Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Proc Natl Acad Sci U S A ; 107(17): 7927-32, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20378838

RESUMO

Huntington's disease (HD), an incurable neurodegenerative disorder, has a complex pathogenesis including protein aggregation and the dysregulation of neuronal transcription and metabolism. Here, we demonstrate that inhibition of sirtuin 2 (SIRT2) achieves neuroprotection in cellular and invertebrate models of HD. Genetic or pharmacologic inhibition of SIRT2 in a striatal neuron model of HD resulted in gene expression changes including significant down-regulation of RNAs responsible for sterol biosynthesis. Whereas mutant huntingtin fragments increased sterols in neuronal cells, SIRT2 inhibition reduced sterol levels via decreased nuclear trafficking of SREBP-2. Importantly, manipulation of sterol biosynthesis at the transcriptional level mimicked SIRT2 inhibition, demonstrating that the metabolic effects of SIRT2 inhibition are sufficient to diminish mutant huntingtin toxicity. These data identify SIRT2 inhibition as a promising avenue for HD therapy and elucidate a unique mechanism of SIRT2-inhibitor-mediated neuroprotection. Furthermore, the ascertainment of SIRT2's role in regulating cellular metabolism demonstrates a central function shared with other sirtuin proteins.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Doença de Huntington/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Sirtuína 2/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Esteróis/biossíntese , Análise de Variância , Animais , Western Blotting , Caenorhabditis elegans , Drosophila , Perfilação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Microscopia Confocal
3.
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
4.
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
5.
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
6.
Cell Chem Biol ; 23(7): 849-861, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27427231

RESUMO

There are currently no disease-modifying therapies for the neurodegenerative disorder Huntington's disease (HD). This study identified novel thiazole-containing inhibitors of the deacetylase sirtuin-2 (SIRT2) with neuroprotective activity in ex vivo brain slice and Drosophila models of HD. A systems biology approach revealed an additional SIRT2-independent property of the lead-compound, MIND4, as an inducer of cytoprotective NRF2 (nuclear factor-erythroid 2 p45-derived factor 2) activity. Structure-activity relationship studies further identified a potent NRF2 activator (MIND4-17) lacking SIRT2 inhibitory activity. MIND compounds induced NRF2 activation responses in neuronal and non-neuronal cells and reduced production of reactive oxygen species and nitrogen intermediates. These drug-like thiazole-containing compounds represent an exciting opportunity for development of multi-targeted agents with potentially synergistic therapeutic benefits in HD and related disorders.


Assuntos
Modelos Animais de Doenças , Doença de Huntington/tratamento farmacológico , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Sirtuína 2/antagonistas & inibidores , Tiazóis/farmacologia , Tiazóis/uso terapêutico , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Drosophila , Doença de Huntington/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/uso terapêutico , Ratos , Sirtuína 2/deficiência , Sirtuína 2/metabolismo , Relação Estrutura-Atividade , Tiazóis/química
7.
EMBO Mol Med ; 4(7): 557-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22610822

RESUMO

From February 12-16, 2012, leading members of the sirtuin scientific community assembled in Tahoe, CA to attend the Keystone Symposium "Sirtuins in Aging, Metabolism, and Disease." It was a vibrant and lively meeting, and in the spirit of Keystone Symposia, both established sirtuin researchers and those new to the field enjoyed a unique opportunity to interact and exchange ideas.


Assuntos
Envelhecimento , Sirtuínas/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Sirtuínas/química , Sirtuínas/genética
8.
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
9.
PLoS Curr ; 22010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20877454

RESUMO

The family of histone deacetylases (HDACs) has recently emerged as important drug targets for treatment of slow progressive neurodegenerative disorders, including Huntington's disease (HD). Broad pharmaceutical inhibition of HDACs has shown neuroprotective effects in various HD models. Here we examined the susceptibility of HDAC targets for drug treatment in affected brain areas during HD progression. We observed increased HDAC1 and decreased HDAC4, 5 and 6 levels, correlating with disease progression, in cortices and striata of HD R6/2 mice. However, there were no significant changes in HDAC protein levels, assessed in an age-dependent manner, in HD knock-in CAG140 mice and we did not observe significant changes in HDAC1 levels in human HD brains. We further assessed acetylation levels of α-tubulin, as a biomarker of HDAC6 activity, and found it unchanged in cortices from R6/2, knock-in, and human subjects at all disease stages. Inhibition of deacetylase activities was identical in cortical extracts from R6/2 and wild-type mice treated with a class II-selective HDAC inhibitor. Lastly, treatment with class I- and II-selective HDAC inhibitors showed similar responses in HD and wild-type rat striatal cells. In conclusion, our results show that class I and class II HDAC targets are present and accessible for chronic drug treatment during HD progression and provide impetus for therapeutic development of brain-permeable class- or isoform-selective inhibitors.

10.
Neuromuscul Disord ; 20(2): 111-21, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20080405

RESUMO

Glucocorticoids are beneficial in many muscular dystrophies but they are ineffective in treating dysferlinopathy, a rare muscular dystrophy caused by loss of dysferlin. We sought to understand the molecular basis for this disparity by studying the effects of a glucocorticoid on differentiation of the myoblast cell line, C2C12, and dysferlin-deficient C2C12s. We found that pharmacologic doses of dexamethasone enhanced the myogenic fusion efficiency of C2C12s and increased the induction of dysferlin, along with specific myogenic transcription factors, sarcolemmal and structural proteins. In contrast, the dysferlin-deficient C2C12 cell line demonstrated a reduction in long myotubes and early induction of particular muscle differentiation proteins, most notably, myosin heavy chain. Dexamethasone partially reversed the defect in myogenic fusion in the dysferlin-deficient C2C12 cells. We hypothesize that a key therapeutic benefit of glucocorticoids may be the up-regulation of dysferlin as an important component of glucocorticoid-enhanced myogenic differentiation.


Assuntos
Dexametasona/farmacologia , Proteínas de Membrana/agonistas , Desenvolvimento Muscular/efeitos dos fármacos , Doenças Musculares/tratamento farmacológico , Mioblastos/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Dexametasona/uso terapêutico , Relação Dose-Resposta a Droga , Disferlina , Glucocorticoides/farmacologia , Glucocorticoides/uso terapêutico , Proteínas de Membrana/biossíntese , Proteínas de Membrana/deficiência , Camundongos , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/efeitos dos fármacos , Proteínas Musculares/metabolismo , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia , Mioblastos/metabolismo , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
11.
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
12.
Proc Natl Acad Sci U S A ; 103(11): 4246-51, 2006 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-16537516

RESUMO

Misfolded proteins accumulate in many neurodegenerative diseases, including huntingtin in Huntington's disease and alpha-synuclein in Parkinson's disease. The disease-causing proteins can take various conformations and are prone to aggregate and form larger cytoplasmic or nuclear inclusions. One approach to the development of therapeutic intervention for these diseases has been to identify chemical compounds that reduce the size or number of inclusions. We have, however, identified a compound that promotes inclusion formation in cellular models of both Huntington's disease and Parkinson's disease. Of particular interest, this compound prevents huntingtin-mediated proteasome dysfunction and reduces alpha-synuclein-mediated toxicity. These results demonstrate that compounds that increase inclusion formation may actually lessen cellular pathology in both Huntington's and Parkinson's diseases, suggesting a therapeutic approach for neurodegenerative diseases caused by protein misfolding.


Assuntos
Doença de Huntington/tratamento farmacológico , Corpos de Inclusão/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Piperazinas/farmacologia , Quinolinas/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Linhagem Celular , Cricetinae , DNA Recombinante/genética , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Técnicas In Vitro , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , alfa-Sinucleína/toxicidade
13.
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
14.
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