Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(23): E4676-E4685, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533375

RESUMO

The activity of the transcription factor nuclear factor-erythroid 2 p45-derived factor 2 (NRF2) is orchestrated and amplified through enhanced transcription of antioxidant and antiinflammatory target genes. The present study has characterized a triazole-containing inducer of NRF2 and elucidated the mechanism by which this molecule activates NRF2 signaling. In a highly selective manner, the compound covalently modifies a critical stress-sensor cysteine (C151) of the E3 ligase substrate adaptor protein Kelch-like ECH-associated protein 1 (KEAP1), the primary negative regulator of NRF2. We further used this inducer to probe the functional consequences of selective activation of NRF2 signaling in Huntington's disease (HD) mouse and human model systems. Surprisingly, we discovered a muted NRF2 activation response in human HD neural stem cells, which was restored by genetic correction of the disease-causing mutation. In contrast, selective activation of NRF2 signaling potently repressed the release of the proinflammatory cytokine IL-6 in primary mouse HD and WT microglia and astrocytes. Moreover, in primary monocytes from HD patients and healthy subjects, NRF2 induction repressed expression of the proinflammatory cytokines IL-1, IL-6, IL-8, and TNFα. Together, our results demonstrate a multifaceted protective potential of NRF2 signaling in key cell types relevant to HD pathology.


Assuntos
Doença de Huntington/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Adulto , Idoso , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Doença de Huntington/genética , Proteína 1 Associada a ECH Semelhante a Kelch/química , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/prevenção & controle , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pessoa de Meia-Idade , Fator 2 Relacionado a NF-E2/química , Células-Tronco Neurais/metabolismo , Fármacos Neuroprotetores/farmacologia , Conformação Proteica/efeitos dos fármacos , Ratos , Transdução de Sinais
2.
Front Pharmacol ; 3: 82, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563317

RESUMO

Sirtuin proteins are conserved regulators of aging that have recently emerged as important modifiers of several diseases which commonly occur later in life such as cancer, diabetes, cardiovascular, and neurodegenerative diseases. In mammals, there are seven sirtuins (SIRT1-7), which display diversity in subcellular localization and function. SIRT1 has received much of attention due to its possible impact on longevity, while important biological and therapeutic roles of other sirtuins have been underestimated and just recently recognized. Here we focus on SIRT2, a member of the sirtuin family, and discuss its role in cellular and tissue-specific functions. This review summarizes the main scientific advances on SIRT2 protein biology and explores its potential as a therapeutic target for treatment of age-related disorders.

3.
Hum Mol Genet ; 20(20): 3986-96, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791548

RESUMO

Sirtuin 2 (SIRT2) is one of seven known mammalian protein deacetylases homologous to the yeast master lifespan regulator Sir2. In recent years, the sirtuin protein deacetylases have emerged as candidate therapeutic targets for many human diseases, including metabolic and age-dependent neurological disorders. In non-neuronal cells, SIRT2 has been shown to function as a tubulin deacetylase and a key regulator of cell division and differentiation. However, the distribution and function of the SIRT2 microtubule (MT) deacetylase in differentiated, postmitotic neurons remain largely unknown. Here, we show abundant and preferential expression of specific isoforms of SIRT2 in the mammalian central nervous system and find that a previously uncharacterized form, SIRT2.3, exhibits age-dependent accumulation in the mouse brain and spinal cord. Further, our studies reveal that focal areas of endogenous SIRT2 expression correlate with reduced α-tubulin acetylation in primary mouse cortical neurons and suggest that the brain-enriched species of SIRT2 may function as the predominant MT deacetylases in mature neurons. Recent reports have demonstrated an association between impaired tubulin acetyltransferase activity and neurodegenerative disease; viewed in this light, our results showing age-dependent accumulation of the SIRT2 neuronal MT deacetylase in wild-type mice suggest a functional link between tubulin acetylation patterns and the aging brain.


Assuntos
Envelhecimento/metabolismo , Sistema Nervoso Central/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Sirtuína 2/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ordem dos Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/genética , Isoformas de Proteínas/metabolismo , Sirtuína 2/genética
4.
ACS Chem Biol ; 6(6): 540-6, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21370928

RESUMO

Sirtuin 2 (SIRT2) deacetylase-dependent inhibition mediates neuroprotective reduction of cholesterol biosynthesis in an in vitro Huntington's disease model. This study sought to identify the first brain-permeable SIRT2 inhibitor and to characterize its cholesterol-reducing properties in neuronal models. Using biochemical sirtuin deacetylation assays, we screened a brain-permeable in silico compound library, yielding 3-(1-azepanylsulfonyl)-N-(3-bromphenyl)benzamide as the most potent and selective SIRT2 inhibitor. Pharmacokinetic studies demonstrated brain-permeability but limited metabolic stability of the selected candidate. In accordance with previous observations, this SIRT2 inhibitor stimulated cytoplasmic retention of sterol regulatory element binding protein-2 and subsequent transcriptional downregulation of cholesterol biosynthesis genes, resulting in reduced total cholesterol in primary striatal neurons. Furthermore, the identified inhibitor reduced cholesterol in cultured naïve neuronal cells and brain slices from wild-type mice. The outcome of this study provides a clear opportunity for lead optimization and drug development, targeting metabolic dysfunctions in CNS disorders where abnormal cholesterol homeostasis is implicated.


Assuntos
Encéfalo/metabolismo , Colesterol/biossíntese , Inibidores Enzimáticos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sirtuína 2/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/química , Camundongos , Modelos Neurológicos , Estrutura Molecular , Neurônios/enzimologia , Permeabilidade , Sirtuína 2/metabolismo , Bibliotecas de Moléculas Pequenas , Estereoisomerismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
5.
Int J Biochem Cell Biol ; 43(1): 20-4, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21056115

RESUMO

Huntington's disease is an autosomal dominant genetic neurodegenerative disorder, which is characterized by progressive motor dysfunction, emotional disturbances, dementia, and weight loss. The disease is caused by pathological CAG-triplet repeat extension(s), encoding polyglutamines, within the gene product, huntingtin. Huntingtin is ubiquitously expressed through the body and is a protein of uncertain molecular function(s). Mutant huntingtin, containing pathologically extended polyglutamines causes the earliest and most dramatic neuropathologic changes in the neostriatum and cerebral cortex. Extended polyglutamines confer structural conformational changes to huntingtin, which gains novel properties, resulting in aberrant interactions with multiple cellular components. The diverse and variable aberrations mediated by mutant huntingtin perturb many cellular functions essential for neuronal homeostasis and underlie pleiotropic mechanisms of Huntington's disease pathogenesis. The only approved drug for Huntington's disease is a symptomatic treatment, tetrabenazine; thus, novel neuroprotective strategies, slowing, blocking and possibly reversing disease progression, are vital for developing effective therapies.


Assuntos
Córtex Cerebral/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Neostriado/metabolismo , Proteínas do Tecido Nervoso , Proteínas Nucleares , Inibidores da Captação Adrenérgica/uso terapêutico , Animais , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Doença de Huntington/terapia , Camundongos , Mutação , Neostriado/patologia , Neostriado/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Conformação Proteica , Tetrabenazina/uso terapêutico , Repetições de Trinucleotídeos/genética
7.
J Biol Chem ; 282(34): 24554-62, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17565993

RESUMO

Increased levels of the repressor element 1/neuron restrictive silencer element (RE1/NRSE) silencing activity promoter, and a consequent reduction in the transcription of many RE1/NRSE-bearing neuronal genes, including brain-derived neurotrophic factor (BDNF), have been demonstrated in Huntington disease (HD) and represent one possible effector of its selective neuronal vulnerability. Restoring the expression levels of neuronal genes in diseased neurons therefore seems to be an attractive therapeutic approach. To this end, we have developed a cell-based reporter assay for monitoring RE1/NRSE silencing activity and validated it by genetically inactivating the RE1/NRSE or pharmacologically stimulating global transcription. In a pilot compound screen, we identified three closely related structural analogues that up-regulate reporter expression at low nanomolar concentrations, and follow-up studies have shown that they efficaciously increase endogenous BDNF levels in HD cells. Moreover, one of the compounds increases the viability of HD cells. Our findings suggest a new avenue for the development of drugs for HD and other neurodegenerative disorders based on the pharmacological up-regulation of the production of the neuronal survival factor BDNF and of other RE1/NRSE-regulated neuronal genes.


Assuntos
Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Inativação Gênica , Proteína Huntingtina , Imuno-Histoquímica , Luciferases/metabolismo , Modelos Químicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/química , Ratos , Fatores de Transcrição/metabolismo
8.
Science ; 317(5837): 516-9, 2007 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-17588900

RESUMO

The sirtuins are members of the histone deacetylase family of proteins that participate in a variety of cellular functions and play a role in aging. We identified a potent inhibitor of sirtuin 2 (SIRT2) and found that inhibition of SIRT2 rescued alpha-synuclein toxicity and modified inclusion morphology in a cellular model of Parkinson's disease. Genetic inhibition of SIRT2 via small interfering RNA similarly rescued alpha-synuclein toxicity. Furthermore, the inhibitors protected against dopaminergic cell death both in vitro and in a Drosophila model of Parkinson's disease. The results suggest a link between neurodegeneration and aging.


Assuntos
Furanos/farmacologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/fisiopatologia , Quinolinas/farmacologia , Sirtuínas/antagonistas & inibidores , Sirtuínas/metabolismo , alfa-Sinucleína/metabolismo , Acetilação , Animais , Animais Geneticamente Modificados , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Dopamina/fisiologia , Relação Dose-Resposta a Droga , Drosophila melanogaster , Humanos , Modelos Moleculares , Neurônios/citologia , Neurônios/efeitos dos fármacos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Conformação Proteica , RNA Interferente Pequeno/genética , Ratos , Sirtuína 1 , Sirtuína 2 , Sirtuínas/química , Sirtuínas/genética , Transfecção , Tubulina (Proteína)/metabolismo , alfa-Sinucleína/genética
9.
J Biomol Screen ; 12(3): 351-60, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17379859

RESUMO

CAG-triplet repeat extension, translated into polyglutamines within the coding frame of otherwise unrelated gene products, causes 9 incurable neurodegenerative disorders, including Huntington's disease. Although an expansion in the CAG repeat length is the autosomal dominant mutation that causes the fully penetrant neurological phenotypes, the repeat length is inversely correlated with the age of onset. The precise molecular mechanism(s) of neurodegeneration remains elusive, but compelling evidence implicates the protein or its proteolytic fragments as the cause for the gain of novel pathological function(s). The authors sought to identify small molecules that target the selective clearance of polypeptides containing pathological polyglutamine extension. In a high-throughput chemical screen, they identified compounds that facilitate the clearance of a small huntingtin fragment with extended polyglutamines fused to green fluorescent protein reporter. Identified hits were validated in dose-response and toxicity tests. Compounds have been further tested in an assay for clearance of a larger huntingtin fragment, containing either pathological or normal polyglutamine repeats. In this assay, the authors identified compounds selectively targeting the clearance of mutant but not normal huntingtin fragments. These compounds were subjected to a functional assay, which yielded a lead compound that rescues cells from induced mutant polyglutamine toxicity.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Relação Dose-Resposta a Droga , Proteínas de Fluorescência Verde/metabolismo , Peso Molecular , Células PC12 , Peptídeos , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Especificidade por Substrato
10.
J Biomol Screen ; 11(7): 729-35, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16928982

RESUMO

Familial amyotrophic lateral sclerosis (ALS) accounts for 10% of all ALS cases; approximately 25% of these cases are due to mutations in the Cu/Zn superoxide dismutase gene (SOD1). To date, 105 different mutations spanning all 5 exons have been identified in the SOD1 gene. Mutant SOD1-associated ALS is caused by a toxic gain of function of the mutated protein. Therefore, regardless of the specific mechanism whereby mutant SOD1 initiates motor neuron death, the authors hypothesize that measures that decrease levels of mutant SOD1 protein should ameliorate the phenotype in transgenic mice and potentially in patients with SOD1-mediated disease. They have designed 2 cell-based screening assays to identify small, brain-permeant molecules that inactivate expression of the SOD1 gene or increase the degradation of the SOD1 protein. Here they describe the development and optimization of these assays and the results of high-throughput screening using a variety of compound libraries, including a total of more than 116,000 compounds. The majority of the hit compounds identified that down-regulated SOD1 were shown to be toxic in a cell-based viability assay or were nonselective transcription inhibitors, but work is continuing on a number of nonspecific inhibitors of SOD1 expression. Ultimately, the authors believe that these 2 cell-based assays will provide powerful strategies to identify novel therapies for the treatment of inherited SOD1-associated forms of ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/enzimologia , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Superóxido Dismutase/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Proteínas Mutantes/metabolismo , Células PC12 , Regiões Promotoras Genéticas/genética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Superóxido Dismutase-1
11.
Chem Biol ; 13(7): 765-70, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16873024

RESUMO

Poly (ADP-ribose) polymerase (PARP1) is a nuclear protein that, when overactivated by oxidative stress-induced DNA damage, ADP ribosylates target proteins leading to dramatic cellular ATP depletion. We have discovered a biologically active small-molecule inhibitor of PARP1. The discovered compound inhibited PARP1 enzymatic activity in vitro and prevented ATP loss and cell death in a surrogate model of oxidative stress in vivo. We also investigated a new use for PARP1 inhibitors in energy-deficient cells by using Huntington's disease as a model. Our results showed that insult with the oxidant hydrogen peroxide depleted cellular ATP in mutant cells below the threshold of viability. The protective role of PARP1 inhibitors against oxidative stress has been shown in this model system.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Trifosfato de Adenosina/metabolismo , Western Blotting , Inibidores Enzimáticos/química , Células HeLa , Humanos , Modelos Moleculares
12.
Proc Natl Acad Sci U S A ; 102(3): 892-7, 2005 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-15642944

RESUMO

Polyglutamine (polyQ) disorders, including Huntington's disease (HD), are caused by expansion of polyQ-encoding repeats within otherwise unrelated gene products. In polyQ diseases, the pathology and death of affected neurons are associated with the accumulation of mutant proteins in insoluble aggregates. Several studies implicate polyQ-dependent aggregation as a cause of neurodegeneration in HD, suggesting that inhibition of neuronal polyQ aggregation may be therapeutic in HD patients. We have used a yeast-based high-throughput screening assay to identify small-molecule inhibitors of polyQ aggregation. We validated the effects of four hit compounds in mammalian cell-based models of HD, optimized compound structures for potency, and then tested them in vitro in cultured brain slices from HD transgenic mice. These efforts identified a potent compound (IC50=10 nM) with long-term inhibitory effects on polyQ aggregation in HD neurons. Testing of this compound in a Drosophila HD model showed that it suppresses neurodegeneration in vivo, strongly suggesting an essential role for polyQ aggregation in HD pathology. The aggregation inhibitors identified in this screen represent four primary chemical scaffolds and are strong lead compounds for the development of therapeutics for human polyQ diseases.


Assuntos
Anilidas/farmacologia , Doença de Huntington/patologia , Doenças Neurodegenerativas/prevenção & controle , Neurônios/patologia , Peptídeos/antagonistas & inibidores , Hidrocarbonetos Policíclicos Aromáticos/farmacologia , Sulfonamidas/farmacologia , Animais , Encéfalo/patologia , Dimerização , Modelos Animais de Doenças , Drosophila , Doença de Huntington/metabolismo , Concentração Inibidora 50 , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Relação Estrutura-Atividade
13.
Proc Natl Acad Sci U S A ; 101(9): 3178-83, 2004 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-14981234

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neurodegenerative disorder resulting from selective death of motor neurons in the brain and spinal cord. In approximately 25% of familial ALS cases, the disease is caused by dominantly acting point mutations in the gene encoding cytosolic Cu,Zn superoxide dismutase (SOD1). In cell culture and in rodent models of ALS, mutant SOD1 proteins exhibit dose-dependent toxicity; thus, agents that reduce mutant protein expression would be powerful therapeutic tools. A wealth of recent evidence has demonstrated that the mechanism of RNA-mediated interference (RNAi) can be exploited to achieve potent and specific gene silencing in vitro and in vivo. We have evaluated the utility of RNAi for selective silencing of mutant SOD1 expression in cultured cells and have identified small interfering RNAs capable of specifically inhibiting expression of ALS-linked mutant, but not wild-type, SOD1. We have investigated the functional effects of RNAi-mediated silencing of mutant SOD1 in cultured murine neuroblastoma cells. In this model, stable expression of mutant, but not wild-type, human SOD1 sensitizes cells to cytotoxic stimuli. We find that silencing of mutant SOD1 protects these cells against cyclosporin A-induced cell death. These results demonstrate a positive physiological effect caused by RNAi-mediated silencing of a dominant disease allele. The present study further supports the therapeutic potential of RNAi-based methods for the treatment of inherited human diseases, including ALS.


Assuntos
Morte Celular/efeitos dos fármacos , Ciclosporina/toxicidade , Inativação Gênica , Neuroblastoma/genética , RNA Interferente Pequeno/genética , Superóxido Dismutase/genética , Sequência de Bases , Morte Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Primers do DNA , DNA Complementar/genética , Células HeLa , Humanos , Doença dos Neurônios Motores/genética , Mutagênese Sítio-Dirigida , Neuroblastoma/patologia , Superóxido Dismutase-1 , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA