Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Proteomics ; 13(3): 760-79, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24390426

RESUMO

Recent clinical evidence suggests that the neuroprotective and beneficial effects of hormone therapy may be limited by factors related to age and reproductive status. The patient's age and length of time without circulating ovarian hormones are likely to be key factors in the specific neurological outcomes of hormone therapy. However, the mechanisms underlying age-related changes in hormone efficacy have not been determined. We hypothesized that there are intrinsic changes in estrogen receptor ß (ERß) function that determine its ability to mediate the actions of 17ß-estradiol (E2) in brain regions such as the ventral hippocampus. In this study, we identified and quantified a subset of ERß protein interactions in the ventral hippocampus that were significantly altered by E2 replacement in young and aged animals, using two-dimensional differential gel electrophoresis coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry. This study demonstrates quantitative changes in ERß protein-protein interactions with E2 replacement that are dependent upon age in the ventral hippocampus and how these changes could alter processes such as transcriptional regulation. Thus, our data provide evidence that changes in ERß protein interactions are a potential mechanism for age-related changes in E2 responsiveness in the brain after menopause.


Assuntos
Envelhecimento/metabolismo , Estradiol/farmacologia , Receptor beta de Estrogênio/metabolismo , Hipocampo/metabolismo , Mapeamento de Interação de Proteínas , Adenosina Trifosfatases/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Anexina A5/metabolismo , Proteínas de Ciclo Celular/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Gelsolina/metabolismo , Técnicas de Silenciamento de Genes , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Células HEK293 , Hipocampo/efeitos dos fármacos , Humanos , Processamento de Imagem Assistida por Computador , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Endogâmicos F344 , Elementos de Resposta/genética , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteína com Valosina
2.
J Neurosci ; 30(2): 591-9, 2010 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20071522

RESUMO

Imbalanced protein load within cells is a critical aspect for most diseases of aging. In particular, the accumulation of proteins into neurotoxic aggregates is a common thread for a host of neurodegenerative diseases. Our previous work demonstrated that age-related changes to the cellular chaperone repertoire contributes to abnormal buildup of the microtubule-associated protein tau that accumulates in a group of diseases termed tauopathies, the most common being Alzheimer's disease. Here, we show that the Hsp90 cochaperone, FK506-binding protein 51 (FKBP51), which possesses both an Hsp90-interacting tetratricopeptide domain and a peptidyl-prolyl cis-trans isomerase (PPIase) domain, prevents tau clearance and regulates its phosphorylation status. Regulation of the latter is dependent on the PPIase activity of FKBP51. FKB51 enhances the association of tau with Hsp90, but the FKBP51/tau interaction is not dependent on Hsp90. In vitro FKBP51 stabilizes microtubules with tau in a reaction depending on the PPIase activity of FKBP51. Based on these new findings, we propose that FKBP51 can use the Hsp90 complex to isomerize tau, altering its phosphorylation pattern and stabilizing microtubules.


Assuntos
Encéfalo/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Microtúbulos/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas tau/metabolismo , Animais , Benzoquinonas/farmacologia , Linhagem Celular Transformada , Quimotripsina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Lactamas Macrocíclicas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/efeitos dos fármacos , Mutação/genética , Oócitos , Peptidilprolil Isomerase/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a Tacrolimo/genética , Transfecção/métodos , Xenopus
3.
J Neurosci ; 29(39): 12079-88, 2009 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-19793966

RESUMO

Alzheimer's disease and other tauopathies have recently been clustered with a group of nervous system disorders termed protein misfolding diseases. The common element established between these disorders is their requirement for processing by the chaperone complex. It is now clear that the individual components of the chaperone system, such as Hsp70 and Hsp90, exist in an intricate signaling network that exerts pleiotropic effects on a host of substrates. Therefore, we have endeavored to identify new compounds that can specifically regulate individual components of the chaperone family. Here, we hypothesized that chemical manipulation of Hsp70 ATPase activity, a target that has not previously been pursued, could illuminate a new pathway toward chaperone-based therapies. Using a newly developed high-throughput screening system, we identified inhibitors and activators of Hsp70 enzymatic activity. Inhibitors led to rapid proteasome-dependent tau degradation in a cell-based model. Conversely, Hsp70 activators preserved tau levels in the same system. Hsp70 inhibition did not result in general protein degradation, nor did it induce a heat shock response. We also found that inhibiting Hsp70 ATPase activity after increasing its expression levels facilitated tau degradation at lower doses, suggesting that we can combine genetic and pharmacologic manipulation of Hsp70 to control the fate of bound substrates. Disease relevance of this strategy was further established when tau levels were rapidly and substantially reduced in brain tissue from tau transgenic mice. These findings reveal an entirely novel path toward therapeutic intervention of tauopathies by inhibition of the previously untargeted ATPase activity of Hsp70.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/fisiologia , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP70/fisiologia , Proteínas tau/fisiologia , Adenosina Trifosfatases/antagonistas & inibidores , Animais , Corantes Azur/química , Corantes Azur/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Dobramento de Proteína/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
J Cell Mol Med ; 13(4): 619-30, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19449461

RESUMO

Molecular chaperones and heat shock proteins (Hsp) have emerged as critical regulators of proteins associated with neurodegenerative disease pathologies. The very nature of the chaperone system, which is to maintain protein quality control, means that most nascent proteins come in contact with chaperone proteins. Thus, amyloid precursor protein (APP), members of the gamma-secretase complex (presenilin 1 [PS1] collectively), the microtubule-associated protein tau (MAPT) as well as a number of neuroinflammatory components are all in contact with chaperones from the moment of their production. Chaperones are often grouped together as one machine presenting abnormal or mutant proteins to the proteasome for degradation, but this is not at all the case. In fact, the chaperone family consists of more than 100 proteins in mammalian cells, and the primary role for most of these proteins is to protect clients following synthesis and during stress; only as a last resort do they facilitate protein degradation. To the best of our current knowledge, the chaperone system in eukaryotic cells revolves around the ATPase activities of Hsp70 and Hsp90, the two primary chaperone scaffolds. Other chaperones and co-chaperones manipulate the ATPase activities of Hsp70 and Hsp90, facilitating either folding of the client or its degradation. In the case of Alzheimer's disease (AD), a number of studies have recently emerged describing the impact that these chaperones have on the proteotoxic effects of tau and amyloid- beta accumulation. Here, we present the current understandings of chaperone biology and examine the literature investigating these proteins in the context of AD.


Assuntos
Doença de Alzheimer/metabolismo , Chaperonas Moleculares/metabolismo , Transdução de Sinais , Doença de Alzheimer/patologia , Animais , Humanos , Modelos Biológicos
5.
Neurobiol Aging ; 61: 13-22, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29031089

RESUMO

Alternative RNA splicing results in the translation of diverse protein products arising from a common nucleotide sequence. These alternative protein products are often functional and can have widely divergent actions from the canonical protein. Studies in humans and other vertebrate animals have demonstrated that alternative splicing events increase with advanced age, sometimes resulting in pathological consequences. Menopause represents a critical transition for women, where the beneficial effects of estrogens are no longer evident; therefore, factors underlying increased pathological conditions in women are confounded by the dual factors of aging and declining estrogens. Estrogen receptors (ERs) are subject to alternative splicing, the spliced variants increase following menopause, and they fail to efficiently activate estrogen-dependent signaling pathways. However, the factors that regulate the alternative splicing of ERs remain unknown. We demonstrate novel evidence supporting a potential biological feedback loop where 17ß-estradiol regulates the RNA-binding protein Nova1, which, in turn, regulates the alternative splicing of ERß. These data increase our understanding of ER alternative splicing and could have potential implications for women taking hormone replacement therapy after menopause.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Processamento Alternativo/genética , Encéfalo/metabolismo , Estradiol/fisiologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica , Proteínas de Ligação a RNA/fisiologia , Animais , Células Cultivadas , Feminino , Humanos , Antígeno Neuro-Oncológico Ventral , Ratos Endogâmicos F344
6.
PLoS One ; 11(8): e0160276, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27487271

RESUMO

Aging and the coincident loss of circulating estrogens at menopause lead to increased risks for neurological and cardiovascular pathologies. Clinical studies show that estrogen therapy (ET) can be beneficial in mitigating these negative effects, in both the brain and heart, when it is initiated shortly after the perimenopausal transition. However, this same therapy is detrimental when initiated >10 years postmenopause. Importantly, the molecular mechanisms underlying this age-related switch in ET efficacy are unknown. Estrogen receptors (ERs) mediate the neuroprotective and cardioprotective functions of estrogens by modulating gene transcription or, non-genomically, by activating second messenger signaling pathways, such as mitogen activated protein kinases (MAPK). These kinases are critical regulators of cell signaling pathways and have widespread downstream effects. Our hypothesis is that age and estrogen deprivation following menopause alters the expression and activation of the MAPK family members p38 and ERK in the brain and heart. To test this hypothesis, we used a surgically induced model of menopause in 18 month old rats through bilateral ovariectomy (OVX) followed by an acute dose of 17ß-estradiol (E2) administered at varying time points post-OVX (1 week, 4 weeks, 8 weeks, or 12 weeks). Age and E2 treatment differentially regulated kinase activity in both the brain and heart, and the effects were also brain region specific. MAPK signaling plays an integral role in aging, and the aberrant regulation of those signaling pathways might be involved in age-related disorders. Clinical studies show benefits of ET during early menopause but detrimental effects later, which might be reflective of changes in kinase expression and activation status.


Assuntos
Envelhecimento/fisiologia , Encéfalo/efeitos dos fármacos , Estradiol/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Coração/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Feminino , Miocárdio/metabolismo , Ovariectomia , Ratos , Ratos Endogâmicos F344 , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Oncotarget ; 6(35): 36965-83, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26460619

RESUMO

Administration of 17ß-estradiol (E2) has beneficial effects on cognitive function in peri- but not post-menopausal women, yet the molecular mechanisms underlying age-related changes in E2 action remain unclear. We propose that there is a biological switch in E2 action that occurs coincident with age and length of time after ovarian hormone depletion, and we hypothesized that age-dependent regulation of microRNAs (miRNAs) could be the molecular basis for that switch. Previously we showed that miRNAs are regulated by E2 in young compared to aged female rats. Here we tested whether increasing lengths of ovarian hormone deprivation in aged females altered E2 regulation of these mature miRNAs. In addition, we determined where along the miRNA biogenesis pathway E2 exerted its effects. Our results showed that age and increased lengths of ovarian hormone deprivation abolished the ability of E2 to regulate mature miRNA expression in the brain. Further, we show that E2 acted at specific points along the miRNA biogenesis pathway.


Assuntos
Envelhecimento , Encéfalo/metabolismo , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , MicroRNAs/genética , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Feminino , Perfilação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Ovariectomia , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Endocrinology ; 156(11): 4187-99, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26295370

RESUMO

Loss of circulating 17ß-estradiol (E2) that occurs during menopause can have detrimental effects on cognitive function. The efficacy of hormone replacement therapy declines as women become farther removed from the menopausal transition, yet the molecular mechanisms underlying this age-related switch in E2 efficacy are unknown. We hypothesized that aging and varying lengths of E2 deprivation alters the ratio of alternatively spliced estrogen receptor (ER)ß isoforms in the brain of female rats. Further, we tested whether changes in global transcriptional activity and splicing kinetics regulate the alternative splicing of ERß. Our results revealed brain region-specific changes in ERß alternative splicing in both aging and E2-deprivation paradigms and showed that ERß could mediate E2-induced alternative splicing. Global transcriptional activity, as measured by phosphorylated RNA polymerase II, was also regulated by age and E2 in specific brain regions. Finally, we show that inhibition of topoisomerase I resulted in increased ERß2 splice variant expression.


Assuntos
Envelhecimento/metabolismo , Processamento Alternativo , Encéfalo/metabolismo , Estradiol/sangue , Receptor beta de Estrogênio/metabolismo , Isoformas de Proteínas/metabolismo , Envelhecimento/genética , Animais , Receptor beta de Estrogênio/genética , Feminino , Fosforilação , Isoformas de Proteínas/genética , Ratos , Ratos Endogâmicos F344 , Transcrição Gênica , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA