Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Glia ; 70(2): 321-336, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34687571

RESUMO

The N-myc downstream regulated gene family member 1 (NDRG1) is a gene whose mutation results in peripheral neuropathy with central manifestations. While most of previous studies characterized NDRG1 role in Schwann cells, the detection of central nervous system symptoms and the identification of NDRG1 as a gene silenced in the white matter of multiple sclerosis brains raise the question regarding its role in oligodendrocytes. Here, we show that NDRG1 is enriched in oligodendrocytes and myelin preparations, and we characterize its expression using a novel reporter mouse (TgNdrg1-EGFP). We report NDRG1 expression during developmental myelination and during remyelination after cuprizone-induced demyelination of the adult corpus callosum. The transcriptome of Ndrg1-EGFP+ cells further supports the identification of late myelinating oligodendrocytes, characterized by expression of genes regulating lipid metabolism and bioenergetics. We also generate a lineage specific conditional knockout (Olig1cre/+ ;Ndrg1fl/fl ) line to study its function. Null mice develop normally, and despite similar numbers of progenitor cells as wild type, they have fewer mature oligodendrocytes and lower levels of myelin proteins than controls, thereby suggesting NDRG1 as important for the maintenance of late myelinating oligodendrocytes. In addition, when control and Ndrg1 null mice are subject to cuprizone-induced demyelination, we observe a higher degree of demyelination in the mutants. Together these data identify NDRG1 as an important molecule for adult myelinating oligodendrocytes, whose decreased levels in the normal appearing white matter of human MS brains may result in greater susceptibility of myelin to damage.


Assuntos
Esclerose Múltipla , Bainha de Mielina , Animais , Cuprizona/toxicidade , Família , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo
2.
J Neurosci ; 35(1): 352-65, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25568127

RESUMO

In many cell types, differentiation requires an interplay between extrinsic signals and transcriptional changes mediated by repressive and activating histone modifications. Oligodendrocyte progenitors (OPCs) are electrically responsive cells receiving synaptic input. The differentiation of these cells into myelinating oligodendrocytes is characterized by temporal waves of gene repression followed by activation of myelin genes and progressive decline of electrical responsiveness. In this study, we used chromatin isolated from rat OPCs and immature oligodendrocytes, to characterize the genome-wide distribution of the repressive histone marks, H3K9me3 and H3K27me3, during differentiation. Although both marks were present at the OPC stage, only H3K9me3 marks (but not H3K27me3) were found to be increased during differentiation, at genes related to neuronal lineage and regulation of membrane excitability. Consistent with these findings, the levels and activity of H3K9 methyltransferases (H3K9 HMT), but not H3K27 HMT, increased more prominently upon exposure to oligodendrocyte differentiating stimuli and were detected in stage-specific repressive protein complexes containing the transcription factors SOX10 or YY1. Silencing H3K9 HMT, but not H3K27 HMT, impaired oligodendrocyte differentiation and functionally altered the response of oligodendrocytes to electrical stimulation. Together, these results identify repressive H3K9 methylation as critical for gene repression during oligodendrocyte differentiation.


Assuntos
Diferenciação Celular/fisiologia , Cromatina/metabolismo , Histonas/metabolismo , Neurogênese/fisiologia , Oligodendroglia/metabolismo , Animais , Imunoprecipitação da Cromatina/métodos , Feminino , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Gravidez , Ratos , Ratos Sprague-Dawley
3.
J Neurosci ; 34(4): 1481-93, 2014 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24453336

RESUMO

Cell cycle exit is an obligatory step for the differentiation of oligodendrocyte progenitor cells (OPCs) into myelinating cells. A key regulator of the transition from proliferation to quiescence is the E2F/Rb pathway, whose activity is highly regulated in physiological conditions and deregulated in tumors. In this paper we report a lineage-specific decline of nuclear E2F1 during differentiation of rodent OPC into oligodendrocytes (OLs) in developing white matter tracts and in cultured cells. Using chromatin immunoprecipitation (ChIP) and deep-sequencing in mouse and rat OPCs, we identified cell cycle genes (i.e., Cdc2) and chromatin components (i.e., Hmgn1, Hmgn2), including those modulating DNA methylation (i.e., Uhrf1), as E2F1 targets. Binding of E2F1 to chromatin on the gene targets was validated and their expression assessed in developing white matter tracts and cultured OPCs. Increased expression of E2F1 gene targets was also detected in mouse gliomas (that were induced by retroviral transformation of OPCs) compared with normal brain. Together, these data identify E2F1 as a key transcription factor modulating the expression of chromatin components in OPC during the transition from proliferation to differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Cromatina/fisiologia , Fator de Transcrição E2F1/metabolismo , Genes cdc/fisiologia , Neurogênese/fisiologia , Oligodendroglia/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Imunoprecipitação da Cromatina , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Células-Tronco/metabolismo
4.
Cell Mol Life Sci ; 70(16): 2887-98, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23124271

RESUMO

The mammalian target of rapamycin (mTOR) pathway is a central controller of growth and homeostasis, and, as such, is implicated in disease states where growth is deregulated, namely cancer, metabolic diseases, and hamartoma syndromes like tuberous sclerosis complex (TSC). Accordingly, mTOR is also a pivotal regulator of the homeostasis of several distinct stem cell pools in which it finely tunes the balance between stem cell self-renewal and differentiation. mTOR hyperactivation in neural stem cells (NSCs) has been etiologically linked to the development of TSC-associated neurological lesions, such as brain hamartomas and benign tumors. Animal models generated by deletion of mTOR upstream regulators in different types of NSCs reproduce faithfully some of the TSC neurological alterations. Thus, mTOR dysregulation in NSCs seems to be responsible for the derangement of their homeostasis, thus leading to TSC development. Here we review recent advances in the molecular dissection of the mTOR cascade, its involvement in the maintenance of stem cell compartments, and in particular the implications of mTOR hyperactivation in NSCs in vivo and in vitro.


Assuntos
Células-Tronco Neurais/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Síndrome do Hamartoma Múltiplo/metabolismo , Síndrome do Hamartoma Múltiplo/patologia , Humanos , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neurais/patologia , Transdução de Sinais
5.
Cell Mol Life Sci ; 70(8): 1439-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23212600

RESUMO

The receptor for activated C-kinase 1 (RACK1) is a conserved structural protein of 40S ribosomes. Strikingly, deletion of RACK1 in yeast homolog Asc1 is not lethal. Mammalian RACK1 also interacts with many nonribosomal proteins, hinting at several extraribosomal functions. A knockout mouse for RACK1 has not previously been described. We produced the first RACK1 mutant mouse, in which both alleles of RACK1 gene are defective in RACK1 expression (ΔF/ΔF), in a pure C57 Black/6 background. In a sample of 287 pups, we observed no ΔF/ΔF mice (72 expected). Dissection and genotyping of embryos at various stages showed that lethality occurs at gastrulation. Heterozygotes (ΔF/+) have skin pigmentation defects with a white belly spot and hypopigmented tail and paws. ΔF/+ have a transient growth deficit (shown by measuring pup size at P11). The pigmentation deficit is partly reverted by p53 deletion, whereas the lethality is not. ΔF/+ livers have mild accumulation of inactive 80S ribosomal subunits by polysomal profile analysis. In ΔF/+ fibroblasts, protein synthesis response to extracellular and pharmacological stimuli is reduced. These results highlight the role of RACK1 as a ribosomal protein converging signaling to the translational apparatus.


Assuntos
Neuropeptídeos/genética , Pigmentação , Biossíntese de Proteínas , Animais , Células Cultivadas , Perda do Embrião/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/ultraestrutura , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/metabolismo , Fenótipo , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Quinase C Ativada , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Seizure ; 27: 30-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25891924

RESUMO

PURPOSE: The development of new drugs for the treatment of epilepsy is a major challenge for modern neurology and its first steps demand basic research. Preclinical studies on animal models of epilepsy are mainly based on the analysis of brain electrical activity to detect seizures, when they are not just limited to behavioral tests like the Racine scale. METHODS: In the present review, we discuss the importance of using time-locked video and EEG recordings (Video-EEG) coupled with behavioral tests as tools to monitor and analyze the effects of anti-epileptic drugs in pre-clinical research. Particularly, we focus on the utility of a multimodal approach based on EEG/behavioral analysis to study the beneficial effects of chronic rapamycin treatment as a potential anti-epileptogenic therapy for a broad spectrum of epilepsy, including both genetic (as in tuberous sclerosis complex) and acquired diseases. RESULTS: Changes and synchronization of neuronal activity of different areas have been correlated with specific behavior in both physiological and pathological conditions. In the epileptic brain, during a seizure there is an abnormal activation of many cells all at once, altering different networks. CONCLUSION: A multimodal approach based on video, EEG analysis and behavioral tests would be the best option in preclinical studies of epilepsy.


Assuntos
Anticonvulsivantes/uso terapêutico , Eletroencefalografia , Epilepsia/tratamento farmacológico , Avaliação de Resultados em Cuidados de Saúde , Sirolimo/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Epilepsia/patologia , Humanos , Reprodutibilidade dos Testes , Gravação de Videoteipe
7.
Neuropharmacology ; 67: 1-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23159330

RESUMO

Tuberous Sclerosis Complex (TSC) is a multisystem genetic disorder caused by mutation in either Tsc1 or Tsc2 genes that leads to the hyper activation of the mTOR pathway, a key signalling pathway for synaptic plasticity. TSC is characterized by benign tumors arising in different organs and severe neuropsychiatric symptoms, such as epilepsy, intellectual disability, autism, anxiety and depressive behaviour. Rapamycin is a potent inhibitor of mTOR and its efficacy in treating epilepsy and neurological symptoms remains elusive. In a mouse model in which Tsc1 has been deleted in embryonic telencephalic neural stem cells, we analyzed anxiety- and depression-like behaviour by elevated-plus maze (EPM), open-field test (OFT), forced-swim test (FST) and tail-suspension test (TST), after chronic administration of rapamycin. In addition, spectral analysis of background EEG was performed. Rapamycin-treated mutant mice displayed a reduction in anxiety- and depression-like phenotype, as shown by the EPM/OFT and FST, respectively. These results were inline with EEG power spectra outcomes. The same effects of rapamycin were observed in wild-type mice. Notably, in heterozygous animals we did not observe any EEG and/or behavioural variation after rapamycin treatment. Together these results suggest that both TSC1 deletion and chronic rapamycin treatment might have a role in modulating behaviour and brain activity, and point out to the potential usefulness of background EEG analysis in tracking brain dysfunction in parallel with behavioural testing.


Assuntos
Comportamento Animal/efeitos dos fármacos , Eletroencefalografia , Sirolimo/administração & dosagem , Esclerose Tuberosa/tratamento farmacológico , Esclerose Tuberosa/fisiopatologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Eletroencefalografia/métodos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Resultado do Tratamento , Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/deficiência
8.
Dis Model Mech ; 6(5): 1185-97, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23744272

RESUMO

Tuberous sclerosis complex (TSC) is a dominantly inherited disease with high penetrance and morbidity, and is caused by mutations in either of two genes, TSC1 or TSC2. Most affected individuals display severe neurological manifestations - such as intractable epilepsy, mental retardation and autism - that are intimately associated with peculiar CNS lesions known as cortical tubers (CTs). The existence of a significant genotype-phenotype correlation in individuals bearing mutations in either TSC1 or TSC2 is highly controversial. Similar to observations in humans, mouse modeling has suggested that a more severe phenotype is associated with mutation in Tsc2 rather than in Tsc1. However, in these mutant mice, deletion of either gene was achieved in differentiated astrocytes. Here, we report that loss of Tsc1 expression in undifferentiated radial glia cells (RGCs) early during development yields the same phenotype detected upon deletion of Tsc2 in the same cells. Indeed, the same aberrations in cortical cytoarchitecture, hippocampal disturbances and spontaneous epilepsy that have been detected in RGC-targeted Tsc2 mutants were observed in RGC-targeted Tsc1 mutant mice. Remarkably, thorough characterization of RGC-targeted Tsc1 mutants also highlighted subventricular zone (SVZ) disturbances as well as STAT3-dependent and -independent developmental-stage-specific defects in the differentiation potential of ex-vivo-derived embryonic and postnatal neural stem cells (NSCs). As such, deletion of either Tsc1 or Tsc2 induces mostly overlapping phenotypic neuropathological features when performed early during neurogenesis, thus suggesting that the timing of mTOR activation is a key event in proper neural development.


Assuntos
Modelos Animais de Doenças , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/enzimologia , Esclerose Tuberosa/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Epilepsia/metabolismo , Epilepsia/patologia , Inativação Gênica/efeitos dos fármacos , Longevidade/efeitos dos fármacos , Megalencefalia/metabolismo , Megalencefalia/patologia , Camundongos , Mutagênese/genética , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fator de Transcrição STAT3/metabolismo , Sirolimo/farmacologia , Fatores de Tempo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
9.
Cell Rep ; 1(2): 141-54, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22832163

RESUMO

Expression of the mannose receptor (MRC1/CD206) identifies macrophage subtypes, such as alternatively activated macrophages (AAMs) and M2-polarized tumor-associated macrophages (TAMs), which are endowed with tissue-remodeling, proangiogenic, and protumoral activity. However, the significance of MRC1 expression for TAM's protumoral activity is unclear. Here, we describe and characterize miR-511-3p, an intronic microRNA (miRNA) encoded by both mouse and human MRC1 genes. By using sensitive miRNA reporter vectors, we demonstrate robust expression and bioactivity of miR-511-3p in MRC1(+) AAMs and TAMs. Unexpectedly, enforced expression of miR-511-3p tuned down the protumoral gene signature of MRC1(+) TAMs and inhibited tumor growth. Our findings suggest that transcriptional activation of Mrc1 in TAMs evokes a genetic program orchestrated by miR-511-3p, which limits rather than enhances their protumoral functions. Besides uncovering a role for MRC1 as gatekeeper of TAM's protumoral genetic programs, these observations suggest that endogenous miRNAs may operate to establish thresholds for inflammatory cell activation in tumors.


Assuntos
Macrófagos/metabolismo , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/patologia , Animais , Pareamento de Bases/genética , Sequência de Bases , Células da Medula Óssea/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Hematopoese/genética , Humanos , Imunofenotipagem , Lectinas Tipo C/genética , Receptor de Manose , Lectinas de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/química , MicroRNAs/genética , Dados de Sequência Molecular , Neoplasias/irrigação sanguínea , Conformação de Ácido Nucleico , Precursores de RNA/química , Precursores de RNA/genética , Precursores de RNA/metabolismo , Receptores de Superfície Celular/genética , Quinases Associadas a rho/metabolismo
10.
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
11.
Cell ; 124(6): 1183-95, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16499958

RESUMO

The interaction between ubiquitinated proteins and intracellular proteins harboring ubiquitin binding domains (UBDs) is critical to a multitude of cellular processes. Here, we report that Rabex-5, a guanine nucleotide exchange factor for Rab5, binds to Ub through two independent UBDs. These UBDs determine a number of properties of Rabex-5, including its coupled monoubiquitination and interaction in vivo with ubiquitinated EGFRs. Structural and biochemical characterization of the UBDs of Rabex-5 revealed that one of them (MIU, motif interacting with ubiquitin) binds to Ub with modes superimposable to those of the UIM (ubiquitin-interacting motif):Ub interaction, although in the opposite orientation. The other UBD, RUZ (Rabex-5 ubiquitin binding zinc finger) binds to a surface of Ub centered on Asp58(Ub) and distinct from the "canonical" Ile44(Ub)-based surface. The two binding surfaces allow Ub to interact simultaneously with different UBDs, thus opening new perspectives in Ub-mediated signaling.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Ubiquitina/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Fator de Crescimento Epidérmico/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa