Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Biosci (Elite Ed) ; 4(8): 2701-8, 2012 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-22652679

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal degenerative motor neuron disease. Approximately 20 percent of familial ALS cases are caused by mutations in the Cu/Zn superoxide dismutase (SOD1) gene. Rodents expressing mutant SOD1 transgenes develop progressive, fatal motor neuron disease and disease onset and progression is dependent on the level of SOD1. We investigated the possibility that a reduction in SOD1 protein may be of therapeutic benefit in ALS and screened 30,000 compounds for inhibition of SOD1 transcription. The most effective inhibitor identified was N-{4-[4-(4-methylbenzoyl)-1-piperazinyl]phenyl}-2-thiophenecarboxamide (Compound ID 7687685), which in PC12 cells showed an EC50 of 10.6 microM for inhibition of SOD1 expression and an LD50 more than 30 microM. This compound was subsequently shown to reduce endogenous SOD1 levels in HeLa cells and to exhibit a modest reduction of SOD1 protein levels in mouse spinal cord tissue. These data suggest that the efficacy of compound 7687685 as an inhibitor of SOD1 gene expression is not likely to be clinically useful, although the strategy reported could be applied broadly to screening for small molecule inhibitors of gene expression.


Assuntos
Superóxido Dismutase/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Western Blotting , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Células PC12 , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Ratos , Relação Estrutura-Atividade , Superóxido Dismutase-1
2.
Bioorg Med Chem ; 20(2): 1029-45, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22189273

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Currently, there is only one FDA-approved treatment for ALS (riluzole), and that drug only extends life, on average, by 2-3 months. Mutations in Cu/Zn superoxide dismutase (SOD1) are found in familial forms of the disease and have played an important role in the study of ALS pathophysiology. On the basis of their activity in a PC12-G93A-YFP high-throughput screening assay, several bioactive compounds have been identified and classified as cyclohexane-1,3-dione (CHD) derivatives. A concise and efficient synthetic route has been developed to provide diverse CHD analogs. The structural modification of the CHD scaffold led to the discovery of a more potent analog (26) with an EC(50) of 700 nM having good pharmacokinetic properties, such as high solubility, low human and mouse metabolic potential, and relatively good plasma stability. It was also found to efficiently penetrate the blood-brain barrier. However, compound 26 did not exhibit any significant life span extension in the ALS mouse model. It was found that, although 26 was active in PC12 cells, it had poor activity in other cell types, including primary cortical neurons, indicating that it can penetrate into the brain, but is not active in neuronal cells, potentially due to poor selective cell penetration. Further structural modification of the CHD scaffold was aimed at improving global cell activity as well as maintaining potency. Two new analogs (71 and 73) were synthesized, which had significantly enhanced cortical neuronal cell permeability, as well as similar potency to that of 26 in the PC12-G93A assay. These CHD analogs are being investigated further as novel therapeutic candidates for ALS.


Assuntos
Cicloexanonas/química , Cicloexanonas/farmacologia , Ciclopropanos/química , Éteres Fenílicos/química , Superóxido Dismutase/antagonistas & inibidores , Substituição de Aminoácidos , Esclerose Lateral Amiotrófica/tratamento farmacológico , Animais , Barreira Hematoencefálica/metabolismo , Cicloexanonas/uso terapêutico , Cicloexanonas/toxicidade , Ciclopropanos/uso terapêutico , Ciclopropanos/toxicidade , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/efeitos dos fármacos , Células PC12 , Éteres Fenílicos/uso terapêutico , Éteres Fenílicos/toxicidade , Ratos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
3.
J Med Chem ; 55(1): 515-27, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22191331

RESUMO

Amyotrophic lateral sclerosis (ALS) is an orphan neurodegenerative disease currently without a cure. The arylsulfanyl pyrazolone (ASP) scaffold was one of the active scaffolds identified in a cell-based high throughput screening assay targeting mutant Cu/Zn superoxide dismutase 1 (SOD1) induced toxicity and aggregation as a marker for ALS. The initial ASP hit compounds were potent and had favorable ADME properties but had poor microsomal and plasma stability. Here, we identify the microsomal metabolite and describe synthesized analogues of these ASP compounds to address the rapid metabolism. Both in vitro potency and pharmacological properties of the ASP scaffold have been dramatically improved via chemical modification to the corresponding sulfone and ether derivatives. One of the ether analogues (13), with superior potency and in vitro pharmacokinetic properties, was tested in vivo for its pharmacokinetic profile, brain penetration, and efficacy in an ALS mouse model. The analogue showed sustained blood and brain levels in vivo and significant activity in the mouse model of ALS, thus validating the new aryloxanyl pyrazolone scaffold as an important novel therapeutic lead for the treatment of this neurodegenerative disorder.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Pirazóis/síntese química , Pirazolonas/síntese química , Superóxido Dismutase/antagonistas & inibidores , Animais , Barreira Hematoencefálica/metabolismo , Células CACO-2 , Permeabilidade da Membrana Celular , Inibidores das Enzimas do Citocromo P-450 , Desenho de Fármacos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Éteres/síntese química , Éteres/farmacocinética , Éteres/farmacologia , Células HEK293 , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/metabolismo , Mutação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Pirazóis/farmacocinética , Pirazóis/farmacologia , Pirazolonas/farmacocinética , Pirazolonas/farmacologia , Ratos , Ratos Sprague-Dawley , Solubilidade , Relação Estrutura-Atividade , Sulfonas/síntese química , Sulfonas/farmacocinética , Sulfonas/farmacologia , Superóxido Dismutase/genética , Superóxido Dismutase-1
4.
J Clin Invest ; 121(11): 4372-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21985783

RESUMO

Huntington disease (HD) is a devastating autosomal-dominant neurodegenerative disorder. It is caused by expansion of a CAG repeat in the first exon of the huntingtin (HTT) gene that encodes a mutant HTT protein with a polyglutamine (polyQ) expansion at the amino terminus. Here, we demonstrate that WT HTT regulates ciliogenesis by interacting through huntingtin-associated protein 1 (HAP1) with pericentriolar material 1 protein (PCM1). Loss of Htt in mouse cells impaired the retrograde trafficking of PCM1 and thereby reduced primary cilia formation. In mice, deletion of Htt in ependymal cells led to PCM1 mislocalization, alteration of the cilia layer, and hydrocephalus. Pathogenic polyQ expansion led to centrosomal accumulation of PCM1 and abnormally long primary cilia in mouse striatal cells. PCM1 accumulation in ependymal cells was associated with longer cilia and disorganized cilia layers in a mouse model of HD and in HD patients. Longer cilia resulted in alteration of the cerebrospinal fluid flow. Thus, our data indicate that WT HTT is essential for protein trafficking to the centrosome and normal ciliogenesis. In HD, hypermorphic ciliogenesis may affect signaling and neuroblast migration so as to dysregulate brain homeostasis and exacerbate disease progression.


Assuntos
Autoantígenos/genética , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Centrossomo/metabolismo , Cílios/genética , Cílios/metabolismo , Cílios/patologia , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Knockout , Microtúbulos/metabolismo , Peptídeos/genética , Transdução de Sinais , Expansão das Repetições de Trinucleotídeos
5.
J Med Chem ; 54(7): 2409-21, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21375347

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons, leading to muscle weakness, paralysis, and death, most often from respiratory failure. The only FDA-approved drug for the treatment of ALS, riluzole, only extends the median survival in patients by 2-3 months. There is an urgent need for novel therapeutic strategies for this devastating disease. Using a high-throughput screening assay targeting an ALS cultured cell model (PC12-G93A-YFP cell line), we previously identified three chemotypes that were neuroprotective. We present a further detailed analysis of one promising scaffold from that group, pyrimidine-2,4,6-triones (PYTs), characterizing a number of PYT analogues using SAR and ADME. The PYT compounds show good potency, superior ADME data, low toxicity, brain penetration, and excellent oral bioavailability. Compounds from this series show 100% efficacy in the protection assay with a good correlation in activity between the protection and protein aggregation assays. The modifications of the PYT scaffold presented here suggest that this chemical structure may be a novel drug candidate scaffold for use in clinical trials in ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Proteínas Mutantes/química , Mutação , Multimerização Proteica/efeitos dos fármacos , Pirimidinas/química , Pirimidinas/farmacologia , Superóxido Dismutase/química , Animais , Humanos , Modelos Moleculares , Proteínas Mutantes/genética , Células PC12 , Estrutura Quaternária de Proteína , Pirimidinas/síntese química , Pirimidinas/uso terapêutico , Ratos , Superóxido Dismutase/genética , Superóxido Dismutase-1
6.
Amyotroph Lateral Scler ; 12(2): 87-96, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21073276

RESUMO

The underlying cause of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder, remains unknown. However, there is strong evidence that one pathophysiological mechanism, toxic protein misfolding and/or aggregation, may trigger motor neuron dysfunction and loss. Since the clinical and pathological features of sporadic and familial ALS are indistinguishable, all forms of the disease may be better understood and ultimately treated by studying pathogenesis and therapy in models expressing mutant forms of SOD1. We developed a cellular model in which cell death depended on the expression of G93A-SOD1, a mutant form of superoxide dismutase found in familial ALS patients that produces toxic protein aggregates. This cellular model was optimized for high throughput screening to identify protective compounds from a >50,000 member chemical library. Three novel chemical scaffolds were selected for further study following screen implementation, counter-screening and secondary testing, including studies with purchased analogs. All three scaffolds blocked SOD1 aggregation in high content screening assays and data on the optimization and further characterization of these compounds will be reported separately. These data suggest that optimization of these chemicals scaffolds may produce therapeutic candidates for ALS patients.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Desenho de Fármacos , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Benzoquinonas/farmacologia , Morte Celular/efeitos dos fármacos , Citoproteção , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Lactamas Macrocíclicas/farmacologia , Leupeptinas/farmacologia , Macrolídeos/farmacologia , Proteínas Mutantes/metabolismo , Células PC12 , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Bibliotecas de Moléculas Pequenas , Superóxido Dismutase/genética
7.
Hum Mol Genet ; 19(15): 3053-67, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20494921

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder previously thought to be of primary neuronal origin, despite ubiquitous expression of mutant huntingtin (mHtt). We tested the hypothesis that mHtt expressed in astrocytes may contribute to the pathogenesis of HD. To better understand the contribution of astrocytes in HD in vivo, we developed a novel mouse model using lentiviral vectors that results in selective expression of mHtt into striatal astrocytes. Astrocytes expressing mHtt developed a progressive phenotype of reactive astrocytes that was characterized by a marked decreased expression of both glutamate transporters, GLAST and GLT-1, and of glutamate uptake. These effects were associated with neuronal dysfunction, as observed by a reduction in DARPP-32 and NR2B expression. Parallel studies in brain samples from HD subjects revealed early glial fibrillary acidic protein expression in striatal astrocytes from Grade 0 HD cases. Astrogliosis was associated with morphological changes that increased with severity of disease, from Grades 0 through 4 and was more prominent in the putamen. Combined immunofluorescence showed co-localization of mHtt in astrocytes in all striatal HD specimens, inclusive of Grade 0 HD. Consistent with the findings from experimental mice, there was a significant grade-dependent decrease in striatal GLT-1 expression from HD subjects. These findings suggest that the presence of mHtt in astrocytes alters glial glutamate transport capacity early in the disease process and may contribute to HD pathogenesis.


Assuntos
Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Doença de Huntington/metabolismo , Neostriado/patologia , Peptídeos/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Idoso , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Astrócitos/patologia , Transporte Biológico , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Regulação para Baixo , Imunofluorescência , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Doença de Huntington/patologia , Lentivirus/genética , Camundongos , Pessoa de Meia-Idade , Proteínas Mutantes/metabolismo , Neostriado/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Receptores de N-Metil-D-Aspartato/metabolismo , Fatores de Tempo
8.
J Neurochem ; 109(5): 1427-39, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19476553

RESUMO

Coenzyme Q(10) (CoQ(10)) and creatine are promising agents for neuroprotection in neurodegenerative diseases via their effects on improving mitochondrial function and cellular bioenergetics and their properties as antioxidants. We examined whether a combination of CoQ(10) with creatine can exert additive neuroprotective effects in a MPTP mouse model of Parkinson's disease, a 3-NP rat model of Huntington's disease (HD) and the R6/2 transgenic mouse model of HD. The combination of the two agents produced additive neuroprotective effects against dopamine depletion in the striatum and loss of tyrosine hydroxylase neurons in the substantia nigra pars compacta (SNpc) following chronic subcutaneous administration of MPTP. The combination treatment resulted in significant reduction in lipid peroxidation and pathologic alpha-synuclein accumulation in the SNpc neurons of the MPTP-treated mice. We also observed additive neuroprotective effects in reducing striatal lesion volumes produced by chronic subcutaneous administration of 3-NP to rats. The combination treatment showed significant effects on blocking 3-NP-induced impairment of glutathione homeostasis and reducing lipid peroxidation and DNA oxidative damage in the striatum. Lastly, the combination of CoQ(10) and creatine produced additive neuroprotective effects on improving motor performance and extending survival in the transgenic R6/2 HD mice. These findings suggest that combination therapy using CoQ(10) and creatine may be useful in the treatment of neurodegenerative diseases such as Parkinson's disease and HD.


Assuntos
Creatina/uso terapêutico , Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Ubiquinona/análogos & derivados , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , 8-Hidroxi-2'-Desoxiguanosina , Análise de Variância , Animais , Cromatografia Líquida de Alta Pressão/métodos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Quimioterapia Combinada , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Doença de Huntington/induzido quimicamente , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nitrocompostos , Doença de Parkinson/etiologia , Propionatos , Ratos , Ratos Endogâmicos Lew , Tirosina 3-Mono-Oxigenase/metabolismo , Ubiquinona/uso terapêutico , alfa-Sinucleína/metabolismo
9.
J Biol Chem ; 284(17): 11318-25, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19240033

RESUMO

Accumulation of expanded polyglutamine proteins is considered to be a major pathogenic biomarker of Huntington disease. We isolated SCAMP5 as a novel regulator of cellular accumulation of expanded polyglutamine track protein using cell-based aggregation assays. Ectopic expression of SCAMP5 augments the formation of ubiquitin-positive and detergent-resistant aggregates of mutant huntingtin (mtHTT). Expression of SCAMP5 is markedly increased in the striatum of Huntington disease patients and is induced in cultured striatal neurons by endoplasmic reticulum (ER) stress or by mtHTT. The increase of SCAMP5 impairs endocytosis, which in turn enhances mtHTT aggregation. On the contrary, down-regulation of SCAMP5 alleviates ER stress-induced mtHTT aggregation and endocytosis inhibition. Moreover, stereotactic injection into the striatum and intraperitoneal injection of tunicamycin significantly increase mtHTT aggregation in the striatum of R6/2 mice and in the cortex of N171-82Q mice, respectively. Taken together, these results suggest that exposure to ER stress increases SCAMP5 in the striatum, which positively regulates mtHTT aggregation via the endocytosis pathway.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Peptídeos/metabolismo , Regulação para Cima , Animais , Encéfalo/embriologia , Endocitose , Humanos , Proteína Huntingtina , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Ratos , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
10.
FASEB J ; 23(6): 1739-49, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19179380

RESUMO

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease characterized by selective degeneration of motor neurons and glial activation. Cell-specific transcriptional regulation induced by oxidative stress may contribute to the survival and activation of astrocytes in the face of motor neuron death. In the present study, we demonstrate an age-dependent increase in Bcl-xL and Ets-2 immunoreactivity that correlates with an increase of glial fibrillary acidic protein (GFAP)-positive cells in the ventral horn of the spinal cord in both ALS transgenic mice [mutant SOD1 (G93A)] and affected humans. Chromatin immunoprecipitation (ChIP) analysis verified that Ets-2 preferentially occupies the Ets-2 binding element in the promoter of Bcl-xL in primary astrocytes under oxidative stress conditions as well as in G93A spinal cords. Ets-2 small-interfering RNA down-regulated the transcriptional activity of Bcl-xL. In primary glial cultures, Bcl-xL overexpression and mutant SOD1 (G93A) both conferred resistance to oxidative stress-induced cell death. Our findings suggest that Ets-2 transcription factor activation of Bcl-xL gene may protect glia from constitutive oxidative stress that is thought to be a key mechanism contributing to the pathogenesis of ALS. This survival pathway may contribute to the glial survival and activation seen in the spinal cord of ALS patients.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Sobrevivência Celular/fisiologia , Neuroglia/fisiologia , Proteína Proto-Oncogênica c-ets-2/metabolismo , Proteína bcl-X/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Neuroglia/citologia , Estresse Oxidativo , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-2/genética , Interferência de RNA , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Proteína bcl-X/genética
11.
Biochim Biophys Acta ; 1782(3): 151-62, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18206128

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disorder for which there is no current therapy preventing cumulative neuronal loss. There is substantial evidence that mitochondrial dysfunction, oxidative stress, and associated caspase activity underlie the neurodegeneration observed. One potential drug therapy is the potent free radical scavenger and antioxidant cystamine, which has demonstrated significant clinical potential in models of neurodegenerative disorders and human neurological disease. This study examined the oral efficacy of cystamine in the MPTP and 6-hydroxydopamine neurotoxin models of PD. The neuroprotective effects of cystamine treatment significantly ameliorated nigral neuronal loss, preserved striatal dopaminergic projections, and improved striatal dopamine and metabolite levels, as compared to MPTP alone. Cystamine normalized striatal 8-hydroxy-2'-deoxyguanosine levels and ATP concentrations, consistent with reduced oxidative stress and improved mitochondrial function. Cystamine also protected against MPTP-induced mitochondrial loss, as identified by mitochondrial heat shock protein 70 and superoxide dismutase 2, with concomitant reductions in cytochrome c and caspase-3 activities. The neuroprotective value of cystamine was confirmed in the 6-hydroxydopamine model. Together these findings show cystamine's therapeutic benefit to reduce neuronal loss through attenuation of oxidative stress and mitochondrial dysfunction, providing the rationale for human clinical trials in PD patients.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Cistamina/uso terapêutico , Modelos Animais de Doenças , Doenças Mitocondriais/tratamento farmacológico , Neurotoxinas , Estresse Oxidativo/efeitos dos fármacos , Oxidopamina , Doença de Parkinson/tratamento farmacológico , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Avaliação Pré-Clínica de Medicamentos , Masculino , Doença de Parkinson/etiologia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia
12.
J Neurosci Res ; 86(5): 961-71, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18041090

RESUMO

Mitochondria are power organelles generating biochemical energy, ATP, in the cell. Mitochondria play a variety of roles, including integrating extracellular signals and executing critical intracellular events, such as neuronal cell survival and death. Increasing evidence suggests that a cross-talk mechanism between mitochondria and the nucleus is closely related to neuronal function and activity. Nuclear receptors (estrogen receptors, thyroid (T3) hormone receptor, peroxisome proliferators-activated receptor gamma2) and transcription factors (cAMP response binding protein, p53) have been found to target mitochondria and exert prosurvival and prodeath pathways. In this context, the regulation of mitochondrial function via the translocation of nuclear receptors and transcription factors may underlie some of the mechanisms involved in neuronal survival and death. Understanding the function of nuclear receptors and transcription factors in the mitochondria may provide important pharmacological utility in the treatment of neurodegenerative conditions. Thus, the modulation of signaling pathways via mitochondria-targeting nuclear receptors and transcription factors is rapidly emerging as a novel therapeutic target.


Assuntos
Núcleo Celular/metabolismo , Mitocôndrias/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/genética , Animais , Núcleo Celular/genética , Metabolismo Energético/genética , Humanos , Mitocôndrias/genética , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética
13.
Neurosci Lett ; 418(2): 175-80, 2007 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-17418947

RESUMO

3-Nitropropionic acid (3-NP), an irreversible inhibitor of succinate dehydrogenase, produces selective lesions in striatal neurons that resemble those observed in Huntington's disease neuropathology. In this study, we evaluated the role of peripheral bone marrow-derived cells (BMDCs) in the 3-NP-induced striatal damage by transplanting bone marrow cells with human SOD1 G93A mutation (mSOD1(G93A)) which induces amyotrophic lateral sclerosis through an unknown gain of toxicity and mitochondrial dysfunction. We assessed striatal damage after 3-NP treatment in the recipient C57BL/6 wild-type (WT) mice that received bone marrow cells from WT or mSOD1(G93A) transgenic donor mice (WT-->WT or mSOD(G93A)-->WT). After intraperitoneal injection of 3-NP, six of the eight mSOD1(G93A)-->WT mice had bilateral striatal lesions while only one out of eight WT-->WT mice had a striatal lesion. The lesion volume was significantly higher in the mSOD1(G93A)-->WT mice than in the WT-->WT mice. However, following an intrastriatal injection of 3-NP, there was no significant difference in the lesion volumes between the WT-->WT mice and mSOD1(G93A)-->WT mice. Thus, the exacerbation of 3-NP-induced striatal damage in mSOD(G93A)-->WT mice was only seen after systemic administration of 3-NP, but not after intrastriatal injection. These results demonstrate that altered SOD1 activity (mSOD(G93A)) in BMDCs affects striatal damage probably through a mechanism involving a systemic factor.


Assuntos
Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Nitrocompostos/toxicidade , Propionatos/toxicidade , Superóxido Dismutase/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Células da Medula Óssea/enzimologia , Transplante de Medula Óssea , Corpo Estriado/fisiopatologia , Predisposição Genética para Doença/genética , Humanos , Doença de Huntington/induzido quimicamente , Doença de Huntington/enzimologia , Doença de Huntington/genética , Injeções Intraperitoneais/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Degeneração Neural/fisiopatologia , Neurotoxinas , Estresse Oxidativo/genética , Superóxido Dismutase-1 , Quimeras de Transplante
14.
J Biol Chem ; 282(15): 11436-45, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17267402

RESUMO

Immunotherapy against the amyloid-beta (Abeta) peptide is a valuable potential treatment for Alzheimer disease (AD). An ideal antigen should be soluble and nontoxic, avoid the C-terminally located T-cell epitope of Abeta, and yet be capable of eliciting antibodies that recognize Abeta fibrils and neurotoxic Abeta oligomers but not the physiological monomeric species of Abeta. We have described here the construction and immunological characterization of a recombinant antigen with these features obtained by tandem multimerization of the immunodominant B-cell epitope peptide Abeta1-15 (Abeta15) within the active site loop of bacterial thioredoxin (Trx). Chimeric Trx(Abeta15)n polypeptides bearing one, four, or eight copies of Abeta15 were constructed and injected into mice in combination with alum, an adjuvant approved for human use. All three polypeptides were found to be immunogenic, yet eliciting antibodies with distinct recognition specificities. The anti-Trx(Abeta15)4 antibody, in particular, recognized Abeta42 fibrils and oligomers but not monomers and exhibited the same kind of conformational selectivity against transthyretin, an amyloidogenic protein unrelated in sequence to Abeta. We have also demonstrated that anti-Trx(Abeta15)4, which binds to human AD plaques, markedly reduces Abeta pathology in transgenic AD mice. The data indicate that a conformational epitope shared by oligomers and fibrils can be mimicked by a thioredoxin-constrained Abeta fragment repeat and identify Trx(Abeta15)4 as a promising new tool for AD immunotherapy.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Anticorpos/imunologia , Epitopos de Linfócito B/imunologia , Fragmentos de Peptídeos/imunologia , Tiorredoxinas/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Modelos Moleculares , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Tiorredoxinas/química , Tiorredoxinas/metabolismo
15.
Subcell Biochem ; 46: 205-43, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18652079

RESUMO

Significant progress has been made in identifying neuroprotective agents and their translation to patients with neurological disorders. While the direct causative pathways of neurodegeneration remain unclear, they are under great clinical and experimental investigation. There are a number of interrelated pathogenic mechanisms triggering molecular events that lead to neuronal death. One putative mechanism reported to play a prominent role in the pathogenesis of neurological diseases is impaired energy metabolism. If reduced energy stores play a role in neuronal loss, then therapeutic strategies that buffer intracellular energy levels may prevent or impede the neurodegenerative process. Recent studies suggest that impaired energy production promotes neurological disease onset and progression. Sustained ATP levels are critical to cellular homeostasis and may have both direct and indirect influence on pathogenic mechanisms associated with neurological disorders. Creatine is a critical component in maintaining cellular energy homeostasis, and its administration has been reported to be neuroprotective in a wide number of both acute and chronic experimental models of neurological disease. In the context of this chapter, we will review the experimental evidence for creatine supplementation as a neurotherapeutic strategy in patients with neurological disorders, including Huntington's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Alzheimer's disease, as well as in ischemic stroke, brain and spinal cord trauma, and epilepsy.


Assuntos
Creatina/uso terapêutico , Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Doença Aguda , Trifosfato de Adenosina/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Doença Crônica , Creatina/metabolismo , Modelos Animais de Doenças , Humanos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo
16.
Proc Natl Acad Sci U S A ; 103(50): 19176-81, 2006 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-17142323

RESUMO

Chromatin remodeling and transcription regulation are tightly controlled under physiological conditions. It has been suggested that altered chromatin modulation and transcription dysfunction may play a role in the pathogenesis of Huntington's disease (HD). Increased histone methylation, a well established mechanism of gene silencing, results in transcriptional repression. ERG-associated protein with SET domain (ESET), a histone H3 (K9) methyltransferase, mediates histone methylation. We show that ESET expression is markedly increased in HD patients and in transgenic R6/2 HD mice. Similarly, the protein level of trimethylated histone H3 (K9) was also elevated in HD patients and in R6/2 mice. We further demonstrate that both specificity protein 1 (Sp1) and specificity protein 3 (Sp3) act as transcriptional activators of the ESET promoter in neurons and that mithramycin, a clinically approved guanosine-cytosine-rich DNA binding antitumor antibiotic, interferes with the DNA binding of these Sp family transcription factors, suppressing basal ESET promoter activity in a dose dependent manner. The combined pharmacological treatment with mithramycin and cystamine down-regulates ESET gene expression and reduces hypertrimethylation of histone H3 (K9). This polytherapy significantly ameliorated the behavioral and neuropathological phenotype in the R6/2 mice and extended survival over 40%, well beyond any existing reported treatment in HD mice. Our data suggest that modulation of gene silencing mechanisms, through regulation of the ESET gene is important to neuronal survival and, as such, may be a promising treatment in HD patients.


Assuntos
Expressão Gênica , Histonas/metabolismo , Doença de Huntington/metabolismo , Proteínas Metiltransferases/metabolismo , Idoso , Animais , Cistamina/uso terapêutico , Feminino , Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Metilação , Camundongos , Pessoa de Meia-Idade , Fenótipo , Plicamicina/uso terapêutico , Regiões Promotoras Genéticas/genética , Proteínas Metiltransferases/genética , Fator de Transcrição Sp3/metabolismo , Taxa de Sobrevida , Regulação para Cima
17.
FASEB J ; 20(13): 2375-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17012241

RESUMO

Cyclooxygenase-2 (COX-2) has been implicated in neuronal survival and death. However, the precise regulatory mechanisms involved in COX-2 function are unclear. In the present study we found that COX-2 is induced in response to glutathione depletion-induced oxidative stress in primary cortical neurons. Two proximal specific Sp1 and Sp3 binding sites are responsible for the COX-2 promoter activity under normal as well as oxidative stress conditions through enhanced Sp1 and Sp3 DNA binding activity. Site-directed mutagenesis confirmed that -268/-267 positions serve as specific Sp1 and Sp3 recognition sites under oxidative stress. Enforced expression of Sp1 and Sp3 using HSV vectors increased the promoter activity, transcription, and protein level of COX-2 in cortical neurons. The dominant negative form of Sp1 abrogated the oxidative stress-induced promoter activity and expression of COX-2. We also demonstrated that adenovirus-mediated COX-2 gene delivery protected neurons from DNA damage induced by oxidative, genotoxic, and excitotoxic stresses and by ischemic injury. Moreover, COX-2(-/-) cortical neurons were more susceptible to DNA damage-induced cell death. These results indicate that in primary neurons Sp1 and Sp3 play an essential role in the modulation of COX-2 transcription, which mediates neuronal homeostasis and survival by preventing DNA damage in response to neuronal stress.


Assuntos
Córtex Cerebral/enzimologia , Ciclo-Oxigenase 2/genética , Dano ao DNA , Neurônios/fisiologia , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp2/metabolismo , Animais , Sequência de Bases , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Sobrevivência Celular , Clonagem Molecular , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/deficiência , Primers do DNA , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/patologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Biochim Biophys Acta ; 1762(6): 616-26, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16647250

RESUMO

There is substantial evidence that a bioenergetic defect may play a role in the pathogenesis of Huntington's Disease (HD). A potential therapy for remediating defective energy metabolism is the mitochondrial cofactor, coenzyme Q10 (CoQ10). We have reported that CoQ10 is neuroprotective in the R6/2 transgenic mouse model of HD. Based upon the encouraging results of the CARE-HD trial and recent evidence that high-dose CoQ10 slows the progressive functional decline in Parkinson's disease, we performed a dose ranging study administering high levels of CoQ10 from two commercial sources in R6/2 mice to determine enhanced efficacy. High dose CoQ10 significantly extended survival in R6/2 mice, the degree of which was dose- and source-dependent. CoQ10 resulted in a marked improvement in motor performance and grip strength, with a reduction in weight loss, brain atrophy, and huntingtin inclusions in treated R6/2 mice. Brain levels of CoQ10 and CoQ9 were significantly lower in R6/2 mice, in comparison to wild type littermate control mice. Oral administration of CoQ10 elevated CoQ10 plasma levels and significantly increased brain levels of CoQ9, CoQ10, and ATP in R6/2 mice, while reducing 8-hydroxy-2-deoxyguanosine concentrations, a marker of oxidative damage. We demonstrate that high-dose administration of CoQ10 exerts a greater therapeutic benefit in a dose dependent manner in R6/2 mice than previously reported and suggest that clinical trials using high dose CoQ10 in HD patients are warranted.


Assuntos
Doença de Huntington/tratamento farmacológico , Ubiquinona/análogos & derivados , 8-Hidroxi-2'-Desoxiguanosina , Trifosfato de Adenosina/metabolismo , Animais , Peso Corporal , Coenzimas , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Desoxiguanosina/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteína Huntingtina , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neostriado/citologia , Neostriado/patologia , Proteínas do Tecido Nervoso/imunologia , Fármacos Neuroprotetores , Proteínas Nucleares/imunologia , Teste de Desempenho do Rota-Rod , Resultado do Tratamento , Ubiquinona/administração & dosagem , Ubiquinona/sangue , Ubiquinona/metabolismo , Ubiquinona/uso terapêutico
19.
J Biol Chem ; 281(24): 16672-80, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16595660

RESUMO

Interactions between mutant huntingtin (Htt) and a variety of transcription factors including specificity proteins (Sp) have been suggested as a central mechanism in Huntington disease (HD). However, the transcriptional activity induced by Htt in neurons that triggers neuronal death has yet to be fully elucidated. In the current study, we characterized the relationship of Sp1 to Htt protein aggregation and neuronal cell death. We found increased levels of Sp1 in neuronal-like PC12 cells expressing mutant Htt, primary striatal neurons, and brain tissue of HD transgenic mice. Sp1 levels were also elevated when 3-nitropropionate (3-NP) was used to induce cell death in PC12 cells. To assess the effects of knocking down Sp1 in HD pathology, we used Sp1 siRNA, a heterozygous Sp1 knock-out mouse, and mithramycin A, a DNA-intercalating agent that inhibits Sp1 function. The three approaches consistently yielded reduced levels of Sp1 which ameliorated toxicity caused by either mutant Htt or 3-NP. In addition, when HD mice were crossed with Sp1 heterozygous knock-out mice, the resulting offspring did not experience the loss of dopamine D2 receptor mRNA characteristic of HD mice, and survived longer than their HD counterparts. Our data suggest that enhancement of transcription factor Sp1 contributes to the pathology of HD and demonstrates that its suppression is beneficial.


Assuntos
Doença de Huntington/genética , Fármacos Neuroprotetores/farmacologia , Fator de Transcrição Sp1/biossíntese , Fator de Transcrição Sp1/fisiologia , Regulação para Cima , Animais , Modelos Animais de Doenças , Feminino , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Células PC12 , Ratos
20.
J Biol Chem ; 280(49): 40398-401, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16207717

RESUMO

Cyclic AMP response element-binding protein (CREB) is a widely expressed transcription factor whose role in neuronal protection is now well established. Here we report that CREB is present in the mitochondrial matrix of neurons and that it binds directly to cyclic AMP response elements (CREs) found within the mitochondrial genome. Disruption of CREB activity in the mitochondria decreases the expression of a subset of mitochondrial genes, including the ND5 subunit of complex I, down-regulates complex I-dependent mitochondrial respiration, and increases susceptibility to 3-nitropropionic acid, a mitochondrial toxin that induces a clinical and pathological phenotype similar to Huntington disease. These results demonstrate that regulation of mitochondrial gene expression by mitochondrial CREB, in part, underlies the protective effects of CREB and raise the possibility that decreased mitochondrial CREB activity contributes to the mitochondrial dysfunction and neuronal loss associated with neurodegenerative disorders.


Assuntos
Encéfalo/ultraestrutura , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Mitocôndrias/química , Neurônios/fisiologia , Animais , Sequência de Bases , Sobrevivência Celular , Córtex Cerebral/citologia , AMP Cíclico , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , DNA Mitocondrial/genética , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Dados de Sequência Molecular , Doenças Neurodegenerativas , Neurônios/ultraestrutura , Nitrocompostos/farmacologia , Consumo de Oxigênio/fisiologia , Fosforilação , Propionatos/farmacologia , Ratos , Elementos de Resposta , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA