Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 1828, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758195

RESUMO

DNA sequences containing consecutive guanines organized in 4-interspaced tandem repeats can form stable single-stranded secondary structures, called G-quadruplexes (G4). Herein, we report that the Polycomb group protein BMI1 is enriched at heterochromatin regions containing putative G4 DNA sequences, and that G4 structures accumulate in cells with reduced BMI1 expression and/or relaxed chromatin, including sporadic Alzheimer's disease (AD) neurons. In AD neurons, G4 structures preferentially accumulate in lamina-associated domains, and this is rescued by re-establishing chromatin compaction. ChIP-seq analyses reveal that G4 peaks correspond to evolutionary conserved Long Interspersed Element-1 (L1) sequences predicted to be transcriptionally active. Hence, G4 structures co-localize with RNAPII, and inhibition of transcription can reverse the G4 phenotype without affecting chromatin's state, thus uncoupling both components. Intragenic G4 structures affecting splicing events are furthermore associated with reduced neuronal gene expression in AD. Active L1 sequences are thus at the origin of most G4 structures observed in human neurons.


Assuntos
Doença de Alzheimer/metabolismo , Eucromatina/metabolismo , Quadruplex G , Elementos Nucleotídeos Longos e Dispersos/genética , Neurogênese/genética , Neurônios/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Doença de Alzheimer/genética , Animais , Células Cultivadas , Sequenciamento de Cromatina por Imunoprecipitação , Eucromatina/genética , Feminino , Imunofluorescência , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Ontologia Genética , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 1/genética , Splicing de RNA/genética
2.
Neural Regen Res ; 16(10): 1958-1964, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33642366

RESUMO

Sporadic late-onset Alzheimer's disease (SLOAD) and familial early-onset Alzheimer's disease (FEOAD) associated with dominant mutations in APP, PSEN1 and PSEN2, are thought to represent a spectrum of the same disorder based on near identical behavioral and histopathological features. Hence, FEOAD transgenic mouse models have been used in past decades as a surrogate to study SLOAD pathogenic mechanisms and as the gold standard to validate drugs used in clinical trials. Unfortunately, such research has yielded little output in terms of therapeutics targeting the disease's development and progression. In this short review, we interrogate the widely accepted view of one, dimorphic disease through the prism of the Bmi1+/- mouse model and the distinct chromatin signatures observed between SLOAD and FEOAD brains.

3.
EMBO Mol Med ; 13(3): e12005, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33587337

RESUMO

Neovascularization contributes to multiple visual disorders including age-related macular degeneration (AMD) and retinopathy of prematurity. Current therapies for treating ocular angiogenesis are centered on the inhibition of vascular endothelial growth factor (VEGF). While clinically effective, some AMD patients are refractory or develop resistance to anti-VEGF therapies and concerns of increased risks of developing geographic atrophy following long-term treatment have been raised. Identification of alternative pathways to inhibit pathological angiogenesis is thus important. We have identified a novel inhibitor of angiogenesis, COCO, a member of the Cerberus-related DAN protein family. We demonstrate that COCO inhibits sprouting, migration and cellular proliferation of cultured endothelial cells. Intravitreal injections of COCO inhibited retinal vascularization during development and in models of retinopathy of prematurity. COCO equally abrogated angiogenesis in models of choroidal neovascularization. Mechanistically, COCO inhibited TGFß and BMP pathways and altered energy metabolism and redox balance of endothelial cells. Together, these data show that COCO is an inhibitor of retinal and choroidal angiogenesis, possibly representing a therapeutic option for the treatment of neovascular ocular diseases.


Assuntos
Neovascularização de Coroide , Cocos , Neovascularização de Coroide/tratamento farmacológico , Células Endoteliais , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Retina , Fator A de Crescimento do Endotélio Vascular
4.
Genes (Basel) ; 11(7)2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32708145

RESUMO

Late-onset sporadic Alzheimer's disease (LOAD) seems to contain a "hidden" component that cannot be explained by classical Mendelian genetics, with advanced aging being the strongest risk factor. More surprisingly, whole genome sequencing analyses of early-onset sporadic Alzheimer's disease cohorts also revealed that most patients do not present classical disease-associated variants or mutations. In this short review, we propose that BMI1 is possibly epigenetically silenced in LOAD. Reduced BMI1 expression is unique to LOAD compared to familial early-onset AD (EOAD) and other related neurodegenerative disorders; moreover, reduced expression of this single gene is sufficient to reproduce most LOAD pathologies in cellular and animal models. We also show the apparent amyloid and Tau-independent nature of this epigenetic alteration of BMI1 expression. Lastly, examples of the mechanisms underlying epigenetic dysregulation of other LOAD-related genes are also illustrated.


Assuntos
Doença de Alzheimer/genética , Complexo Repressor Polycomb 1/genética , Doença de Alzheimer/patologia , Animais , Epigênese Genética , Humanos , Mutação , Complexo Repressor Polycomb 1/metabolismo
5.
Stem Cell Reports ; 14(3): 357-373, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32160518

RESUMO

Ciliopathies are heterogeneous genetic diseases affecting primary cilium structure and function. Meckel-Gruber (MKS) and Bardet-Biedl (BBS) syndromes are severe ciliopathies characterized by skeletal and neurodevelopment anomalies, including polydactyly, cognitive impairment, and retinal degeneration. We describe the generation and molecular characterization of human induced pluripotent stem cell (iPSC)-derived retinal sheets (RSs) from controls, and MKS (TMEM67) and BBS (BBS10) cases. MKS and BBS RSs displayed significant common alterations in the expression of hundreds of developmental genes and members of the WNT and BMP pathways. Induction of crystallin molecular chaperones was prominent in MKS and BBS RSs suggesting a stress response to misfolded proteins. Unique to MKS photoreceptors was the presence of supernumerary centrioles and cilia, and aggregation of ciliary proteins. Unique to BBS photoreceptors was the accumulation of DNA damage and activation of the mitotic spindle checkpoint. This study reveals how combining cell reprogramming, organogenesis, and next-generation sequencing enables the elucidation of mechanisms involved in human ciliopathies.


Assuntos
Ciliopatias/genética , Citoesqueleto/patologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes Induzidas/patologia , Sistema Nervoso/crescimento & desenvolvimento , Retina/patologia , Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/patologia , Centríolos/metabolismo , Centríolos/ultraestrutura , Cílios/patologia , Cílios/ultraestrutura , Cristalinas/metabolismo , Instabilidade Genômica , Humanos , Morfogênese/genética , Células Fotorreceptoras de Vertebrados/patologia , Retina/ultraestrutura , Fuso Acromático , Síndrome
6.
NPJ Precis Oncol ; 4: 1, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31934644

RESUMO

Glioblastoma multiforme (GBM) is an incurable primary brain tumor containing a sub-population of cancer stem cells (CSCs). Polycomb Repressive Complex (PRC) proteins BMI1 and EZH2 are enriched in CSCs, promoting clonogenic growth and resistance to genotoxic therapies. We report here that when used at appropriate concentrations, pharmaceutical inhibitors of BMI1 could efficiently prevent GBM colony growth and CSC self-renewal in vitro and significantly extend lifespan in terminally ill tumor-bearing mice. Notably, molecular analyses revealed that the commonly used PTC596 molecule targeted both BMI1 and EZH2, possibly providing beneficial therapeutic effects in some contexts. On the other hand, treatment with PTC596 resulted in instant reactivation of EZH2 target genes and induction of a molecular program of epithelial-mesenchymal transition (EMT), possibly explaining the modified phenotype of some PTC596-treated tumors. Treatment with a related but more specific BMI1 inhibitor resulted in tumor regression and maintenance of cell identity. We conclude that inhibition of BMI1 alone is efficient at inducing GBM regression, and that dual inhibition of BMI1 and EZH2 using PTC596 may be also beneficial but only in specific contexts.

7.
Sci Rep ; 9(1): 594, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679733

RESUMO

Sporadic Alzheimer's disease (AD) is the most common cause of dementia. However, representative experimental models of AD have remained difficult to produce because of the disease's uncertain origin. The Polycomb group protein BMI1 regulates chromatin compaction and gene silencing. BMI1 expression is abundant in adult brain neurons but down-regulated in AD brains. We show here that mice lacking one allele of Bmi1 (Bmi1+/-) develop normally but present with age cognitive deficits and neurodegeneration sharing similarities with AD. Bmi1+/- mice also transgenic for the amyloid beta precursor protein died prematurely and present aggravated disease. Loss of heterochromatin and DNA damage response (DDR) at repetitive DNA sequences were predominant in Bmi1+/- mouse neurons and inhibition of the DDR mitigated the amyloid and Tau phenotype. Heterochromatin anomalies and DDR at repetitive DNA sequences were also found in AD brains. Aging Bmi1+/- mice may thus represent an interesting model to identify and study novel pathogenic mechanisms related to AD.


Assuntos
Doença de Alzheimer/patologia , Instabilidade Genômica , Heterocromatina/metabolismo , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/mortalidade , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Estimativa de Kaplan-Meier , Potenciação de Longa Duração , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Memória Espacial , Proteínas tau/metabolismo
8.
Stem Cell Res ; 33: 215-227, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30453152

RESUMO

During development, multipotent progenitors undergo temporally-restricted differentiation into post-mitotic retinal cells; however, the mechanisms of progenitor division that occurs during retinogenesis remain controversial. Using clonal analyses (lineage tracing and single cell cultures), we identify rod versus cone lineage-specific progenitors derived from both adult retinal stem cells and embryonic neural retinal precursors. Taurine and retinoic acid are shown to act in an instructive and lineage-restricted manner early in the progenitor lineage hierarchy to produce rod-restricted progenitors from stem cell progeny. We also identify an instructive, but lineage-independent, mechanism for the specification of cone-restricted progenitors through the suppression of multiple differentiation signaling pathways. These data indicate that exogenous signals play critical roles in directing lineage decisions and resulting in fate-restricted rod or cone photoreceptor progenitors in culture. Additional factors may be involved in governing photoreceptor fates in vivo.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Retina/fisiopatologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Camundongos
9.
Stem Cell Reports ; 10(6): 1721-1733, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29706499

RESUMO

Brain neurogenesis is severely impaired following exposure to ionizing radiation (IR). We and others have shown that the expression of the tumor suppressor gene p16INK4a is increased in tissues exposed to IR and thus hypothesized that its expression could limit neurogenesis in the irradiated brain. Here, we found that exposure to IR leads to persistent DNA damage and the expression of p16INK4a in the hippocampus and subventricular zone regions. This was accompanied by a decline in neurogenesis, as determined by doublecortin expression and bromodeoxyuridine incorporation, an effect partially restored in Ink4a/arf-null mice. Increased neurogenesis in the absence of INK4a/ARF expression was independent of apoptosis and activation of the microglia. Moreover, treatment of irradiated mice with a superoxide dismutase mimetic or clearance of p16INK4a-expressing cells using mouse genetics failed to increase neurogenesis. In conclusion, our results suggest that IR-induced p16INK4a expression is a mechanism that limits neurogenesis.


Assuntos
Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação da Expressão Gênica/efeitos da radiação , Neurogênese/genética , Neurogênese/efeitos da radiação , Radiação Ionizante , Animais , Apoptose/genética , Biomarcadores , Biomimética , Encéfalo/patologia , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dano ao DNA/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Imagem Molecular , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/efeitos da radiação , Neurogênese/efeitos dos fármacos , Doses de Radiação , Superóxido Dismutase/metabolismo , Superóxido Dismutase/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Cell Rep ; 23(9): 2653-2666, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29847796

RESUMO

Late-onset sporadic Alzheimer's disease (AD) is the most prevalent form of dementia, but its origin remains poorly understood. The Bmi1/Ring1 protein complex maintains transcriptional repression of developmental genes through histone H2A mono-ubiquitination, and Bmi1 deficiency in mice results in growth retardation, progeria, and neurodegeneration. Here, we demonstrate that BMI1 is silenced in AD brains, but not in those with early-onset familial AD, frontotemporal dementia, or Lewy body dementia. BMI1 expression was also reduced in cortical neurons from AD patient-derived induced pluripotent stem cells but not in neurons overexpressing mutant APP and PSEN1. BMI1 knockout in human post-mitotic neurons resulted in amyloid beta peptide secretion and deposition, p-Tau accumulation, and neurodegeneration. Mechanistically, BMI1 was required to repress microtubule associated protein tau (MAPT) transcription and prevent GSK3beta and p53 stabilization, which otherwise resulted in neurodegeneration. Restoration of BMI1 activity through genetic or pharmaceutical approaches could represent a therapeutic strategy against AD.


Assuntos
Doença de Alzheimer/patologia , Modelos Biológicos , Complexo Repressor Polycomb 1/deficiência , Idade de Início , Doença de Alzheimer/genética , Amiloide/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Demência/metabolismo , Demência/patologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Fosforilação , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas tau/metabolismo
11.
J Biol Chem ; 292(50): 20669-20682, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29054928

RESUMO

Recent studies have reported that many proteases, besides the canonical α-, ß-, and γ-secretases, cleave the amyloid precursor protein (APP) and modulate ß-amyloid (Aß) peptide production. Moreover, specific APP isoforms contain Kunitz protease-inhibitory domains, which regulate the proteolytic activity of serine proteases. This prompted us to investigate the role of matriptase, a member of the type II transmembrane serine protease family, in APP processing. Using quantitative RT-PCR, we detected matriptase mRNA in several regions of the human brain with an enrichment in neurons. RNA sequencing data of human dorsolateral prefrontal cortex revealed relatively high levels of matriptase RNA in young individuals, whereas lower levels were detected in older individuals. We further demonstrate that matriptase and APP directly interact with each other and that matriptase cleaves APP at a specific arginine residue (Arg-102) both in vitro and in cells. Site-directed (Arg-to-Ala) mutagenesis of this cleavage site abolished matriptase-mediated APP processing. Moreover, we observed that a soluble, shed matriptase form cleaves endogenous APP in SH-SY5Y cells and that this cleavage significantly reduces APP processing to Aß40. In summary, this study identifies matriptase as an APP-cleaving enzyme, an activity that could have important consequences for the abundance of Aß and in Alzheimer's disease pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Fragmentos de Peptídeos/metabolismo , Serina Endopeptidases/metabolismo , Fatores Etários , Idoso , Encéfalo/metabolismo , Cadáver , Linhagem Celular , Biologia Computacional , Regulação Enzimológica da Expressão Gênica , Humanos , Mutagênese Sítio-Dirigida , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Especificidade de Órgãos , Córtex Pré-Frontal/enzimologia , Córtex Pré-Frontal/metabolismo , Proteólise , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/genética , Especificidade por Substrato , Adulto Jovem
12.
Development ; 143(9): 1571-84, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26965367

RESUMO

Retinal development occurs through the sequential but overlapping generation of six types of neuronal cells and one glial cell type. Of these, rod and cone photoreceptors represent the functional unit of light detection and phototransduction and are frequently affected in retinal degenerative diseases. During mouse development, the Polycomb group protein Bmi1 is expressed in immature retinal progenitors and differentiated retinal neurons, including cones. We show here that Bmi1 is required to prevent post natal degeneration of cone photoreceptors and bipolar neurons and that inactivation of Chk2 or p53 could improve but not overcome cone degeneration in Bmi1(-/-) mice. The retinal phenotype of Bmi1(-/-) mice was also characterized by loss of heterochromatin, activation of tandem repeats, oxidative stress and Rip3-associated necroptosis. In the human retina, BMI1 was preferentially expressed in cones at heterochromatic foci. BMI1 inactivation in human embryonic stem cells was compatible with retinal induction but impaired cone terminal differentiation. Despite this developmental arrest, BMI1-deficient cones recapitulated several anomalies observed in Bmi1(-/-) photoreceptors, such as loss of heterochromatin, activation of tandem repeats and induction of p53, revealing partly conserved biological functions between mouse and man.


Assuntos
Células-Tronco Embrionárias/citologia , Necrose/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Animais , Linhagem Celular , Quinase do Ponto de Checagem 2/genética , Heterocromatina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Proteína Serina-Treonina Quinases de Interação com Receptores , Retina/embriologia , Células Fotorreceptoras Retinianas Bastonetes/citologia , Proteína Supressora de Tumor p53/genética
13.
J Biol Chem ; 291(1): 182-97, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26468281

RESUMO

The polycomb repressive complex 1 (PRC1), containing the core BMI1 and RING1A/B proteins, mono-ubiquitinylates histone H2A (H2A(ub)) and is associated with silenced developmental genes at facultative heterochromatin. It is, however, assumed that the PRC1 is excluded from constitutive heterochromatin in somatic cells based on work performed on mouse embryonic stem cells and oocytes. We show here that BMI1 is required for constitutive heterochromatin formation and silencing in human and mouse somatic cells. BMI1 was highly enriched at intergenic and pericentric heterochromatin, co-immunoprecipitated with the architectural heterochromatin proteins HP1, DEK1, and ATRx, and was required for their localization. In contrast, BRCA1 localization was BMI1-independent and partially redundant with that of BMI1 for H2A(ub) deposition, constitutive heterochromatin formation, and silencing. These observations suggest a dynamic and developmentally regulated model of PRC1 occupancy at constitutive heterochromatin, and where BMI1 function in somatic cells is to stabilize the repetitive genome.


Assuntos
Inativação Gênica , Heterocromatina/metabolismo , Mamíferos/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proteína BRCA1/metabolismo , Córtex Cerebral/citologia , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Complexo Repressor Polycomb 1/deficiência , Proteínas Proto-Oncogênicas/deficiência , Sequências Repetitivas de Ácido Nucleico/genética , Ubiquitina/metabolismo
14.
Development ; 142(19): 3294-306, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443633

RESUMO

Cone photoreceptors are required for color discrimination and high-resolution central vision and are lost in macular degenerations, cone and cone/rod dystrophies. Cone transplantation could represent a therapeutic solution. However, an abundant source of human cones remains difficult to obtain. Work performed in model organisms suggests that anterior neural cell fate is induced 'by default' if BMP, TGFß and Wnt activities are blocked, and that photoreceptor genesis operates through an S-cone default pathway. We report here that Coco (Dand5), a member of the Cerberus gene family, is expressed in the developing and adult mouse retina. Upon exposure to recombinant COCO, human embryonic stem cells (hESCs) differentiated into S-cone photoreceptors, developed an inner segment-like protrusion, and could degrade cGMP when exposed to light. Addition of thyroid hormone resulted in a transition from a unique S-cone population toward a mixed M/S-cone population. When cultured at confluence for a prolonged period of time, COCO-exposed hESCs spontaneously developed into a cellular sheet composed of polarized cone photoreceptors. COCO showed dose-dependent and synergistic activity with IGF1 at blocking BMP/TGFß/Wnt signaling, while its cone-inducing activity was blocked in a dose-dependent manner by exposure to BMP, TGFß or Wnt-related proteins. Our work thus provides a unique platform to produce human cones for developmental, biochemical and therapeutic studies and supports the hypothesis that photoreceptor differentiation operates through an S-cone default pathway during human retinal development.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Retina/embriologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Western Blotting , Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
15.
J Neurosci ; 33(5): 2205-16, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23365256

RESUMO

The transcription factor p53 mediates neuronal death in a variety of stress-related and neurodegenerative conditions. The proapoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1 and ASPP2. However, whether ASPP1/2 play a role in the regulation of p53-dependent neuronal death in the CNS is currently unknown. To address this, we asked whether ASPP1/2 contribute to the death of retinal ganglion cells (RGCs) using in vivo models of acute optic nerve damage in mice and rats. Here, we show that p53 is activated in RGCs soon after injury and that axotomy-induced RGC death is attenuated in p53 heterozygote and null mice. We demonstrate that ASPP1/2 proteins are abundantly expressed by injured RGCs, and that short interfering (si)RNA-based ASPP1 or ASPP2 knockdown promotes robust RGC survival. Comparative gene expression analysis revealed that siASPP-mediated downregulation of p53-upregulated-modulator-of-apoptosis (PUMA), Fas/CD95, and Noxa depends on p53 transcriptional activity. Furthermore, siRNA against PUMA or Fas/CD95 confers neuroprotection, demonstrating a functional role for these p53 targets in RGC death. Our study demonstrates a novel role for ASPP1 and ASPP2 in the death of RGCs and provides evidence that blockade of the ASPP-p53 pathway is beneficial for central neuron survival after axonal injury.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Morte Celular/fisiologia , Células Ganglionares da Retina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Receptor fas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Axônios/metabolismo , Regulação para Baixo , Feminino , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Receptor fas/genética
16.
PLoS One ; 7(2): e31870, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384090

RESUMO

Aging increases the risk to develop several neurodegenerative diseases, although the underlying mechanisms are poorly understood. Inactivation of the Polycomb group gene Bmi1 in mice results in growth retardation, cerebellar degeneration, and development of a premature aging-like phenotype. This progeroid phenotype is characterized by formation of lens cataracts, apoptosis of cortical neurons, and increase of reactive oxygen species (ROS) concentrations, owing to p53-mediated repression of antioxidant response (AOR) genes. Herein we report that Bmi1 expression progressively declines in the neurons of aging mouse and human brains. In old brains, p53 accumulates at the promoter of AOR genes, correlating with a repressed chromatin state, down-regulation of AOR genes, and increased oxidative damages to lipids and DNA. Comparative gene expression analysis further revealed that aging brains display an up-regulation of the senescence-associated genes IL-6, p19(Arf) and p16(Ink4a), along with the pro-apoptotic gene Noxa, as seen in Bmi1-null mice. Increasing Bmi1 expression in cortical neurons conferred robust protection against DNA damage-induced cell death or mitochondrial poisoning, and resulted in suppression of ROS through activation of AOR genes. These observations unveil that Bmi1 genetic deficiency recapitulates aspects of physiological brain aging and that Bmi1 over-expression is a potential therapeutic modality against neurodegeneration.


Assuntos
Envelhecimento , Encéfalo/fisiologia , Doenças Neurodegenerativas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose , Encéfalo/metabolismo , Cromatina/metabolismo , DNA/metabolismo , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes p53 , Humanos , Peroxidação de Lipídeos , Lipídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças Neurodegenerativas/prevenção & controle , Neurônios/metabolismo , Estresse Oxidativo , Complexo Repressor Polycomb 1 , Regiões Promotoras Genéticas , Espécies Reativas de Oxigênio , Proteína Supressora de Tumor p53/metabolismo
17.
Cancers (Basel) ; 3(2): 1777-97, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-24212782

RESUMO

Glioblastoma multiforme (GBM), an aggressive brain tumor of astrocytic/neural stem cell origin, represents one of the most incurable cancers. GBM tumors are highly heterogeneous. However, most tumors contain a subpopulation of cells that display neural stem cell characteristics in vitro and that can generate a new brain tumor upon transplantation in mice. Hence, previously identified molecular pathways regulating neural stem cell biology were found to represent the cornerstone of GBM stem cell self-renewal mechanism. GBM tumors are also notorious for their resistance to radiation therapy. Notably, GBM "cancer stem cells" were also found to be responsible for this radioresistance. Herein, we will analyze the data supporting or not the cancer stem cell model in GBM, overview the current knowledge regarding GBM stem cell self-renewal and radioresistance molecular mechanisms, and discuss the potential therapeutic application of these findings.

18.
Antioxid Redox Signal ; 15(6): 1729-37, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20849375

RESUMO

Recent advances in delineating the biological functions of p53 had shed the light on its key role in the multifacets of cellular homeostasis. After its activation, via DNA damage, oxidative stress, or aberrant expression of oncogenes, p53 transduces its classical effect through several mechanisms comprising activation of the DNA repair machinery, cell cycle arrest, and initiation of apoptosis or senescence. In the mammalian brain, p53 plays critical functions in normal development, tumor suppression, neurodegenerative diseases, and aging. Herein, we focus on the constitutive pro-oxidant activity of p53 in neurons and discuss the potential implication of this finding in the context of neurodegenerative diseases and normal brain aging.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Humanos , Camundongos , Neurônios/metabolismo , Oxirredução
19.
J Neurosci ; 30(30): 10096-111, 2010 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-20668194

RESUMO

Glioblastoma multiforme (GBM) is an aggressive brain tumor that is resistant to all known therapies. Within these tumors, a CD133-positive cancer-initiating neural stem cell (NSC) population was shown to be resistant to gamma radiation through preferential activation of the DNA double-strand break (DSB) response machinery, including the ataxia-telangiectasia-mutated (ATM) kinase. The polycomb group protein BMI1 is enriched in CD133-positive GBM cells and required for their self-renewal in an INK4A/ARF-independent manner through transcriptional repression of alternate tumor suppressor pathways. We report here that BMI1 copurifies with DNA DSB response and nonhomologous end joining (NHEJ) repair proteins in GBM cells. BMI1 was enriched at the chromatin after irradiation and colocalized and copurified with ATM and the histone gammaH2AX. BMI1 also preferentially copurified with NHEJ proteins DNA-PK, PARP-1, hnRNP U, and histone H1 in CD133-positive GBM cells. BMI1 deficiency in GBM cells severely impaired DNA DSB response, resulting in increased sensitivity to radiation. In turn, BMI1 overexpression in normal NSCs enhanced ATM recruitment to the chromatin, the rate of gammaH2AX foci resolution, and resistance to radiation. BMI1 thus displays a previously uncharacterized function in controlling DNA DSB response and repair. Pharmacological inhibition of BMI1 combined with radiation therapy may provide an effective mean to target GBM stem cells.


Assuntos
Dano ao DNA/fisiologia , Células-Tronco Embrionárias/efeitos da radiação , Células-Tronco Neoplásicas/efeitos da radiação , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tolerância a Radiação , Proteínas Repressoras/metabolismo , Antígeno AC133 , Antígenos CD/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Transformação Celular Neoplásica/efeitos da radiação , Quinase do Ponto de Checagem 2 , Cromatografia Líquida/métodos , Ensaio Cometa/métodos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/fisiologia , Feto , Citometria de Fluxo/métodos , Glioblastoma/patologia , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Imunoprecipitação/métodos , Células-Tronco Neoplásicas/fisiologia , Proteínas Nucleares/genética , Peptídeos/metabolismo , Fosforilação/genética , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Espectrometria de Massas em Tandem/métodos , Transfecção/métodos
20.
Stem Cells ; 28(8): 1412-23, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20549707

RESUMO

The developing mammalian retina is generated by the proliferation and differentiation of multipotent retinal progenitor cells (RPCs) giving rise to neuronal and glial lineages. Whether an immature progenitor/stem cell subpopulation is present in the developing mammalian retina remains undefined. Deficiency in the polycomb group gene Bmi1 results in reduced proliferation and postnatal depletion of neural and hematopoietic stem cells. Here, we show that Bmi1 is required for the self-renewal of most immature RPCs and for postnatal retinal development. In the embryo, Bmi1 is highly enriched in a rare stage-specific embryonic antigen-1-positive RPC subpopulation expressing the stem cell markers Sox2, Lhx2, and Musashi. Gain-of-function experiments revealed that Bmi1 overexpression could convert RPCs having limited proliferation capacity into RPCs showing extensive proliferation and multiple differentiation capacities over time. At all developmental stages analyzed using the neurosphere assay, Bmi1 deficiency resulted in reduced proliferation and self-renewal of most immature RPCs. Reduced RPCs proliferation was also observed in the peripheral retina of Bmi1(-/-) fetus and newborn mice. The biological impact of these developmental anomalies was revealed by the reduced retinal diameter of Bmi1-deficient pups. P19(Arf) and p16(Ink4a) were upregulated in vivo and in vitro and coinactivation of p53, which lies downstream of p19(Arf), partially restored Bmi1-deficient RPCs self-renewal phenotype. Bmi1 thus distinguishes immature RPCs from the main RPC population and is required for normal retinal development.


Assuntos
Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Retina/citologia , Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Citometria de Fluxo , Camundongos , Proteínas Nucleares/genética , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA