Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmacopsychiatry ; 54(1): 37-46, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33254260

RESUMO

BACKGROUND: Silexan, a special essential oil from flowering tops of lavandula angustifolia, is used to treat subsyndromal anxiety disorders. In a recent clinical trial, Silexan also showed antidepressant effects in patients suffering from mixed anxiety-depression (ICD-10 F41.2). Since preclinical data explaining antidepressant properties of Silexan are missing, we decided to investigate if Silexan also shows antidepressant-like effects in vitro as well as in vivo models. METHODS: We used the forced swimming test (FST) in rats as a simple behavioral test indicative of antidepressant activity in vivo. As environmental events and other risk factors contribute to depression through converging molecular and cellular mechanisms that disrupt neuronal function and morphology-resulting in dysfunction of the circuitry that is essential for mood regulation and cognitive function-we investigated the neurotrophic properties of Silexan in neuronal cell lines and primary hippocampal neurons. RESULTS: The antidepressant activity of Silexan (30 mg/kg BW) in the FST was comparable to the tricyclic antidepressant imipramine (20 mg/kg BW) after 9-day treatment. Silexan triggered neurite outgrowth and synaptogenesis in 2 different neuronal cell models and led to a significant increase in synaptogenesis in primary hippocampal neurons. Silexan led to a significant phosphorylation of protein kinase A and subsequent CREB phosphorylation. CONCLUSION: Taken together, Silexan demonstrates antidepressant-like effects in cellular as well as animal models for antidepressant activity. Therefore, our data provides preclinical evidence for the clinical antidepressant effects of Silexan in patients with mixed depression and anxiety.


Assuntos
Antidepressivos/farmacologia , Óleos Voláteis/farmacologia , Óleos de Plantas/farmacologia , Animais , Proteína de Ligação a CREB/metabolismo , Técnicas de Cultura de Células , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Imipramina/farmacologia , Lavandula , Pregabalina/farmacologia , Ratos , Ratos Sprague-Dawley
2.
J Alzheimers Dis ; 64(s1): S455-S467, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29504539

RESUMO

Because of the failure of all amyloid-ß directed treatment strategies for Alzheimer's disease (AD), the concept of mitochondrial dysfunction as a major pathomechanism of the cognitive decline in aging and AD has received substantial support. Accordingly, improving mitochondrial function as an alternative strategy for new drug development became of increasing interest and many different compounds have been identified which improve mitochondrial function in preclinical in vitro and in vivo experiments. However, very few if any have been investigated in clinical trials, representing a major drawback of the mitochondria directed drug development. To overcome these problems, we used a top-down approach by investigating several older antidementia drugs with clinical evidence of therapeutic efficacy. These include EGb761® (standardized ginkgo biloba extract), piracetam, and Dimebon. All improve experimentally many aspects of mitochondrial dysfunction including mitochondrial dynamics and also improve cognition and impaired neuronal plasticity, the functionally most relevant consequences of mitochondrial dysfunction. All partially inhibit opening events of the mitochondrial permeability transition pore (mPTP) which previously has mainly been discussed as a mechanism relevant for the induction of apoptosis. However, as more recent work suggests the mPTP as a master regulator of many mitochondrial functions, our data suggest the mPTP as a possible relevant drug target within the love triangle between mPTP regulation, mitochondrial dynamics, and mitochondrial function including regulation of neuronal plasticity. Drugs interfering with mPTP function will improve not only mitochondrial impairment in aging and AD but also will have beneficial effects on impaired neuronal plasticity, the pathomechanism which correlates best with functional deficits (cognition, behavior) in aging and AD.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos
4.
Neuron ; 93(3): 560-573.e6, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28111078

RESUMO

Precise regulation of cellular metabolism is hypothesized to constitute a vital component of the developmental sequence underlying the life-long generation of hippocampal neurons from quiescent neural stem cells (NSCs). The identity of stage-specific metabolic programs and their impact on adult neurogenesis are largely unknown. We show that the adult hippocampal neurogenic lineage is critically dependent on the mitochondrial electron transport chain and oxidative phosphorylation machinery at the stage of the fast proliferating intermediate progenitor cell. Perturbation of mitochondrial complex function by ablation of the mitochondrial transcription factor A (Tfam) reproduces multiple hallmarks of aging in hippocampal neurogenesis, whereas pharmacological enhancement of mitochondrial function ameliorates age-associated neurogenesis defects. Together with the finding of age-associated alterations in mitochondrial function and morphology in NSCs, these data link mitochondrial complex function to efficient lineage progression of adult NSCs and identify mitochondrial function as a potential target to ameliorate neurogenesis-defects in the aging hippocampus.


Assuntos
Células-Tronco Adultas/metabolismo , Envelhecimento/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Neurogênese , Neurônios/metabolismo , Células-Tronco Adultas/citologia , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Hipocampo/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais , Neurônios/citologia , Fosforilação Oxidativa
5.
Neural Plast ; 2016: 8075903, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27747106

RESUMO

The mitochondrial cascade hypothesis of dementia assumes mitochondrial dysfunction leading to reduced energy supply, impaired neuroplasticity, and finally cell death as one major pathomechanism underlying the continuum from brain aging over mild cognitive impairment to initial and advanced late onset Alzheimer's disease. Accordingly, improving mitochondrial function has become an important strategy to treat the early stages of this continuum. The metabolic enhancer piracetam has been proposed as possible prototype for those compounds by increasing impaired mitochondrial function and related aspects like mechanisms of neuroplasticity. We here report that piracetam at therapeutically relevant concentrations improves neuritogenesis in the human cell line SH-SY5Y over conditions mirroring the whole spectrum of age-associated cognitive decline. These effects go parallel with improvement of impaired mitochondrial dynamics shifting back fission and fusion balance to the energetically more favorable fusion site. Impaired fission and fusion balance can also be induced by a reduction of the mitochondrial permeability transition pore (mPTP) function as atractyloside which indicates the mPTP has similar effects on mitochondrial dynamics. These changes are also reduced by piracetam. These findings suggest the mPTP as an important target for the beneficial effects of piracetam on mitochondrial function.


Assuntos
Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Plasticidade Neuronal/fisiologia , Piracetam/farmacologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Crescimento Neuronal/efeitos dos fármacos , Crescimento Neuronal/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Células PC12 , Ratos
6.
J Alzheimers Dis ; 50(1): 201-15, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26639968

RESUMO

Aberrant neuronal network activity associated with neuronal hyperexcitability seems to be an important cause of cognitive decline in aging and Alzheimer's disease (AD). Out of many antiepileptics, only levetiracetam improved cognitive dysfunction in AD patients and AD animal models by reducing hyperexcitability. As impaired inhibitory interneuronal function, rather than overactive neurons, seems to be the underlying cause, improving impaired neuronal function rather than quieting overactive neurons might be relevant in explaining the lack of activity of the other antiepileptics. Interestingly, improvement of cognitive deficits by levetiracetam caused by small levels of soluble Aß was accompanied by improvement of synaptic function and plasticity. As the negative effects of Aß on synaptic plasticity strongly correlate with mitochondrial dysfunction, wehypothesized that the effect of levetiracetam on synaptic activity might be raised by an improved mitochondrial function. Accordingly, we investigated possible effects of levetiracetam on neuronal deficits associated with mitochondrial dysfunction linked to aging and AD. Levetiracetam improved several aspects of mitochondrial dysfunction including alterations of fission and fusion balance in a cell model for aging and early late-onset AD. We demonstrate for the first time, using immunohistochemistry and proteomics, that the synaptic vesicle protein 2A (SV2a), the molecular target of levetiracetam, is expressed in mitochondria. In addition, levetiracetam shows significant effect on the opening of the mitochondrial permeability transition pore. Importantly, the effects of levetiracetam were significantly abolished when SV2a was knockdown using siRNA. In conclusion, interfering with the SV2a protein at the mitochondrial level and thereby improving mitochondrial function might represent an additional therapeutic effect of levetiracetam to improve symptoms of late-onset AD.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/tratamento farmacológico , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nootrópicos/uso terapêutico , Piracetam/análogos & derivados , Trifosfato de Adenosina/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Animais , Encéfalo/ultraestrutura , Linhagem Celular , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Feminino , Proteína GAP-43/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Levetiracetam , Masculino , Glicoproteínas de Membrana/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Proteínas do Tecido Nervoso/genética , Nitroprussiato/farmacologia , Piracetam/uso terapêutico , Proteômica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos
7.
Prog Mol Biol Transl Sci ; 127: 183-210, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25149218

RESUMO

The etiology of common, nonfamiliar late-onset Alzheimer's disease (LOAD) is only partly understood and seems to be extremely complex including many genetic and environmental factors. The most important environmental risk factor to develop LOAD is aging itself. Aging and LOAD are considered to be strongly linked to mitochondrial dysfunction and enhanced oxidative stress. In this review, we focus on the interaction between mitochondrial dysfunction in aging especially on defects of the respiratory chain of the oxidative phosphorylation system resulting in enhanced oxidative stress and the interplay between aging-associated mitochondrial defects and LOAD-associated mitochondrial failure. The deleterious effects of the two hallmarks of LOAD, amyloid beta, and hyperphosphorylated tau, on mitochondrial function, movement, and morphology are described as well as the toxic effects of the most relevant genetic risk factor of LOAD, the apolipoprotein E4 allele. Finally, the review provides an overview about drugs and nutritional ingredients which improve mitochondrial function or/and act as antioxidants and discusses their potential role in the treatment of LOAD.


Assuntos
Doença de Alzheimer/patologia , Mitocôndrias/patologia , Envelhecimento/patologia , Animais , Antioxidantes/metabolismo , Encéfalo/patologia , Humanos , Fosforilação Oxidativa
8.
J Alzheimers Dis ; 42(2): 395-411, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24898661

RESUMO

Recent data suggest that the combined effect of oxidative stress due to aging and slightly elevated amyloid-ß (Aß) levels initiate Alzheimer's disease (AD) long before the clinical onset. Investigations of this early phase are hampered by the lack of cellular or animal models reflecting this scenario. We used SH-SY5Y cells stably transfected with an additional copy of the human AßPP gene and artificial aging by complex I inhibition. These cells show slightly elevated Aß levels, moderately decreased ATP levels, impaired mitochondrial membrane potential, and decreased mitochondrial respiration. Assessing mitochondrial dynamics with three different methods reveals a distinct shift toward mitochondrial fission and fragmentation in SH-SY5Y AßPPwt cells. We also performed electron cryo-tomography of isolated mitochondria to reveal that there were no major differences between SH-SY5Y control and SH-SY5Y AßPPwt mitochondria with respect to swelling or loss of cristae. Dystrophic neurites are an early pathological feature of AD. Interestingly, SH-SY5Y AßPPwt cells exhibit significantly longer neurites, likely due to substantially elevated levels of sAßPPα. Complex I inhibition also shows substantial effects on mitochondrial dynamics, impairs neuritogenesis, and elevates Aß levels in both cell types. In SH-SY5Y AßPPwt cells, these defects were more pronounced due to a relatively elevated Aß and a reduced sAßPPα production. Our findings suggest that the progression from low Aß levels to the beginning of AD takes place in the presence of oxidative stress during normal aging. This mechanism not only results from additive effects of both mechanisms on mitochondrial function but might also be additionally aggravated by altered amyloidogenic processing.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Modelos Biológicos , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Potencial da Membrana Mitocondrial/fisiologia , Microscopia Eletrônica , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Neuroblastoma/patologia , Transfecção
9.
Biochem Soc Trans ; 41(5): 1331-4, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24059528

RESUMO

The metabolic enhancer piracetam is used in many countries to treat cognitive impairment in aging, brain injuries, as well as dementia such as AD (Alzheimer's disease). As a specific feature of piracetam, beneficial effects are usually associated with mitochondrial dysfunction. In previous studies we were able to show that piracetam enhanced ATP production, mitochondrial membrane potential as well as neurite outgrowth in cell and animal models for aging and AD. To investigate further the effects of piracetam on mitochondrial function, especially mitochondrial fission and fusion events, we decided to assess mitochondrial morphology. Human neuroblastoma cells were treated with the drug under normal conditions and under conditions imitating aging and the occurrence of ROS (reactive oxygen species) as well as in stably transfected cells with the human wild-type APP (amyloid precursor protein) gene. This AD model is characterized by expressing only 2-fold more human Aß (amyloid ß-peptide) compared with control cells and therefore representing very early stages of AD when Aß levels gradually increase over decades. Interestingly, these cells exhibit an impaired mitochondrial function and morphology under baseline conditions. Piracetam is able to restore this impairment and shifts mitochondrial morphology back to elongated forms, whereas there is no effect in control cells. After addition of a complex I inhibitor, mitochondrial morphology is distinctly shifted to punctate forms in both cell lines. Under these conditions piracetam is able to ameliorate morphology in cells suffering from the mild Aß load, as well as mitochondrial dynamics in control cells.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/efeitos dos fármacos , Piracetam/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Metabolismo , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...