receptors implicated the RBPjk-association molecule and transactivation domains of Notch3 in generating choroidal and glial tumors, respectively. In contrast to our findings in the optic nerve and retina, introduction of active Notch receptors, including Notch3, into the brain never caused glial tumors. Our results highlight the differential ability of Notch receptor paralogs to initiate malignant tumor formation, and suggest that glial precursors of the optic nerve, but not the brain, are susceptible to transformation by Notch3.


Assuntos
Glioma/metabolismo , Receptores Notch/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Neoplasias da Coroide/metabolismo , Neoplasias da Coroide/patologia , Glioma/patologia , Humanos , Cristalino/patologia , Camundongos , Neoplasias do Nervo Óptico/metabolismo , Neoplasias do Nervo Óptico/patologia , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Receptor Notch3 , Proteínas Recombinantes de Fusão/metabolismo , Neoplasias da Retina/metabolismo , Neoplasias da Retina/patologia , Transdução de Sinais
11.
Circ Res ; 108(1): 51-9, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21106942

RESUMO

RATIONALE: Transgenic Notch reporter mice express enhanced green fluorescent protein in cells with C-promoter binding factor-1 response element transcriptional activity (CBF1-RE(x)4-EGFP), providing a unique and powerful tool for identifying and isolating "Notch-activated" progenitors. OBJECTIVE: We asked whether, as in other tissues of this mouse, EGFP localized and functionally tagged adult cardiac tissue progenitors, and, if so, whether this cell-based signal could serve as a quantitative and qualitative biosensor of the injury repair response of the heart. METHODS AND RESULTS: In addition to scattered endothelial and interstitial cells, Notch-activated (EGFP(+)) cells unexpectedly richly populated the adult epicardium. We used fluorescence-activated cell sorting to isolate EGFP(+) cells and excluded hematopoietic (CD45(+)) and endothelial (CD31(+)) subsets. We analyzed EGFP(+)/CD45⁻/CD31⁻ cells, a small (<2%) but distinct subpopulation, by gene expression profiling and functional analyses. We called this mixed cell pool, which had dual multipotent stromal cell and epicardial lineage signatures, Notch-activated epicardial-derived cells (NECs). Myocardial infarction and thoracic aortic banding amplified the NEC pool, increasing fibroblast differentiation. Validating the functional vitality of clonal NEC lines, serum growth factors triggered epithelial-mesenchymal transition and the immobilized Notch ligand Delta-like 1-activated downstream target genes. Moreover, cardiomyocyte coculture and engraftment in NOD-SCID (nonobese diabetic-severe combined immunodeficiency) mouse myocardium increased cardiac gene expression in NECs. CONCLUSIONS: A dynamic Notch injury response activates adult epicardium, producing a multipotent cell population that contributes to fibrosis repair.


Assuntos
Células-Tronco Multipotentes/metabolismo , Infarto do Miocárdio/metabolismo , Pericárdio/metabolismo , Receptores Notch/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Fibrose , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Antígenos Comuns de Leucócito , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/patologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Pericárdio/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas , Receptores Notch/genética
12.
Biochem Biophys Res Commun ; 404(1): 133-8, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21108929

RESUMO

Notch signaling pathway enhances neural stem cell characters and regulates cell fate decisions during neural development. Interestingly, besides Notch, other γ-secretase substrates such as APP, LRP2, and ErbB4 have also proven to have biological functions in neural development. We designed a unique experimental setting, combining gain-of- (expression of Notch intracellular domain, NICD) and loss-of-function (γ-secretase inhibition) methods, and were able to examine the function of Notch alone by excluding the activity of other γ-secretase substrates. Here, we show that the frequency and size of neurospheres generated from embryonic neural stem cells (NSCs) significantly decreased by 62.7% and 37.2%, respectively, in the presence of γ-secretase inhibitor even when NICD was expressed. Under the condition of differentiation, however, the γ-secretase inhibitor treatment did not influence the promotion of astrogenesis at the expense of neurogenesis by NICD. These results indicate that other γ-secretase substrate(s) along with Notch are important in the maintenance of the stemness of NSCs, but that Notch alone can sufficiently inhibit neurogenesis without the action of the other γ-secretase substrates during differentiation.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Receptor Notch1/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Carbamatos/farmacologia , Dipeptídeos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Receptor Notch1/genética , Ativação Transcricional
13.
Clin Cancer Res ; 16(24): 6060-70, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21169257

RESUMO

PURPOSE: Multiple developmental pathways including Notch, Hedgehog, and Wnt are active in malignant brain tumors such as medulloblastoma and glioblastoma (GBM). This raises the possibility that tumors might compensate for therapy directed against one pathway by upregulating a different one. We investigated whether brain tumors show resistance to therapies against Notch, and whether targeting multiple pathways simultaneously would kill brain tumor cells more effectively than monotherapy. EXPERIMENTAL DESIGN: We used GBM neurosphere lines to investigate the effects of a gamma-secretase inhibitor (MRK-003) on tumor growth, and chromatin immunoprecipitation to study the regulation of other genes by Notch targets. We also evaluated the effect of combined therapy with a Hedgehog inhibitor (cyclopamine) in GBM and medulloblastoma lines, and in primary human GBM cultures. RESULTS: GBM cells are at least partially resistant to long-term MRK-003 treatment, despite ongoing Notch pathway suppression, and show concomitant upregulation of Wnt and Hedgehog activity. The Notch target Hes1, a repressive transcription factor, bound the Gli1 first intron, and may inhibit its expression. Similar results were observed in a melanoma-derived cell line. Targeting Notch and Hedgehog simultaneously induced apoptosis, decreased cell growth, and inhibited colony-forming ability more dramatically than monotherapy. Low-passage neurospheres isolated from freshly resected human GBMs were also highly susceptible to coinhibition of the two pathways, indicating that targeting multiple developmental pathways can be more effective than monotherapy at eliminating GBM-derived cells. CONCLUSIONS: Notch may directly suppress Hedgehog via Hes1 mediated inhibition of Gli1 transcription, and targeting both pathways simultaneously may be more effective at eliminating GBMs cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Óxidos S-Cíclicos/administração & dosagem , Avaliação Pré-Clínica de Medicamentos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas Hedgehog/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Receptores Notch/metabolismo , Receptores Notch/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Tiadiazóis/administração & dosagem , Fatores de Transcrição HES-1 , Fatores de Transcrição/metabolismo , Células U937 , Alcaloides de Veratrum/administração & dosagem , Proteína GLI1 em Dedos de Zinco
14.
J Vis Exp ; (45)2010 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-21113114

RESUMO

In utero survival surgery in mice permits the molecular manipulation of gene expression during development. However, because the uterine wall is opaque during early embryogenesis, the ability to target specific parts of the embryo for microinjection is greatly limited. Fortunately, high-frequency ultrasound imaging permits the generation of images that can be used in real time to guide a microinjection needle into the embryonic region of interest. Here we describe the use of such imaging to guide the injection of retroviral vectors into the ventricular system of the mouse forebrain at embryonic day (E) 9.5. This method uses a laparotomy to permit access to the uterine horns, and a specially designed plate that permits host embryos to be bathed in saline while they are imaged and injected. Successful surgeries often result in most or all of the injected embryos surviving to any subsequent time point of interest (embryonically or postnatally). The principles described here can be used with slight modifications to perform injections into the amnionic fluid of E8.5 embryos (thereby permitting infection along the anterior posterior extent of the neural tube, which has not yet closed), or into the ventricular system of the brain at E10.5/11.5. Furthermore, at mid-neurogenic ages (~E13.5), ultrasound imaging can be used direct injection into specific brain regions for viral infection or cell transplantation. The use of ultrasound imaging to guide in utero injections in mice is a very powerful technique that permits the molecular and cellular manipulation of mouse embryos in ways that would otherwise be exceptionally difficult if not impossible.


Assuntos
Terapias Fetais/métodos , Microinjeções/métodos , Prosencéfalo/cirurgia , Ultrassonografia Pré-Natal/métodos , Animais , Vetores Genéticos/administração & dosagem , Camundongos , Prosencéfalo/embriologia , Retroviridae/genética
15.
Stroke ; 41(10 Suppl): S64-71, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20876509

RESUMO

BACKGROUND AND PURPOSE: Notch receptors (1-4) are membrane proteins that, on ligand stilumation, release their cytoplasmic domains to serve as transcription factors. Notch-2 promotes proliferation both during development and cancer, but its role in response to ischemic injury is less well understood. The purpose of this study was to understand whether Notch-2 is induced after neonatal stroke and to investigate its functional relevance. METHODS: P12 CD1 mice were subjected to permanent unilateral (right-sided) double ligation of the common carotid artery. RESULTS: Neonatal ischemia induces a progressive brain injury with prolonged apoptosis and Notch-2 up-regulation. Notch-2 expression was induced shortly after injury in hippocampal areas with elevated c-fos activation and increased cell death. Long-term induction of Notch-2 also occurred in CA1 and CA3 in and around areas of cell death, and had a distinct pattern of expression as compared to Notch-1. In vitro oxygen glucose deprivation treatment showed a similar increase in Notch-2 in apoptotic cells. In vitro gain of function experiments, using an active form of Notch-2, show that Notch-2 induction is neurotoxic to a comparable extent as oxygen glucose deprivation treatment. CONCLUSIONS: These results suggest that Notch-2 up-regulation after neonatal ischemia is detrimental to neuronal survival.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptor Notch2/metabolismo , Acidente Vascular Cerebral/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Apoptose , Western Blotting , Isquemia Encefálica/patologia , Contagem de Células , Hipocampo/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia de Fluorescência , Neurônios/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Acidente Vascular Cerebral/patologia , Fatores de Tempo , Regulação para Cima
16.
Nat Neurosci ; 13(5): 551-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20364144

RESUMO

Brain structure and size require precise division of neural stem cells (NSCs), which self-renew and generate intermediate neural progenitors (INPs) and neurons. The factors that regulate NSCs remain poorly understood, and mechanistic explanations of how aberrant NSC division causes the reduced brain size seen in microcephaly are lacking. Here we show that Magoh, a component of the exon junction complex (EJC) that binds RNA, controls mouse cerebral cortical size by regulating NSC division. Magoh haploinsufficiency causes microcephaly because of INP depletion and neuronal apoptosis. Defective mitosis underlies these phenotypes, as depletion of EJC components disrupts mitotic spindle orientation and integrity, chromosome number and genomic stability. In utero rescue experiments showed that a key function of Magoh is to control levels of the microcephaly-associated protein Lis1 during neurogenesis. Our results uncover requirements for the EJC in brain development, NSC maintenance and mitosis, thereby implicating this complex in the pathogenesis of microcephaly.


Assuntos
Encéfalo/patologia , Divisão Celular/genética , Microcefalia/patologia , Neurônios/patologia , Proteínas Nucleares/metabolismo , Células-Tronco/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Apoptose/genética , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Bromodesoxiuridina/metabolismo , Diferenciação Celular/genética , Análise Mutacional de DNA , Embrião de Mamíferos , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento/genética , Genótipo , Proteínas de Fluorescência Verde/genética , Células HeLa , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microcefalia/genética , Microcefalia/fisiopatologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Tamanho do Órgão/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Interferência de RNA/fisiologia , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transfecção
17.
J Neurosurg Spine ; 12(2): 117-21, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20121344

RESUMO

The relative rarity of spinal cord tumors has hampered the study of these uncommon nervous system malignancies. Consequently, the understanding of the fundamental biology and optimal treatment of spinal cord tumors is limited, and these cancers continue to inflict considerable morbidity and mortality in children and adults. As a first step to improving the outcome of patients affected with spinal cord tumors, the National Institutes of Health Office of Rare Diseases Research in cooperation with the National Cancer Institute and the National Institute of Neurological Disorders and Stroke convened a workshop to discuss the current status of research and clinical management of these tumors. The overall goal of this meeting was to initiate a process that would eventually translate fundamental basic science research into improved clinical care for this group of patients. Investigational priorities for each of these areas were established, and the opportunities for future multidisciplinary research collaborations were identified.


Assuntos
Pesquisa Biomédica , Neoplasias da Medula Espinal , Animais , Pesquisa Biomédica/tendências , Humanos , National Cancer Institute (U.S.) , National Institute of Neurological Disorders and Stroke (USA) , Doenças Raras/epidemiologia , Doenças Raras/patologia , Doenças Raras/fisiopatologia , Doenças Raras/terapia , Neoplasias da Medula Espinal/epidemiologia , Neoplasias da Medula Espinal/patologia , Neoplasias da Medula Espinal/fisiopatologia , Neoplasias da Medula Espinal/terapia , Estados Unidos
18.
Cell Stem Cell ; 5(5): 455-6, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19896433

RESUMO

During corticogenesis, neural stem cells switch from symmetric to asymmetric divisions to generate intermediate progenitor cells and neurons. Recently published in Cell, Siegenthaler et al. (2009) show that retinoic acid derived from the meninges is a key signal controlling this transition.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...