Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
FASEB J ; 32(7): 3844-3858, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29485903

RESUMO

Evolutionary considerations suggest that the body has been optimized to perform at a high level in the food-deprived state when fatty acids and their ketone metabolites are a major fuel source for muscle cells. Because controlled food deprivation in laboratory animals and intermittent energy restriction in humans is a potent physiologic stimulus for ketosis, we designed a study to determine the impact of intermittent food deprivation during endurance training on performance and to elucidate the underlying cellular and molecular mechanisms. Male mice were randomly assigned to either ad libitum feeding or alternate-day food deprivation (ADF) groups, and half of the mice in each diet group were trained daily on a treadmill for 1 mo. A run to exhaustion endurance test performed at the end of the training period revealed superior performance in the mice maintained on ADF during training compared to mice fed ad libitum during training. Maximal O2 consumption was increased similarly by treadmill training in mice on ADF or ad libitum diets, whereas respiratory exchange ratio was reduced in ADF mice on food-deprivation days and during running. Analyses of gene expression in liver and soleus tissues, and metabolomics analysis of blood suggest that the metabolic switch invoked by ADF and potentiated by exercise strongly modulates molecular pathways involved in mitochondrial biogenesis, metabolism, and cellular plasticity. Our findings demonstrate that ADF engages metabolic and cellular signaling pathways that result in increased metabolic efficiency and endurance capacity.-Marosi, K., Moehl, K., Navas-Enamorado, I., Mitchell, S. J., Zhang, Y., Lehrmann, E., Aon, M. A., Cortassa, S., Becker, K. G., Mattson, M. P. Metabolic and molecular framework for the enhancement of endurance by intermittent food deprivation.


Assuntos
Privação de Alimentos , Condicionamento Físico Animal/métodos , Resistência Física , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Biogênese de Organelas
2.
Proc Natl Acad Sci U S A ; 113(44): 12502-12507, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27791127

RESUMO

Cockayne syndrome is a neurodegenerative accelerated aging disorder caused by mutations in the CSA or CSB genes. Although the pathogenesis of Cockayne syndrome has remained elusive, recent work implicates mitochondrial dysfunction in the disease progression. Here, we present evidence that loss of CSA or CSB in a neuroblastoma cell line converges on mitochondrial dysfunction caused by defects in ribosomal DNA transcription and activation of the DNA damage sensor poly-ADP ribose polymerase 1 (PARP1). Indeed, inhibition of ribosomal DNA transcription leads to mitochondrial dysfunction in a number of cell lines. Furthermore, machine-learning algorithms predict that diseases with defects in ribosomal DNA (rDNA) transcription have mitochondrial dysfunction, and, accordingly, this is found when factors involved in rDNA transcription are knocked down. Mechanistically, loss of CSA or CSB leads to polymerase stalling at non-B DNA in a neuroblastoma cell line, in particular at G-quadruplex structures, and recombinant CSB can melt G-quadruplex structures. Indeed, stabilization of G-quadruplex structures activates PARP1 and leads to accelerated aging in Caenorhabditis elegans In conclusion, this work supports a role for impaired ribosomal DNA transcription in Cockayne syndrome and suggests that transcription-coupled resolution of secondary structures may be a mechanism to repress spurious activation of a DNA damage response.


Assuntos
DNA Helicases/genética , Enzimas Reparadoras do DNA/genética , DNA de Neoplasias/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , Fatores de Transcrição/genética , Transcrição Gênica , Linhagem Celular Tumoral , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Dano ao DNA , DNA Helicases/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , DNA Ribossômico/genética , Quadruplex G , Técnicas de Silenciamento de Genes , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Fatores de Transcrição/metabolismo
3.
J Neurochem ; 142(2): 286-296, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28382744

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by aggregation of toxic forms of amyloid ß peptide (Aß). Treatment strategies have largely been focused on inhibiting the enzymes (ß- and γ-secretases) that liberate Aß from the amyloid precursor protein (APP). While evidence suggests that individuals who exercise regularly are at reduced risk for AD and studies of animal models demonstrate that running can ameliorate brain Aß pathology and associated cognitive deficits, the underlying mechanisms are unknown. However, considerable evidence suggests that brain-derived neurotrophic factor (BDNF) mediates beneficial effects of exercise on neuroplasticity and cellular stress resistance. Here, we tested the hypothesis that BDNF promotes non-amyloidogenic APP processing. Using a transgenic mouse model of Alzheimer's disease and cultured human neural cells, we demonstrate that exercise and BDNF reduce production of toxic Aß peptides through a mechanism involving enhanced α-secretase processing of APP. This anti-amyloidogenic APP processing involves subcellular redistribution of α-secretase and an increase in intracellular neuroprotective APP peptides capable of binding and inhibiting ß-secretase. Moreover, our results suggest that BDNF's ability to promote neurite outgrowth is primarily exerted through pathways other than APP processing. Exercise and other factors that enhance BDNF signaling may therefore have both therapeutic and prophylactic value in the battle against AD. Read the Editorial Highlight for this article on page 191.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/biossíntese , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Condicionamento Físico Animal , Transdução de Sinais/fisiologia
4.
Alzheimers Dement ; 13(5): 582-591, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27599210

RESUMO

Recent findings have elucidated roles for mitochondrial uncoupling proteins (UCPs) in neuronal plasticity and resistance to metabolic and oxidative stress. UCPs are induced by bioenergetic challenges such as caloric restriction and exercise and may protect neurons against dysfunction and degeneration. The pharmacological uncoupler 2,4-dinitrophenol (DNP), which was once prescribed to >100,000 people as a treatment for obesity, stimulates several adaptive cellular stress-response signaling pathways in neurons including those involving the brain-derived neurotrophic factor (BDNF), the transcription factor cyclic AMP response element-binding protein (CREB), and autophagy. Preclinical data show that low doses of DNP can protect neurons and improve functional outcome in animal models of Alzheimer's and Parkinson's diseases, epilepsy, and cerebral ischemic stroke. Repurposing of DNP and the development of novel uncoupling agents with hormetic mechanisms of action provide opportunities for new breakthrough therapeutic interventions in a range of acute and chronic insidious neurodegenerative/neuromuscular conditions, all paradoxically at body weight-preserving doses.


Assuntos
2,4-Dinitrofenol/administração & dosagem , Neuroproteção/fisiologia , Proteína Desacopladora 1/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo , Modelos Animais de Doenças , Humanos , Doenças Neurodegenerativas/prevenção & controle , Plasticidade Neuronal
5.
J Neurochem ; 139(5): 769-781, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27739595

RESUMO

During fasting and vigorous exercise, a shift of brain cell energy substrate utilization from glucose to the ketone 3-hydroxybutyrate (3OHB) occurs. Studies have shown that 3OHB can protect neurons against excitotoxicity and oxidative stress, but the underlying mechanisms remain unclear. Neurons maintained in the presence of 3OHB exhibited increased oxygen consumption and ATP production, and an elevated NAD+ /NADH ratio. We found that 3OHB metabolism increases mitochondrial respiration which drives changes in expression of brain-derived neurotrophic factor (BDNF) in cultured cerebral cortical neurons. The mechanism by which 3OHB induces Bdnf gene expression involves generation of reactive oxygen species, activation of the transcription factor NF-κB, and activity of the histone acetyltransferase p300/EP300. Because BDNF plays important roles in synaptic plasticity and neuronal stress resistance, our findings suggest cellular signaling mechanisms by which 3OHB may mediate adaptive responses of neurons to fasting, exercise, and ketogenic diets.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Córtex Cerebral/metabolismo , Metabolismo Energético/fisiologia , Neurônios/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
6.
Biogerontology ; 13(4): 413-27, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22722983

RESUMO

Ageing of the brain is accompanied by variable degrees of cognitive decline. Estrogens have profound effects on brain ageing by exerting neurotrophic and neuroprotective types of action. Furthermore, exercise has also been claimed to play a role in the non-pharmacological prevention of psycho-neuronal decline with ageing. In the present study the question was asked whether chronic physical exercise might substitute the action of estrogens in aged rats. We compared the effects of 17ß-estradiol (E2) treatment and long-term moderate physical exercise in ageing (15 months, early stage of ageing) and old (27 months) female rats, on cognitive functions and the relevant intracellular molecular signaling pathways in the hippocampus. Results showed that both treatments improved attention and memory functions of the 15 months old rats. Like E2, physical training enhanced the level of brain derived nerve growth factor and the activation of PKA/Akt/CREB and MAPK/CREB pathways. The treatments also enhanced the levels of synaptic molecules synaptophysin and synapsin I, which could explain the improved cognitive functions. In the 27 months old rats the behavioral and molecular effects of E2 were indistinguishable from those found in the 15 months old animals but the effects of physical exercise in most of the measures proved to be practically ineffective. It is concluded that the effectiveness of regular and moderate intensity physical exercise is age-dependent while the action of E2 treatment is comparable between the ageing and old female rats on maintaining cognition and its underlying molecular mechanisms.


Assuntos
Envelhecimento/fisiologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Estradiol/farmacologia , Fármacos Neuroprotetores , Condicionamento Físico Animal/fisiologia , Animais , Estradiol/administração & dosagem , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Injeções Subcutâneas , Memória/efeitos dos fármacos , Memória/fisiologia , Modelos Animais , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resultado do Tratamento
7.
Orv Hetil ; 153(32): 1247-55, 2012 Aug 12.
Artigo em Húngaro | MEDLINE | ID: mdl-22878034

RESUMO

There is compelling evidence for a genetic contribution to physical performance. In addition, there is an advanced scientific knowledge on the predisposition to sports-related diseases and injuries. Genetic testing of performance related polymorphisms can serve as a new opportunity for developing the process of talent selection. Sport-related genetic information may also allow for individualization of the training and improve performance. Genetic testing may also play an important role in the pre-participation screening for injuries and disease risks.


Assuntos
Atletas , Traumatismos em Atletas/genética , Traumatismos em Atletas/prevenção & controle , Pesquisa em Genética , Testes Genéticos , Polimorfismo Genético , Esportes , Actinina/genética , Atletas/educação , Comportamento de Escolha/ética , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/prevenção & controle , Humanos , Comunicação Interdisciplinar , Força Muscular/genética , Peptidil Dipeptidase A/genética , Educação Física e Treinamento/ética , Educação Física e Treinamento/normas , Educação Física e Treinamento/tendências , Desempenho Psicomotor , Traumatismos dos Tendões/genética
8.
Orv Hetil ; 153(12): 445-53, 2012 Mar 25.
Artigo em Húngaro | MEDLINE | ID: mdl-22411217

RESUMO

Cardiovascular diseases (CVDs) are the leading causes of death in the developed countries. Elevated homocysteine level is as an independent risk factor of CVDs. The C677T and A1298C variants of methylenetetrahydrofolate reductase gene (MTHFR) have been shown to influence folate and homocysteine metabolisms. However, the relationship between MTHFR polymorphisms and hyperhomocysteinemia has not been well established yet. The gene variants were also reported to be associated with CVDs. In addition, the C677T polymorphisms may play a role in the development of hypertension. Recent research evidence has suggested that MTHFR variants might be independently linked to CVDs and hypertension, because of the involvement of the MTHFR enzyme product (5-methyl-tetrahydrofolate /5-MTHF) in the regulation of endothelial functions. Further research is required to investigate the association between gene polymorphisms of folate-metabolizing enzymes and CVDs, and to identify the possible role of the relevant gene variants in the molecular pathogenesis of hyperhomocysteinemia.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/genética , Ferredoxina-NADP Redutase/genética , Homocisteína/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Ácido Fólico/metabolismo , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Hipertensão/enzimologia , Hipertensão/genética
9.
Orv Hetil ; 153(52): 2070-6, 2012 Dec 30.
Artigo em Húngaro | MEDLINE | ID: mdl-23261995

RESUMO

Elevated plasma lipid level is one of the main risk factors for cardiovascular diseases, which are considered to be primary causes of death. Apolipoprotein E plays a part in the lipid transport in the blood, thus polimophisms of that affect the lipid composition of the plasma. The three most common alleles of apolipoprotein E are e2, e3, e4. Out of the two non-wild type alleles, the e2 and e4, the latter was shown to play a role in the development of cardiovascular diseases and Alzheimer's disease. Some studies mention the e2/e2 homozygote genotype as one of the causes of hyperlipoproteinemia type III. Besides lipid metabolism, apolipoprotein E also influences the manifestation of cardiovascular diseases through other biochemical pathways, therefore it is essential to explore the molecular background of these metabolic pathways.


Assuntos
Apolipoproteínas E/metabolismo , Doenças Cardiovasculares/metabolismo , Polimorfismo Genético , Doença de Alzheimer/metabolismo , Apolipoproteína E2/metabolismo , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteínas E/sangue , Apolipoproteínas E/genética , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/genética , LDL-Colesterol/sangue , Frequência do Gene , Genótipo , Humanos , Hiperlipoproteinemia Tipo III/complicações , Hiperlipoproteinemia Tipo III/genética , Fatores de Risco
10.
Orv Hetil ; 153(8): 296-302, 2012 Feb 26.
Artigo em Húngaro | MEDLINE | ID: mdl-22330841

RESUMO

UNLABELLED: Marfan syndrome is a genetic disorder of the connective tissue, which affects approximately 2000-3000 individuals in Hungary. Given its multi-systemic manifestations, this disorder is often difficult to diagnose. To date, the National Marfan Register system contains approximately 250 cases, and this number is dynamically increasing. AIMS: Collection of data from biological samples, clinical parameters, and lifestyle factors in Hungarian patients with Marfan syndrome. METHODS: In terms of the criteria used for selection, those cases were chosen where the disorder could be clearly diagnosed on the basis of the patients' cardiovascular and systemic symptoms, as well as of their family history, in line with the guidelines set by the Revised Ghent Nosology. RESULTS: For the purposes of developing the biobank used for the research, 102 cases were selected from the Marfan Register (cDNA from 55 patients, genomic DNA and serum from 102 patients). In addition to the samples, data have been obtained by using internationally validated surveys to further examine the role of physical activity, nutrition and various psychological factors. CONCLUSIONS: The establishment of the Marfan Biobank enables scientists to effectively carry out research based on genetic, gene-expression and protein analysis. The biobank also provides new opportunities to study Hungarian patients with Marfan syndrome.


Assuntos
Síndrome de Marfan , Sistema de Registros , Bancos de Tecidos , Adolescente , Adulto , Idoso , Criança , DNA , Feminino , Humanos , Hungria , Estimativa de Kaplan-Meier , Estilo de Vida , Masculino , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/genética , Síndrome de Marfan/mortalidade , Síndrome de Marfan/fisiopatologia , Síndrome de Marfan/psicologia , Pessoa de Meia-Idade , Atividade Motora , Mucosa Bucal , Estado Nutricional , Seleção de Pacientes , RNA , Inquéritos e Questionários
11.
Alzheimers Dement (N Y) ; 7(1): e12168, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35005201

RESUMO

INTRODUCTION: Analyses of off-label use of acetylcholinesterase inhibitors (AChEIs) in mild cognitive impairment (MCI) has produced mixed results. Post hoc analyses of observational cohorts, such as the Alzheimer's Disease Neuroimaging Initiative (ADNI), have reported deleterious effects in AChEI-treated subjects (AChEI+). Here, we used neuroimaging biomarkers to determine whether AChEI+ subjects had a greater rate of neurodegeneration than untreated (AChEI-) subjects while accounting for baseline differences. METHODS: We selected 121 ADNI MCI AChEI+ subjects and 151 AChEI- subjects with a magnetic resonance imaging (MRI) scan; 82 AChEI+ and 110 AChEI- also had a fluorodeoxyglucose (FDG) scan. A subset (83 AChEI+ and 98 AChEI-) had cerebrospinal fluid (CSF) or amyloid positron emission tomography (PET) assessment for amyloid positivity. Linear regression models were used to compare the effect of treatment on changes in Mini-Mental State Examination and Clinical Dementia Rating-Sum of Boxes scores. We used standard regression in SPM (for baseline) and the SPM toolbox sandwich estimator, SwE (for longitudinal) for comparisons of AChEI+ and AChEI- FDG PET and MRI data. RESULTS: At baseline, the AChEI+ group had significantly reduced cortical gray matter density (GMD) and more hypometabolism than AChEI- subjects. The greater rate of atrophy and hypometabolic changes over time in AChEI+ compared to AChEI- subjects did not survive correction for baseline differences. AChEI+ participants were more likely to be amyloid-positive and have lower GMD and FDG standardized uptake value ratio than AChEI- at baseline. AChEI+ subjects showed greater atrophy over time, which remained significant after controlling for amyloid status. DISCUSSION: Our data suggest that the observed differences in rates of cognitive decline, atrophy, and hypometabolism are likely the result of significant baseline differences between the groups. Furthermore, the data indicate no treatment effect of AChEI (positive of negative), rather that physicians prescribe AChEI to subjects who present with more severe clinical impairment. This alone may account for the negative effect seen previously in the ADNI population of AChEI use among MCI subjects.

12.
Drug Discov Today ; 25(2): 438-445, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31562982

RESUMO

Large-scale biobanks can yield unprecedented insights into our health and provide discoveries of new and potentially targetable biomarkers. Several protective loss-of-function alleles have been identified, including variants that protect against cardiovascular disease, obesity, type 2 diabetes, and asthma and allergic diseases. These alleles serve as indicators of efficacy, mimicking the effects of drugs and suggesting that inhibiting these genes could provide therapeutic benefit, as has been observed for PCSK9. We provide a context for these findings through a multifaceted review covering the use of genetics in drug discovery efforts through genome-wide and phenome-wide association studies, linking deep mutation scanning data to molecular function and highlighting some additional tools that might help in the interpretation of newly discovered variants.


Assuntos
Bancos de Espécimes Biológicos , Descoberta de Drogas , Fenômenos Genéticos , Animais , Desenvolvimento de Medicamentos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação
13.
Neurobiol Aging ; 85: 123-130, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31718928

RESUMO

Dopaminergic neuronal cell loss in the substantia nigra is responsible for the motor symptoms that are the clinical hallmark of Parkinson's disease (PD). As of yet there are no treatments that slow or prevent the degeneration of dopaminergic neurons in PD patients. Here we tested the hypothesis that dopaminergic neurons can be protected by treatment with the mitochondrial uncoupling agent 2,4-dinitrophenol (DNP) and the novel DNP prodrug MP201. We found that mice treated with low doses of DNP and MP201 were protected against motor dysfunction and dopamine neuron loss in the 6-hydroxydopamine PD model, with MP201 being more efficacious than DNP. Amelioration of motor deficits and dopamine neuron loss by MP201 treatment was associated with reductions in microglial and astrocyte activation and neuroinflammation. These preclinical findings suggest the potential application of mitochondrial uncoupling agents such as MP201 as disease-modifying therapies for PD.


Assuntos
2,4-Dinitrofenol/análogos & derivados , 2,4-Dinitrofenol/uso terapêutico , Neurônios Dopaminérgicos/patologia , Doença de Parkinson/tratamento farmacológico , Pró-Fármacos/uso terapêutico , 2,4-Dinitrofenol/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Pró-Fármacos/farmacologia
14.
Obesity (Silver Spring) ; 26(2): 254-268, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29086496

RESUMO

OBJECTIVE: Intermittent fasting (IF) is a term used to describe a variety of eating patterns in which no or few calories are consumed for time periods that can range from 12 hours to several days, on a recurring basis. This review is focused on the physiological responses of major organ systems, including the musculoskeletal system, to the onset of the metabolic switch: the point of negative energy balance at which liver glycogen stores are depleted and fatty acids are mobilized (typically beyond 12 hours after cessation of food intake). RESULTS AND CONCLUSIONS: Emerging findings suggest that the metabolic switch from glucose to fatty acid-derived ketones represents an evolutionarily conserved trigger point that shifts metabolism from lipid/cholesterol synthesis and fat storage to mobilization of fat through fatty acid oxidation and fatty acid-derived ketones, which serve to preserve muscle mass and function. Thus, IF regimens that induce the metabolic switch have the potential to improve body composition in overweight individuals. Moreover, IF regimens also induce the coordinated activation of signaling pathways that optimize physiological function, enhance performance, and slow aging and disease processes. Future randomized controlled IF trials should use biomarkers of the metabolic switch (e.g., plasma ketone levels) as a measure of compliance and of the magnitude of negative energy balance during the fasting period.


Assuntos
Ingestão de Alimentos/fisiologia , Ingestão de Energia/fisiologia , Jejum/fisiologia , Comportamento Alimentar/fisiologia , Jejum/metabolismo , Humanos
16.
Sci Rep ; 7: 46208, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28397803

RESUMO

Aging is a major international concern that brings formidable socioeconomic and healthcare challenges. Small molecules capable of improving the health of older individuals are being explored. Small molecules that enhance cellular stress resistance are a promising avenue to alleviate declines seen in human aging. Tomatidine, a natural compound abundant in unripe tomatoes, inhibits age-related skeletal muscle atrophy in mice. Here we show that tomatidine extends lifespan and healthspan in C. elegans, an animal model of aging which shares many major longevity pathways with mammals. Tomatidine improves many C. elegans behaviors related to healthspan and muscle health, including increased pharyngeal pumping, swimming movement, and reduced percentage of severely damaged muscle cells. Microarray, imaging, and behavioral analyses reveal that tomatidine maintains mitochondrial homeostasis by modulating mitochondrial biogenesis and PINK-1/DCT-1-dependent mitophagy. Mechanistically, tomatidine induces mitochondrial hormesis by mildly inducing ROS production, which in turn activates the SKN-1/Nrf2 pathway and possibly other cellular antioxidant response pathways, followed by increased mitophagy. This mechanism occurs in C. elegans, primary rat neurons, and human cells. Our data suggest that tomatidine may delay some physiological aspects of aging, and points to new approaches for pharmacological interventions for diseases of aging.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas de Ligação a DNA/metabolismo , Longevidade/fisiologia , Mitofagia/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tomatina/análogos & derivados , Fatores de Transcrição/metabolismo , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Longevidade/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Músculos/efeitos dos fármacos , Músculos/fisiologia , Biogênese de Organelas , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Tomatina/farmacologia , Transcriptoma/genética
17.
Cell Metab ; 23(1): 128-42, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26698917

RESUMO

The impact of mitochondrial protein acetylation status on neuronal function and vulnerability to neurological disorders is unknown. Here we show that the mitochondrial protein deacetylase SIRT3 mediates adaptive responses of neurons to bioenergetic, oxidative, and excitatory stress. Cortical neurons lacking SIRT3 exhibit heightened sensitivity to glutamate-induced calcium overload and excitotoxicity and oxidative and mitochondrial stress; AAV-mediated Sirt3 gene delivery restores neuronal stress resistance. In models relevant to Huntington's disease and epilepsy, Sirt3(-/-) mice exhibit increased vulnerability of striatal and hippocampal neurons, respectively. SIRT3 deficiency results in hyperacetylation of several mitochondrial proteins, including superoxide dismutase 2 and cyclophilin D. Running wheel exercise increases the expression of Sirt3 in hippocampal neurons, which is mediated by excitatory glutamatergic neurotransmission and is essential for mitochondrial protein acetylation homeostasis and the neuroprotective effects of running. Our findings suggest that SIRT3 plays pivotal roles in adaptive responses of neurons to physiological challenges and resistance to degeneration.


Assuntos
Mitocôndrias/enzimologia , Neurônios/fisiologia , Sirtuína 3/fisiologia , Acetilação , Adaptação Fisiológica , Animais , Cálcio/metabolismo , Células Cultivadas , Metabolismo Energético , Hipocampo/citologia , Potencial da Membrana Mitocondrial , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Neostriado/citologia , Degeneração Neural/enzimologia , Condicionamento Físico Animal , Fatores de Proteção , Processamento de Proteína Pós-Traducional , Corrida/fisiologia , Estresse Fisiológico
18.
Artigo em Inglês | MEDLINE | ID: mdl-27928512

RESUMO

Alzheimer's Disease (AD) is an age-related neurodegenerative disorder in which aggregation-prone neurotoxic amyloid ß-peptide (Aß) accumulates in the brain. Extracellular vesicles (EVs) are small 50-150 nanometer membrane vesicles that have recently been implicated in the prion-like spread of self-aggregating proteins. Here we report that EVs isolated from AD patient CSF and plasma, from the plasma of two AD mouse models, and from the medium of neural cells expressing familial AD presenilin 1 mutations, destabilize neuronal Ca2+ homeostasis, impair mitochondrial function, and sensitize neurons to excitotoxicity. EVs contain a relatively low amount of Aß but have an increased Aß42/ Aß40 ratio; the majority of Aß is located on the surface of the EVs. Impairment of lysosome function results in increased generation EVs with elevated Aß42 levels. EVs may mediate transcellular spread of pathogenic Aß species and that impair neuronal Ca2+ handling and mitochondrial function, and may thereby render neurons vulnerable to excitotoxicity.

19.
Cell Metab ; 24(4): 566-581, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27732836

RESUMO

Ataxia telangiectasia (A-T) is a rare autosomal recessive disease characterized by progressive neurodegeneration and cerebellar ataxia. A-T is causally linked to defects in ATM, a master regulator of the response to and repair of DNA double-strand breaks. The molecular basis of cerebellar atrophy and neurodegeneration in A-T patients is unclear. Here we report and examine the significance of increased PARylation, low NAD+, and mitochondrial dysfunction in ATM-deficient neurons, mice, and worms. Treatments that replenish intracellular NAD+ reduce the severity of A-T neuropathology, normalize neuromuscular function, delay memory loss, and extend lifespan in both animal models. Mechanistically, treatments that increase intracellular NAD+ also stimulate neuronal DNA repair and improve mitochondrial quality via mitophagy. This work links two major theories on aging, DNA damage accumulation, and mitochondrial dysfunction through nuclear DNA damage-induced nuclear-mitochondrial signaling, and demonstrates that they are important pathophysiological determinants in premature aging of A-T, pointing to therapeutic interventions.


Assuntos
Ataxia Telangiectasia/patologia , Reparo do DNA/efeitos dos fármacos , Saúde , Longevidade/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , NAD/farmacologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Comportamento Animal , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/ultraestrutura , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Homeostase/efeitos dos fármacos , Metabolômica , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Ftalazinas/farmacologia , Piperazinas/farmacologia , Proteômica , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/metabolismo
20.
Cell Metab ; 21(4): 509-10, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25863241

RESUMO

In a recent publication in Neuron, Choe et al. (2015) demonstrate that brain-derived neurotrophic factor (BDNF) signaling mediates salt-induced blood pressure elevation by increasing the excitability of hypothalamic vasopressin-secreting neurons. These findings suggest complex roles for BDNF in adaptive cardiovascular responses to physiological challenges and in the pathogenesis of hypertension.


Assuntos
Barorreflexo/fisiologia , Pressão Sanguínea/fisiologia , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cloreto de Sódio na Dieta/efeitos adversos , Simportadores/metabolismo , Vasopressinas/metabolismo , Animais , Masculino
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa