Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging (Albany NY) ; 13(3): 3269-3289, 2021 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-33550278

RESUMO

Geroprotectors are compounds that slow the biological aging process in model organisms and may therefore extend healthy lifespan in humans. It is hypothesized that they do so by preserving the more youthful function of multiple organ systems. However, this hypothesis has rarely been tested in any organisms besides C. elegans and D. melanogaster. To determine if two life-extending compounds for Drosophila maintain a more youthful phenotype in old mice, we asked if they had anti-aging effects in both the brain and kidney. We utilized rapidly aging senescence-accelerated SAMP8 mice to investigate age-associated protein level alterations in these organs. The test compounds were two cognition-enhancing Alzheimer's disease drug candidates, J147 and CMS121. Mice were fed the compounds in the last quadrant of their lifespan, when they have cognitive deficits and are beginning to develop CKD. Both compounds improved physiological markers for brain and kidney function. However, these two organs had distinct, tissue-specific protein level alterations that occurred with age, but in both cases, drug treatments restored a more youthful level. These data show that geroprotective AD drug candidates J147 and CMS121 prevent age-associated disease in both brain and kidney, and that their apparent mode of action in each tissue is distinct.


Assuntos
Envelhecimento/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Rim/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Doença de Alzheimer , Animais , Caenorhabditis elegans , Curcumina/análogos & derivados , Curcumina/farmacologia , Modelos Animais de Doenças , Drosophila melanogaster , Feminino , Masculino , Camundongos , Insuficiência Renal Crônica
2.
Alzheimers Res Ther ; 12(1): 75, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32560738

RESUMO

BACKGROUND: All cells accumulate insoluble protein aggregates throughout their lifespan. While many studies have characterized the canonical disease-associated protein aggregates, such as those associated with amyloid plaques, additional, undefined proteins aggregate in the brain and may be directly associated with disease and lifespan. METHODS: A proteomics approach was used to identify a large subset of insoluble proteins in the mild cognitively impaired (MCI) and Alzheimer's disease (AD) human brain. Cortical samples from control, MCI, and AD patients were separated into detergent-soluble and detergent-insoluble fractions, and high-resolution LC/MS/MS technology was used to determine which proteins became more insoluble in the disease state. Bioinformatics analyses were used to determine if the alteration of protein aggregation between AD and control patients was associated with any specific biological process. Western blots were used to validate the proteomics data and to assess the levels of secondary protein modifications in MCI and AD. RESULTS: There was a stage-dependent increase in detergent-insoluble proteins, with more extreme changes occurring in the AD cohort. Glycolysis was the most significantly overrepresented gene ontology biological process associated with the alteration of protein aggregation between AD and control patients. It was further shown that many low molecular weight proteins that were enriched in the AD brain were also highly aggregated, migrating on SDS-PAGE far above their predicted molecular masses. Glucose-6-phosphate isomerase, ubiquitin carboxyl-terminal hydrolase isoenzyme L1 (UCHL1/PARK5), and the DNA damage repair enzyme KU70 were among the top insoluble proteins identified by proteomics and validated by Western blot to be increased in the insoluble fractions of both MCI and AD brain samples. CONCLUSIONS: Diverse proteins became more detergent-insoluble in the brains of both MCI and AD patients compared to age-matched controls, suggesting that multiple proteins aggregate in these diseases, likely posing a direct toxic insult to neurons. Furthermore, detergent-insoluble proteins included those with important biological activities for critical cellular processes such as energetics, proteolysis, and DNA damage repair. Thus, reduced protein solubility likely promotes aggregation and limits functionality, reducing the efficiency of multiple aspects of cell physiology. Pharmaceutical interventions that increase autophagy may provide a useful therapeutic treatment to combat protein aggregation.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Encéfalo , Humanos , Placa Amiloide , Espectrometria de Massas em Tandem
3.
Mol Neurobiol ; 56(11): 7719-7730, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31104297

RESUMO

Finding a therapy for Alzheimer's disease (AD) is perhaps the greatest challenge for modern medicine. The chemical scaffolds of many drugs in the clinic today are based upon natural products from plants, yet Cannabis has not been extensively examined as a source of potential AD drug candidates. Here, we determine if a number of non-psychoactive cannabinoids are neuroprotective in a novel pre-clinical AD and neurodegeneration drug-screening platform that is based upon toxicities associated with the aging brain. This drug discovery paradigm has yielded several compounds in or approaching clinical trials for AD. Eleven cannabinoids were assayed for neuroprotection in assays that recapitulate proteotoxicity, loss of trophic support, oxidative stress, energy loss, and inflammation. These compounds were also assayed for their ability to remove intraneuronal amyloid and subjected to a structure-activity relationship analysis. Pairwise combinations were assayed for their ability to synergize to produce neuroprotective effects that were greater than additive. Nine of the 11 cannabinoids have the ability to protect cells in four distinct phenotypic neurodegeneration screening assays, including those using neurons that lack CB1 and CB2 receptors. They are able to remove intraneuronal Aß, reduce oxidative damage, and protect from the loss of energy or trophic support. Structure-activity relationship (SAR) data show that functional antioxidant groups such as aromatic hydroxyls are necessary but not sufficient for neuroprotection. Therefore, there is a need to focus upon CB1 agonists that have these functionalities if neuroprotection is the goal. Pairwise combinations of THC and CBN lead to a synergistic neuroprotective interaction. Together, these results significantly extend the published data by showing that non-psychoactive cannabinoids are potential lead drug candidates for AD and other neurodegenerative diseases.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Canabinoides/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Animais , Canabinoides/química , Canabinoides/farmacologia , Linhagem Celular , Sinergismo Farmacológico , Humanos , Camundongos , Resultado do Tratamento
4.
Aging Cell ; 17(2)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29316249

RESUMO

Aging is a major driving force underlying dementia, such as that caused by Alzheimer's disease (AD). While the idea of targeting aging as a therapeutic strategy is not new, it remains unclear how closely aging and age-associated diseases are coupled at the molecular level. Here, we discover a novel molecular link between aging and dementia through the identification of the molecular target for the AD drug candidate J147. J147 was developed using a series of phenotypic screening assays mimicking disease toxicities associated with the aging brain. We have previously demonstrated the therapeutic efficacy of J147 in several mouse models of AD. Here, we identify the mitochondrial α-F1 -ATP synthase (ATP5A) as a target for J147. By targeting ATP synthase, J147 causes an increase in intracellular calcium leading to sustained calcium/calmodulin-dependent protein kinase kinase ß (CAMKK2)-dependent activation of the AMPK/mTOR pathway, a canonical longevity mechanism. Accordingly, modulation of mitochondrial processes by J147 prevents age-associated drift of the hippocampal transcriptome and plasma metabolome in mice and extends lifespan in drosophila. Our results link aging and age-associated dementia through ATP synthase, a molecular drug target that can potentially be exploited for the suppression of both. These findings demonstrate that novel screens for new AD drug candidates identify compounds that act on established aging pathways, suggesting an unexpectedly close molecular relationship between the two.


Assuntos
Envelhecimento/genética , Demência/genética , Mitocôndrias/enzimologia , ATPases Mitocondriais Próton-Translocadoras/genética , Humanos , Mitocôndrias/metabolismo
5.
J Gerontol A Biol Sci Med Sci ; 73(3): 299-307, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28575152

RESUMO

Alzheimer's disease (AD) is rarely addressed in the context of aging even though there is an overlap in pathology. We previously used a phenotypic screening platform based on old age-associated brain toxicities to identify the flavonol fisetin as a potential therapeutic for AD and other age-related neurodegenerative diseases. Based on earlier results with fisetin in transgenic AD mice, we hypothesized that fisetin would be effective against brain aging and cognitive dysfunction in rapidly aging senescence-accelerated prone 8 (SAMP8) mice, a model for sporadic AD and dementia. An integrative approach was used to correlate protein expression and metabolite levels in the brain with cognition. It was found that fisetin reduced cognitive deficits in old SAMP8 mice while restoring multiple markers associated with impaired synaptic function, stress, and inflammation. These results provide further evidence for the potential benefits of fisetin for the treatment of age-related neurodegenerative diseases.


Assuntos
Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Flavonoides/farmacologia , Envelhecimento/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Eicosanoides/metabolismo , Flavonóis , Imuno-Histoquímica , Aprendizagem em Labirinto/efeitos dos fármacos , Metabolômica/métodos , Camundongos , Camundongos Transgênicos , Percepção Visual/efeitos dos fármacos
6.
Alzheimers Res Ther ; 9(1): 50, 2017 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-28709449

RESUMO

BACKGROUND: CAD-31 is an Alzheimer's disease (AD) drug candidate that was selected on the basis of its ability to stimulate the replication of human embryonic stem cell-derived neural precursor cells as well as in APPswe/PS1ΔE9 AD mice. To move CAD-31 toward the clinic, experiments were undertaken to determine its neuroprotective and pharmacological properties, as well as to assay its therapeutic efficacy in a rigorous mouse model of AD. RESULTS: CAD-31 has potent neuroprotective properties in six distinct nerve cell assays that mimic toxicities observed in the old brain. Pharmacological and preliminary toxicological studies show that CAD-31 is brain-penetrant and likely safe. When fed to old, symptomatic APPswe/PS1ΔE9 AD mice starting at 10 months of age for 3 additional months in a therapeutic model of the disease, there was a reduction in the memory deficit and brain inflammation, as well as an increase in the expression of synaptic proteins. Small-molecule metabolic data from the brain and plasma showed that the major effect of CAD-31 is centered on fatty acid metabolism and inflammation. Pathway analysis of gene expression data showed that CAD-31 had major effects on synapse formation and AD energy metabolic pathways. CONCLUSIONS: All of the multiple physiological effects of CAD-31 were favorable in the context of preventing some of the toxic events in old age-associated neurodegenerative diseases.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Antipsicóticos/uso terapêutico , Ácidos Graxos/metabolismo , Inflamação/tratamento farmacológico , Inflamação/etiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Antipsicóticos/síntese química , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Medo/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Presenilina-1/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
7.
Aging (Albany NY) ; 7(11): 937-55, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26564964

RESUMO

Because age is the greatest risk factor for sporadic Alzheimer's disease (AD), phenotypic screens based upon old age-associated brain toxicities were used to develop the potent neurotrophic drug J147. Since certain aspects of aging may be primary cause of AD, we hypothesized that J147 would be effective against AD-associated pathology in rapidly aging SAMP8 mice and could be used to identify some of the molecular contributions of aging to AD. An inclusive and integrative multiomics approach was used to investigate protein and gene expression, metabolite levels, and cognition in old and young SAMP8 mice. J147 reduced cognitive deficits in old SAMP8 mice, while restoring multiple molecular markers associated with human AD, vascular pathology, impaired synaptic function, and inflammation to those approaching the young phenotype. The extensive assays used in this study identified a subset of molecular changes associated with aging that may be necessary for the development of AD.


Assuntos
Envelhecimento , Doença de Alzheimer/etiologia , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/análise , Animais , Comportamento Animal/efeitos dos fármacos , Barreira Hematoencefálica , Eicosanoides/metabolismo , Glutationa/metabolismo , Hipocampo/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Aprendizagem em Labirinto/efeitos dos fármacos , Metabolômica , Camundongos , Transcriptoma
8.
ACS Chem Neurosci ; 5(7): 503-13, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-24902068

RESUMO

There are no disease-modifying drugs for any old age associated neurodegenerative disease or stroke. This is at least in part due to the failure of drug developers to recognize that the vast majority of neurodegenerative diseases arise from a confluence of multiple toxic insults that accumulate during normal aging and interact with genetic and environmental risk factors. Thus, it is unlikely that the current single target approach based upon rare dominant mutations or even a few preselected targets is going to yield useful drugs for these conditions. Therefore, the identification of drug candidates for neurodegeneration should be based upon their efficacy in phenotypic screening assays that reflect the biology of the aging brain, not a single, preselected target. It is argued here that this approach to drug discovery is the most likely to produce safe and effective drugs for neurodegenerative diseases.


Assuntos
Envelhecimento/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Doenças Neurodegenerativas/tratamento farmacológico , Fenótipo , Envelhecimento/fisiologia , Animais , Encéfalo/fisiopatologia , Avaliação Pré-Clínica de Medicamentos/instrumentação , Humanos , Doenças Neurodegenerativas/fisiopatologia , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia
9.
Aging Cell ; 13(2): 379-90, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24341874

RESUMO

Alzheimer's disease (AD) is the most common type of dementia. It is the only one of the top ten causes of death in the USA for which prevention strategies have not been developed. Although AD has traditionally been associated with the deposition of amyloid ß plaques and tau tangles, it is becoming increasingly clear that it involves disruptions in multiple cellular systems. Therefore, it is unlikely that hitting a single target will result in significant benefits to patients with AD. An alternative approach is to identify molecules that have multiple biological activities that are relevant to the disease. Fisetin is a small, orally active molecule which can act on many of the target pathways implicated in AD. We show here that oral administration of fisetin to APPswe/PS1dE9 double transgenic AD mice from 3 to 12 months of age prevents the development of learning and memory deficits. This correlates with an increase in ERK phosphorylation along with a decrease in protein carbonylation, a marker of oxidative stress. Importantly, fisetin also reduces the levels of the cyclin-dependent kinase 5 (Cdk5) activator p35 cleavage product, p25, in both control and AD brains. Elevated levels of p25 relative to p35 cause dysregulation of Cdk5 activity leading to neuroinflammation and neurodegeneration. These fisetin-dependent changes correlate with additional anti-inflammatory effects, including alterations in global eicosanoid synthesis, and the maintenance of markers of synaptic function in the AD mice. Together, these results suggest that fisetin may provide a new approach to the treatment of AD.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Cognição/efeitos dos fármacos , Flavonoides/farmacologia , Inflamação/patologia , Proteínas do Tecido Nervoso/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Animais , Ácido Araquidônico/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonóis , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Hipocampo/patologia , Humanos , Inflamação/complicações , Inflamação/metabolismo , Inflamação/fisiopatologia , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Fosfotransferases , Carbonilação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/metabolismo
10.
J Neurosci ; 33(25): 10512-25, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23785163

RESUMO

The accumulation of intracellular ß amyloid (Aß) may be one of the factors leading to neuronal cell death in Alzheimer's disease (AD). Using a pyrazole called CNB-001, which was selected for its ability to reduce intracellular Aß, we show that the activation of the eIF2α/ATF4 arm of the unfolded protein response is sufficient to degrade aggregated intracellular Aß. CNB-001 is a potent inhibitor of 5-lipoxygenase (5-LOX), decreases 5-LOX expression, and increases proteasome activity. 5-LOX inhibition induces eIF2α and PERK (protein kinase R-like extracellular signal-regulated kinase) phosphorylation, and HSP90 and ATF4 levels. When fed to AD transgenic mice, CNB-001 also increases eIF2α phosphorylation and HSP90 and ATF4 levels, and limits the accumulation of soluble Aß and ubiquitinated aggregated proteins. Finally, CNB-001 maintains the expression of synapse-associated proteins and improves memory. Therefore, 5-LOX metabolism is a key element in the promotion of endoplasmic reticulum dysfunction, and its inhibition under conditions of stress is sufficient to reduce proteotoxicity both in vivo and in vitro.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/toxicidade , Araquidonato 5-Lipoxigenase/metabolismo , Curcumina/análogos & derivados , Inibidores de Lipoxigenase/farmacologia , Pirazóis/farmacologia , Fator 4 Ativador da Transcrição/metabolismo , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Animais , Apomorfina/farmacologia , Comportamento Animal/fisiologia , Western Blotting , Curcumina/farmacologia , Agonistas de Dopamina/farmacologia , Eletroforese em Gel de Poliacrilamida , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/genética , Dobramento de Proteína , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ubiquitina/metabolismo
11.
Alzheimers Res Ther ; 5(3): 25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23673233

RESUMO

INTRODUCTION: Despite years of research, there are no disease-modifying drugs for Alzheimer's disease (AD), a fatal, age-related neurodegenerative disorder. Screening for potential therapeutics in rodent models of AD has generally relied on testing compounds before pathology is present, thereby modeling disease prevention rather than disease modification. Furthermore, this approach to screening does not reflect the clinical presentation of AD patients which could explain the failure to translate compounds identified as beneficial in animal models to disease modifying compounds in clinical trials. Clearly a better approach to pre-clinical drug screening for AD is required. METHODS: To more accurately reflect the clinical setting, we used an alternative screening strategy involving the treatment of AD mice at a stage in the disease when pathology is already advanced. Aged (20-month-old) transgenic AD mice (APP/swePS1ΔE9) were fed an exceptionally potent, orally active, memory enhancing and neurotrophic molecule called J147. Cognitive behavioral assays, histology, ELISA and Western blotting were used to assay the effect of J147 on memory, amyloid metabolism and neuroprotective pathways. J147 was also investigated in a scopolamine-induced model of memory impairment in C57Bl/6J mice and compared to donepezil. Details on the pharmacology and safety of J147 are also included. RESULTS: Data presented here demonstrate that J147 has the ability to rescue cognitive deficits when administered at a late stage in the disease. The ability of J147 to improve memory in aged AD mice is correlated with its induction of the neurotrophic factors NGF (nerve growth factor) and BDNF (brain derived neurotrophic factor) as well as several BDNF-responsive proteins which are important for learning and memory. The comparison between J147 and donepezil in the scopolamine model showed that while both compounds were comparable at rescuing short term memory, J147 was superior at rescuing spatial memory and a combination of the two worked best for contextual and cued memory. CONCLUSION: J147 is an exciting new compound that is extremely potent, safe in animal studies and orally active. J147 is a potential AD therapeutic due to its ability to provide immediate cognition benefits, and it also has the potential to halt and perhaps reverse disease progression in symptomatic animals as demonstrated in these studies.

12.
J Med Chem ; 55(1): 378-89, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22192055

RESUMO

Many factors are implicated in age-related central nervous system (CNS) disorders, making it unlikely that modulating only a single factor will provide effective treatment. Perhaps a better approach is to identify small molecules that have multiple biological activities relevant to the maintenance of brain function. Recently, we identified an orally active, neuroprotective, and cognition-enhancing molecule, the flavonoid fisetin, that is effective in several animal models of CNS disorders. Fisetin has direct antioxidant activity and can also increase the intracellular levels of glutathione (GSH), the major endogenous antioxidant. In addition, fisetin has both neurotrophic and anti-inflammatory activity. However, its relatively high EC(50) in cell based assays, low lipophilicity, high topological polar surface area (tPSA), and poor bioavailability suggest that there is room for medicinal chemical improvement. Here we describe a multitiered approach to screening that has allowed us to identify fisetin derivatives with significantly enhanced activity in an in vitro neuroprotection model while at the same time maintaining other key activities.


Assuntos
Anti-Inflamatórios não Esteroides/síntese química , Antioxidantes/síntese química , Flavonoides/síntese química , Fármacos Neuroprotetores/síntese química , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Chalconas/síntese química , Chalconas/química , Chalconas/farmacologia , Flavonoides/química , Flavonoides/farmacologia , Flavonóis , Glutationa/metabolismo , Humanos , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/biossíntese , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Quinolinas/síntese química , Quinolinas/química , Quinolinas/farmacologia , Ratos , Relação Estrutura-Atividade
13.
PLoS One ; 6(12): e27865, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194796

RESUMO

Currently, the major drug discovery paradigm for neurodegenerative diseases is based upon high affinity ligands for single disease-specific targets. For Alzheimer's disease (AD), the focus is the amyloid beta peptide (Aß) that mediates familial Alzheimer's disease pathology. However, given that age is the greatest risk factor for AD, we explored an alternative drug discovery scheme that is based upon efficacy in multiple cell culture models of age-associated pathologies rather than exclusively amyloid metabolism. Using this approach, we identified an exceptionally potent, orally active, neurotrophic molecule that facilitates memory in normal rodents, and prevents the loss of synaptic proteins and cognitive decline in a transgenic AD mouse model.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Cognição/efeitos dos fármacos , Curcumina/análogos & derivados , Fatores de Crescimento Neural/farmacologia , Pirazóis/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Curcumina/química , Curcumina/farmacologia , Modelos Animais de Doenças , Proteínas de Choque Térmico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Inflamação/patologia , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Oxidantes/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Pirazóis/química , Ratos , Solubilidade/efeitos dos fármacos , Relação Estrutura-Atividade , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
PLoS One ; 6(6): e21226, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738623

RESUMO

The elevated glycation of macromolecules by the reactive dicarbonyl and α-oxoaldehyde methylglyoxal (MG) has been associated with diabetes and its complications. We have identified a rare flavone, fisetin, which increases the level and activity of glyoxalase 1, the enzyme required for the removal of MG, as well as the synthesis of its essential co-factor, glutathione. It is shown that fisetin reduces two major complications of diabetes in Akita mice, a model of type 1 diabetes. Although fisetin had no effect on the elevation of blood sugar, it reduced kidney hypertrophy and albuminuria and maintained normal levels of locomotion in the open field test. This correlated with a reduction in proteins glycated by MG in the blood, kidney and brain of fisetin-treated animals along with an increase in glyoxalase 1 enzyme activity and an elevation in the expression of the rate-limiting enzyme for the synthesis of glutathione, a co-factor for glyoxalase 1. The expression of the receptor for advanced glycation end products (RAGE), serum amyloid A and serum C-reactive protein, markers of protein oxidation, glycation and inflammation, were also increased in diabetic Akita mice and reduced by fisetin. It is concluded that fisetin lowers the elevation of MG-protein glycation that is associated with diabetes and ameliorates multiple complications of the disease. Therefore, fisetin or a synthetic derivative may have potential therapeutic use for the treatment of diabetic complications.


Assuntos
Complicações do Diabetes/prevenção & controle , Diabetes Mellitus/tratamento farmacológico , Flavonoides/uso terapêutico , Glicosilação/efeitos dos fármacos , Aldeído Pirúvico/metabolismo , Animais , Ansiedade/tratamento farmacológico , Linhagem Celular , Diabetes Mellitus/metabolismo , Flavonóis , Camundongos , Camundongos Endogâmicos C57BL , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Proteína Amiloide A Sérica/metabolismo
15.
Hum Mol Genet ; 20(2): 261-70, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20952447

RESUMO

Huntington's disease (HD) is an inherited, progressive and ultimately fatal neurodegenerative disorder that is characterized by psychiatric, cognitive and motor symptoms. Among the pathways implicated in HD are those involving mitogen-activated protein kinase signaling and particularly the Ras-extracellular signal-regulated kinase (ERK) cascade. Studies in both cells and animal models suggest that ERK activation might provide a novel therapeutic target for the treatment of HD but compounds that specifically activate ERK are few. To test the hypothesis that pharmaceutical activation of ERK might be protective for HD, a polyphenol, fisetin, which was previously shown to activate the Ras-ERK cascade, was tested in three different models of HD: PC12 cells expressing mutant Httex1 under the control of an inducible promoter, Drosophila expressing mutant Httex1 and the R6/2 mouse model of HD. The results indicate that fisetin can reduce the impact of mutant huntingtin in each of these disease models. Prompted by this observation, we determined that the related polyphenol, resveratrol, also activates ERK and is protective in HD models. Notably, although more than a dozen small molecule inhibitors of ERK activation are in clinical trials, very few small molecule activators of ERK signaling are reported. Thus, fisetin, resveratrol and related compounds might be useful for the treatment of HD by virtue of their unique ability to activate ERK.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Doença de Huntington/enzimologia , Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Animais , Modelos Animais de Doenças , Drosophila/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Flavonóis , Dosagem de Genes/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Huntingtina , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Células PC12 , Ratos , Resveratrol , Análise de Sobrevida
16.
J Neurochem ; 113(2): 502-14, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20132475

RESUMO

Lactate and acidosis increase infarct size in humans and in animal models of cerebral ischemia but the mechanisms by which they exert their neurotoxic effects are poorly understood. Oxidative glutamate toxicity is a form of nerve cell death, wherein glutamate inhibits cystine uptake via the cystine/glutamate antiporter system leading to glutathione depletion, accumulation of reactive oxygen species and, ultimately, programmed cell death. Using the hippocampal cell line, HT22, we show that lactate and acidosis exacerbate oxidative glutamate toxicity and further decrease glutathione levels. Acidosis but not lactate inhibits system , whereas both acidosis and lactate inhibit the enzymatic steps of glutathione synthesis downstream of cystine uptake. In contrast, when glutathione synthesis is completely inhibited by cystine-free medium, acidosis partially protects against glutathione depletion and cell death. Both effects of acidosis are also present in primary neuronal and astrocyte cultures. Furthermore, we show that some neuroprotective compounds are much less effective in the presence of lactacidosis. Our findings indicate that lactacidosis modulates glutathione metabolism and neuronal cell death. Furthermore, lactacidosis may interfere with the action of some neuroprotective drugs rendering these less likely to be therapeutically effective in cerebral ischemia.


Assuntos
Acidose/metabolismo , Ácido Glutâmico/farmacologia , Glutationa/metabolismo , Ácido Láctico/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Acidose/induzido quimicamente , Animais , Antioxidantes/farmacologia , Astrócitos , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Cistina/metabolismo , Cistina/farmacologia , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Flavonoides/farmacologia , Flavonóis , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Ácido Láctico/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Quercetina/farmacologia , Isótopos de Enxofre/metabolismo , Fatores de Tempo , Trítio/metabolismo
17.
J Neurochem ; 105(4): 1336-45, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18208543

RESUMO

The plant polyphenolic curcumin alters the response of nerve cells to some forms of toxic stress. The steroid-like compound, cyclohexyl bisphenol A, has broad neuroprotective properties that are very distinct from those of curcumin. To incorporate both families of biological activities into a single molecule, a pyrazole derivative of curcumin, called CNB-001, was synthesized. CNB-001 acquires a new activity and is far superior in neuroprotection assays to either parental molecule, but retains some of the properties of both. It is neuroprotective in cell culture assays for trophic factor withdrawal, oxidative stress, excitotoxicity, and glucose starvation, as well as toxicity from both intracellular and extracellular amyloid. While the creation of CNB-001 was based upon an uncommon approach to drug design, it has the potential of a lead drug candidate for treating multiple conditions involving nerve cell death.


Assuntos
Curcumina/análogos & derivados , Curcumina/farmacologia , Fármacos Neuroprotetores/farmacologia , Pirazóis/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Curcumina/síntese química , Relação Dose-Resposta a Droga , Camundongos , Fármacos Neuroprotetores/síntese química , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Pirazóis/síntese química , Ratos
18.
J Biol Chem ; 282(42): 30523-34, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17761673

RESUMO

Peroxiredoxins (Prxs) are a ubiquitously expressed family of thiol peroxidases that reduce hydrogen peroxide, peroxynitrite, and hydroperoxides using a highly conserved cysteine. There is substantial evidence that oxidative stress elicited by amyloid beta (Abeta) accumulation is a causative factor in the pathogenesis of Alzheimer disease (AD). Here we show that Abeta-resistant PC12 cell lines exhibit increased expression of multiple Prx isoforms with reduced cysteine oxidation. Abeta-resistant PC12 cells also display higher levels of thioredoxin and thioredoxin reductase, two enzymes critical for maintaining Prx activity. PC12 cells and rat primary hippocampal neurons transfected with wild type Prx1 exhibit increased Abeta resistance, whereas mutant Prx1, lacking a catalytic cysteine, confers no protection. Using an antibody that specifically recognizes sulfinylated and sulfonylated Prxs, it is demonstrated that primary rat cortical nerve cells exposed to Abeta display a time-dependent increase in cysteine oxidation of the catalytic site of Prxs that can be blocked by the addition of the thiol-antioxidant N-acetylcysteine. In support of previous findings, expression of Prx1 is higher in post-mortem human AD cortex tissues than in age-matched controls. In addition, two-dimensional gel electrophoresis and mass spectrometry analysis revealed that Prx2 exists in a more oxidized state in AD brains than in control brains. These findings suggest that increased Prx expression and resistance to sulfhydryl oxidation in Abeta-resistant nerve cells is a compensatory response to the oxidative stress initiated by chronic pro-oxidant Abeta exposure.


Assuntos
Doença de Alzheimer/etnologia , Peptídeos beta-Amiloides/metabolismo , Neurônios/enzimologia , Peroxirredoxinas/metabolismo , Acetilcisteína/farmacologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Domínio Catalítico/genética , Córtex Cerebelar/enzimologia , Córtex Cerebelar/patologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica , Hipocampo/enzimologia , Hipocampo/patologia , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Peroxirredoxinas/antagonistas & inibidores , Peroxirredoxinas/genética , Ratos , Compostos de Sulfidrila/metabolismo
19.
Antioxid Redox Signal ; 8(3-4): 661-70, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16677109

RESUMO

Hydrogen sulfide (H2S) is a neuromodulator in the brain and a relaxant for smooth muscle. H2S protects primary cortical neurons from oxidative stress by increasing the intracellular concentrations of glutathione, the major antioxidant in cells. However, changes in glutathione alone are not sufficient to account for full protection in all types of nerve cells. H2S is here shown to protect an immortalized mouse hippocampal cell line from oxidative glutamate toxicity by activating ATP-dependent K+ (KATP) and Cl- channels, in addition to increasing the levels of glutathione. The present study therefore identifies a novel pathway for H2S protection from oxidative stress.


Assuntos
Sulfeto de Hidrogênio/farmacologia , Neurônios/patologia , Estresse Oxidativo , Trifosfato de Adenosina/química , Animais , Linhagem Celular , Meios de Cultura/farmacologia , Cisteína/química , Glutationa/química , Glutationa/metabolismo , Sulfeto de Hidrogênio/química , L-Lactato Desidrogenase/metabolismo , Camundongos , Neurônios/metabolismo , Potássio/química , Fatores de Tempo
20.
Biochem Biophys Res Commun ; 342(1): 86-91, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16480682

RESUMO

The responses of cells exposed to nanoparticles have been studied with regard to toxicity, but very little attention has been paid to the possibility that some types of particles can protect cells from various forms of lethal stress. It is shown here that nanoparticles composed of cerium oxide or yttrium oxide protect nerve cells from oxidative stress and that the neuroprotection is independent of particle size. The ceria and yttria nanoparticles act as direct antioxidants to limit the amount of reactive oxygen species required to kill the cells. It follows that this group of nanoparticles could be used to modulate oxidative stress in biological systems.


Assuntos
Cério/farmacologia , Nanoestruturas , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ítrio/farmacologia , Óxido de Alumínio/química , Óxido de Alumínio/farmacologia , Óxido de Alumínio/toxicidade , Antioxidantes/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cério/toxicidade , Humanos , Nanomedicina , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ítrio/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...