Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Antioxid Redox Signal ; 35(12): 1016-1080, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33726509

RESUMO

Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.


Assuntos
Proteínas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Humanos , Oxirredução , Transdução de Sinais
2.
Redox Biol ; 37: 101676, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32956978

RESUMO

UCH-L1 is a deubiquitinating enzyme (DUB), highly abundant in neurons, with a sub-cellular localization dependent on its farnesylation state. Despite UCH-L1's association with familial Parkinson's Disease (PD), the effects on mitochondrial bioenergetics and quality control remain unexplored. Here we investigated the role of UCHL-1 in mitochondrial dynamics and bioenergetics. We demonstrate that knock-down (KD) of UCH-L1 in different cell lines reduces the levels of the mitochondrial fusion protein Mitofusin-2, but not Mitofusin-1, resulting in mitochondrial enlargement and disruption of the tubular network. This was associated with lower tethering between mitochondria and the endoplasmic reticulum, consequently altering mitochondrial calcium uptake. Respiratory function was also altered, as UCH-L1 KD cells displayed higher proton leak and maximum respiratory capacity. Conversely, overexpression of UCH-L1 increased Mfn2 levels, an effect dramatically enhanced by the mutation of the farnesylation site (C220S), which drives UCH-L1 binding to membranes. These data indicate that the soluble cytosolic form of UCH-L1 regulates Mitofusin-2 levels and mitochondrial function. These effects are biologically conserved, since knock-down of the corresponding UCH-L1 ortholog in D. melanogaster reduces levels of the mitofusin ortholog Marf and also increases mitochondrial respiratory capacity. We thus show that Mfn-2 levels are directly affected by UCH-L1, demonstrating that the mitochondrial roles of DUBs go beyond controlling mitophagy rates.


Assuntos
Cálcio , Drosophila melanogaster , Mitocôndrias , Ubiquitina Tiolesterase , Animais , Transporte Biológico , Cálcio/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Retículo Endoplasmático/metabolismo , GTP Fosfo-Hidrolases , Mitocôndrias/genética , Mitocôndrias/metabolismo , Ubiquitina Tiolesterase/metabolismo
3.
Biochem J ; 477(2): 461-475, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-32003437

RESUMO

Mitochondrial turnover is required for proper cellular function. Both mitochondrial biogenesis and mitophagy are impaired in several degenerative and age-related diseases. The search for mitophagy activators recently emerged as a new therapeutical approach; however, there is a lack in suitable tools to follow mitochondrial turnover in a high-throughput manner. We demonstrate that the fluorescent protein, MitoTimer, is a reliable and robust probe to follow mitochondrial turnover. The screening of 15 000 small molecules led us to two chemically-related benzothiophenes that stimulate basal mitophagy in the beta-cell line, INS1. Enhancing basal mitophagy was associated with improved mitochondrial function, higher Complex I activity and Complex II and III expressions in INS1 cells, as well as better insulin secretion performance in mouse islets. The possibility of further enhancing mitophagy in the absence of mitochondrial stressors points to the existence of a 'basal mitophagy spare capacity'. To this end, we found two small molecules that can be used as models to better understand the physiological regulation of mitophagy.


Assuntos
Envelhecimento/genética , Secreção de Insulina/genética , Mitocôndrias/genética , Mitofagia/genética , Envelhecimento/patologia , Animais , Autofagia/genética , Linhagem Celular , Citometria de Fluxo , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Renovação Mitocondrial , Mitofagia/efeitos dos fármacos , Complexos Multienzimáticos/química , Complexos Multienzimáticos/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Tiofenos/química , Tiofenos/farmacologia
4.
FEBS J ; 283(5): 822-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26732506

RESUMO

ß-cells quickly adjust insulin secretion to oscillations in nutrients carried by the blood, acting as fuel sensors. However, most studies of ß-cell responses to nutrients do not discriminate between fuel levels and signaling components present in the circulation. Here we studied the effect of serum from calorie-restricted rats versus serum from rats fed ad libitum, diluted tenfold in the medium, which did not contribute significantly to the pool of nutrients, on ß-cell mitochondrial function and dynamics under regular and high-nutrient culture conditions. Insulin secreting beta-cell derived line (INS1) cells incubated with serum from calorie-restricted rats (CR serum) showed higher levels of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α) and active nitric oxide synthase. The expression of mitofusin-2 (Mfn-2) and optic atrophy 1 (OPA-1), proteins involved in mitochondrial fusion, was increased, while the levels of the mitochondrial fission mediator dynamin related protein 1 (DRP-1) were reduced. Consistent with changes in mitochondrial dynamics protein levels, CR serum treatment increased mitochondrial fusion rates, as well as their length and connectivity. These changes in mitochondrial morphology were associated with prolonged glucose-stimulated insulin secretion and mitochondrial respiration. When combining CR serum and high levels of glucose and palmitate (20 and 0.4 mm, respectively), an in vitro model of type II diabetes, we observed that signaling promoted by CR serum was enough to overcome glucolipotoxicity, as indicated by CR-mediated prevention of mitochondrial fusion arrest and reduced respiratory function in INS1 cells under glucolipotoxicity. Overall, our results provide evidence that non-nutrient factors in serum have a major impact on ß-cell mitochondrial adaptations to changes in metabolism.


Assuntos
Restrição Calórica , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Animais , Linhagem Celular , Respiração Celular , Separação Celular , Dinaminas/metabolismo , Citometria de Fluxo , GTP Fosfo-Hidrolases/metabolismo , Insulina/metabolismo , Masculino , Dinâmica Mitocondrial , Óxido Nítrico Sintase Tipo III/metabolismo , Consumo de Oxigênio , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
5.
Ageing Res Rev ; 12(1): 22-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22504406

RESUMO

Mitochondrial energy metabolism and mitochondrially-derived oxidants have, for many years, been recognized as central toward the effects of aging. A body of recent work has focused on the relationship between mitochondrial redox state, aging and dietary interventions that affect lifespan. These studies have uncovered mechanisms through which diet alters mitochondrial metabolism, in addition to determining how these changes affect oxidant generation, which in itself has an impact on mitochondrial function in aged animals. Many of the studies conducted to date, however, are correlative, and it remains to be determined which of the energy metabolism and redox modifications induced by diet are central toward lifespan extent. Furthermore, dietary interventions used for laboratory animals are often unequal, and of difficult comparison with humans (for whom, by nature, no long-term sound scientific information on the effects of diet on mitochondrial redox state and aging is available). We hope future studies will be able to mechanistically characterize which energy metabolism and redox changes promoted by dietary interventions have positive lifespan effects, and translate these findings into human prevention and treatment of age-related disease.


Assuntos
Envelhecimento/metabolismo , Dieta , Mitocôndrias/metabolismo , Animais , Restrição Calórica , Metabolismo Energético/fisiologia , Humanos , Longevidade , Oxidantes , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Nitrogênio/fisiologia , Espécies Reativas de Oxigênio/metabolismo
6.
J Vis Exp ; (65): e3991, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22847388

RESUMO

Mitochondrial fusion plays an essential role in mitochondrial calcium homeostasis, bioenergetics, autophagy and quality control. Fusion is quantified in living cells by photo-conversion of matrix targeted photoactivatable GFP (mtPAGFP) in a subset of mitochondria. The rate at which the photoconverted molecules equilibrate across the entire mitochondrial population is used as a measure of fusion activity. Thus far measurements were performed using a single cell time lapse approach, quantifying the equilibration in one cell over an hour. Here, we scale up and automate a previously published live cell method based on using mtPAGFP and a low concentration of TMRE (15 nm). This method involves photoactivating a small portion of the mitochondrial network, collecting highly resolved stacks of confocal sections every 15 min for 1 hour, and quantifying the change in signal intensity. Depending on several factors such as ease of finding PAGFP expressing cells, and the signal of the photoactivated regions, it is possible to collect around 10 cells within the 15 min intervals. This provides a significant improvement in the time efficiency of this assay while maintaining the highly resolved subcellular quantification as well as the kinetic parameters necessary to capture the detail of mitochondrial behavior in its native cytoarchitectural environment. Mitochondrial dynamics play a role in many cellular processes including respiration, calcium regulation, and apoptosis. The structure of the mitochondrial network affects the function of mitochondria, and the way they interact with the rest of the cell. Undergoing constant division and fusion, mitochondrial networks attain various shapes ranging from highly fused networks, to being more fragmented. Interestingly, Alzheimer's disease, Parkinson's disease, Charcot Marie Tooth 2A, and dominant optic atrophy have been correlated with altered mitochondrial morphology, namely fragmented networks. Often times, upon fragmentation, mitochondria become depolarized, and upon accumulation this leads to impaired cell function. Mitochondrial fission has been shown to signal a cell to progress toward apoptosis. It can also provide a mechanism by which to separate depolarized and inactive mitochondria to keep the bulk of the network robust. Fusion of mitochondria, on the other hand, leads to sharing of matrix proteins, solutes, mtDNA and the electrochemical gradient, and also seems to prevent progression to apoptosis. How fission and fusion of mitochondria affects cell homeostasis and ultimately the functioning of the organism needs further understanding, and therefore the continuous development and optimization of how to gather information on these phenomena is necessary. Existing mitochondrial fusion assays have revealed various insights into mitochondrial physiology, each having its own advantages. The hybrid PEG fusion assay, mixes two populations of differently labeled cells (mtRFP and mtYFP), and analyzes the amount of mixing and colocalization of fluorophores in fused, multinucleated, cells. Although this method has yielded valuable information, not all cell types can fuse, and the conditions under which fusion is stimulated involves the use of toxic drugs that likely affect the normal fusion process. More recently, a cell free technique has been devised, using isolated mitochondria to observe fusion events based on a luciferase assay. Two human cell lines are targeted with either the amino or a carboxy terminal part of Renilla luciferase along with a leucine zipper to ensure dimerization upon mixing. Mitochondria are isolated from each cell line, and fused. The fusion reaction can occur without the cytosol under physiological conditions in the presence of energy, appropriate temperature and inner mitochondrial membrane potential. Interestingly, the cytosol was found to modulate the extent of fusion, demonstrating that cell signaling regulates the fusion process. This assay will be very useful for high throughput screening to identify components of the fusion machinery and also pharmacological compounds that may affect mitochondrial dynamics. However, more detailed whole cell mitochondrial assays will be needed to complement this in vitro assay to observe these events within a cellular environment. A technique for monitoring whole-cell mitochondrial dynamics has been in use for some time and is based on a mitochondrially-targeted photoactivatable GFP (mtPAGFP). Upon expression of the mtPAGFP, a small portion of the mitochondrial network is photoactivated (10-20%), and the spread of the signal to the rest of the mitochondrial network is recorded every 15 minutes for 1 hour using time lapse confocal imaging. Each fusion event leads to a dilution of signal intensity, enabling quantification of the fusion rate. Although fusion and fission are continuously occurring in cells, this technique only monitors fusion as fission does not lead to a dilution of the PAGFP signal. Co-labeling with low levels of TMRE (7-15 nM in INS1 cells) allows quantification of the membrane potential of mitochondria. When mitochondria are hyperpolarized they uptake more TMRE, and when they depolarize they lose the TMRE dye. Mitochondria that depolarize no longer have a sufficient membrane potential and tend not to fuse as efficiently if at all. Therefore, active fusing mitochondria can be tracked with these low levels of TMRE. Accumulation of depolarized mitochondria that lack a TMRE signal may be a sign of phototoxicity or cell death. Higher concentrations of TMRE render mitochondria very sensitive to laser light, and therefore great care must be taken to avoid overlabeling with TMRE. If the effect of depolarization of mitochondria is the topic of interest, a technique using slightly higher levels of TMRE and more intense laser light can be used to depolarize mitochondria in a controlled fashion (Mitra and Lippincott-Schwartz, 2010). To ensure that toxicity due to TMRE is not an issue, we suggest exposing loaded cells (3-15 nM TMRE) to the imaging parameters that will be used in the assay (perhaps 7 stacks of 6 optical sections in a row), and assessing cell health after 2 hours. If the mitochondria appear too fragmented and cells are dying, other mitochondrial markers, such as dsRED or Mitotracker red could be used instead of TMRE. The mtPAGFP method has revealed details about mitochondrial network behavior that could not be visualized using other methods. For example, we now know that mitochondrial fusion can be full or transient, where matrix content can mix without changing the overall network morphology. Additionally, we know that the probability of fusion is independent of contact duration and organelle dimension, is influenced by organelle motility, membrane potential and history of previous fusion activity. In this manuscript, we describe a methodology for scaling up the previously published protocol using mtPAGFP and 15 nM TMRE in order to examine multiple cells at a time and improve the time efficiency of data collection without sacrificing the subcellular resolution. This has been made possible by the use of an automated microscope stage, and programmable image acquisition software. Zen software from Zeiss allows the user to mark and track several designated cells expressing mtPAGFP. Each of these cells can be photoactivated in a particular region of interest, and stacks of confocal slices can be monitored for mtPAGFP signal as well as TMRE at specified intervals. Other confocal systems could be used to perform this protocol provided there is an automated stage that is programmable, an incubator with CO2, and a means by which to photoactivate the PAGFP; either a multiphoton laser, or a 405 nm diode laser.


Assuntos
Proteínas de Fluorescência Verde/química , Microscopia Confocal/métodos , Mitocôndrias/química , Fusão Celular , Linhagem Celular , Humanos , Mitocôndrias/fisiologia , Compostos Organometálicos/química
7.
PLoS One ; 7(2): e31155, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22319612

RESUMO

eNOS activation resulting in mitochondrial biogenesis is believed to play a central role in life span extension promoted by calorie restriction (CR). We investigated the mechanism of this activation by treating vascular cells with serum from CR rats and found increased Akt and eNOS phosphorylation, in addition to enhanced nitrite release. Inhibiting Akt phosphorylation or immunoprecipitating adiponectin (found in high quantities in CR serum) completely prevented the increment in nitrite release and eNOS activation. Overall, we demonstrate that adiponectin in the serum from CR animals increases NO• signaling by activating the insulin pathway. These results suggest this hormone may be a determinant regulator of the beneficial effects of CR.


Assuntos
Adiponectina/metabolismo , Restrição Calórica , Insulina/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Adiponectina/sangue , Animais , Células Cultivadas , Endotélio Vascular/citologia , Proteína Oncogênica v-akt/metabolismo , Fosforilação , Ratos , Transdução de Sinais
8.
Free Radic Biol Med ; 52(7): 1236-41, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22310960

RESUMO

Calorie restriction (CR) enhances animal life span and prevents age-related diseases, including neurological decline. Recent evidence suggests that a mechanism involved in CR-induced life-span extension is NO(•)-stimulated mitochondrial biogenesis. We examine here the effects of CR on brain mitochondrial content. CR increased eNOS and nNOS and the content of mitochondrial proteins (cytochrome c oxidase, citrate synthase, and mitofusin) in the brain. Furthermore, we established an in vitro system to study the neurological effects of CR using serum extracted from animals on this diet. In cultured neurons, CR serum enhanced nNOS expression and increased levels of nitrite (a NO(•) product). CR serum also enhanced the levels of cytochrome c oxidase and increased citrate synthase activity and respiratory rates in neurons. CR serum effects were inhibited by L-NAME and mimicked by the NO(•) donor SNAP. Furthermore, both CR sera and SNAP were capable of improving neuronal survival. Overall, our results indicate that CR increases mitochondrial biogenesis in a NO(•)-mediated manner, resulting in enhanced reserve respiratory capacity and improved survival in neurons.


Assuntos
Encéfalo/metabolismo , Restrição Calórica , Radicais Livres/metabolismo , Mitocôndrias/metabolismo , Neurônios/citologia , Óxido Nítrico/metabolismo , Sistema Respiratório/metabolismo , Animais , Western Blotting , Encéfalo/citologia , Células Cultivadas , Citrato (si)-Sintase , Feminino , Camundongos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Oxirredução
9.
Free Radic Biol Med ; 51(7): 1454-60, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21816219

RESUMO

Calorie restriction is a dietary intervention known to improve redox state, glucose tolerance, and animal life span. Other interventions have been adopted as study models for caloric restriction, including nonsupplemented food restriction and intermittent, every-other-day feedings. We compared the short- and long-term effects of these interventions to ad libitum protocols and found that, although all restricted diets decrease body weight, intermittent feeding did not decrease intra-abdominal adiposity. Short-term calorie restriction and intermittent feeding presented similar results relative to glucose tolerance. Surprisingly, long-term intermittent feeding promoted glucose intolerance, without a loss in insulin receptor phosphorylation. Intermittent feeding substantially increased insulin receptor nitration in both intra-abdominal adipose tissue and muscle, a modification associated with receptor inactivation. All restricted diets enhanced nitric oxide synthase levels in the insulin-responsive adipose tissue and skeletal muscle. However, whereas calorie restriction improved tissue redox state, food restriction and intermittent feedings did not. In fact, long-term intermittent feeding resulted in largely enhanced tissue release of oxidants. Overall, our results show that restricted diets are significantly different in their effects on glucose tolerance and redox state when adopted long-term. Furthermore, we show that intermittent feeding can lead to oxidative insulin receptor inactivation and glucose intolerance.


Assuntos
Restrição Calórica/métodos , Dieta Redutora/métodos , Gordura Intra-Abdominal/metabolismo , Músculo Esquelético/metabolismo , Obesidade/dietoterapia , Receptor de Insulina/metabolismo , Adiposidade , Animais , Western Blotting , Peso Corporal , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/análise , Proteínas Substratos do Receptor de Insulina/biossíntese , Masculino , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/biossíntese , Nitrocompostos , Obesidade/metabolismo , Oxirredução , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/antagonistas & inibidores
10.
PLoS One ; 6(3): e18433, 2011 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-21483800

RESUMO

Enhanced mitochondrial biogenesis promoted by eNOS activation is believed to play a central role in the beneficial effects of calorie restriction (CR). Since treatment of mice with dinitrophenol (DNP) promotes health and lifespan benefits similar to those observed in CR, we hypothesized that it could also impact biogenesis. We found that DNP and CR increase citrate synthase activity, PGC-1α, cytochrome c oxidase and mitofusin-2 expression, as well as fasting plasma levels of NO• products. In addition, eNOS and Akt phosphorylation in skeletal muscle and visceral adipose tissue was activated in fasting CR and DNP animals. Overall, our results indicate that systemic mild uncoupling activates eNOS and Akt-dependent pathways leading to mitochondrial biogenesis.


Assuntos
Restrição Calórica , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Western Blotting , Citrato (si)-Sintase/metabolismo , Jejum/sangue , Feminino , Camundongos , Nitritos/sangue
11.
Curr Drug Targets ; 12(6): 783-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21275885

RESUMO

Mild mitochondrial uncoupling, or the reduction of the efficiency of energy conversion without compromising intracellular high energy phosphate levels, is a protective therapeutic strategy under many laboratory conditions. Here we discuss these conditions, which include both cell and animal models of ischemia reperfusion and complications associated with the metabolic syndrome. We also discuss drugs that promote mild mitochondrial uncoupling and naturally occurring mild mitochondrial uncoupling pathways involving free fatty acid cycling and K(+) transport.


Assuntos
Síndrome Metabólica/complicações , Mitocôndrias/efeitos dos fármacos , Traumatismo por Reperfusão/fisiopatologia , Animais , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/metabolismo , Humanos , Canais Iônicos/metabolismo , Transporte de Íons , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Potássio/metabolismo , Proteína Desacopladora 1
12.
PLoS One ; 5(5): e10666, 2010 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-20498724

RESUMO

BACKGROUND: The aim of the present study was to evaluate the protective effects of the 4-anilinoquinazoline derivative PD153035 on cardiac ischemia/reperfusion and mitochondrial function. METHODOLOGY/PRINCIPAL FINDINGS: Perfused rat hearts and cardiac HL-1 cells were used to determine cardioprotective effects of PD153035. Isolated rat heart mitochondria were studied to uncover mechanisms of cardioprotection. Nanomolar doses of PD153035 strongly protect against heart and cardiomyocyte damage induced by ischemia/reperfusion and cyanide/aglycemia. PD153035 did not alter oxidative phosphorylation, nor directly prevent Ca(2+) induced mitochondrial membrane permeability transition. The protective effect of PD153035 on HL-1 cells was also independent of AKT phosphorylation state. Interestingly, PD153035 activated K(+) transport in isolated mitochondria, in a manner prevented by ATP and 5-hydroxydecanoate, inhibitors of mitochondrial ATP-sensitive K(+) channels (mitoK(ATP)). 5-Hydroxydecanoate also inhibited the cardioprotective effect of PD153035 in cardiac HL-1 cells, demonstrating that this protection is dependent on mitoK(ATP) activation. CONCLUSIONS/SIGNIFICANCE: We conclude that PD153035 is a potent cardioprotective compound and acts in a mechanism involving mitoK(ATP) activation.


Assuntos
Cardiotônicos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio/metabolismo , Quinazolinas/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Cálcio/metabolismo , Respiração Celular/efeitos dos fármacos , Cianetos/toxicidade , Testes de Função Cardíaca , Técnicas In Vitro , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/enzimologia , Miocárdio/patologia , Permeabilidade/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Potássio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar
13.
Ageing Res Rev ; 9(4): 424-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20493280

RESUMO

Undernutrition without malnutrition is an intervention that enhances laboratory animal life span, and is widely studied to uncover factors limiting longevity. In a search of the literature over a course of four years, we found that most protocols currently adopted as caloric restriction do not meet micronutrient standards set by the National Research Council for laboratory rats and mice. We provide evidence that the most commonly adopted caloric restriction protocol, a 40% restriction of the AIN-93 diet without vitamin or mineral supplementation, leads to malnutrition in both mice and rats. Furthermore, others and we find that every other day feeding, another dietary intervention often referred to as caloric restriction, does not limit the total amount of calories consumed. Altogether, we propose that the term "caloric restriction" should be used specifically to describe diets that decrease calorie intake but not micronutrient availability, and that protocols adopted should be described in detail in order to allow for comparisons and better understanding of the effects of these diets.


Assuntos
Restrição Calórica , Desnutrição , Ração Animal/efeitos adversos , Animais , Restrição Calórica/efeitos adversos , Suplementos Nutricionais/normas , Longevidade/fisiologia , Desnutrição/etiologia , Camundongos , Micronutrientes/administração & dosagem , Micronutrientes/deficiência , Modelos Animais , Ratos , Ratos Sprague-Dawley , Projetos de Pesquisa
14.
Biochim Biophys Acta ; 1802(5): 462-71, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20097285

RESUMO

Mutations in the gene encoding cytosolic Cu,Zn-superoxide dismutase (SOD1) have been linked to familial amyotrophic lateral sclerosis (FALS). However the molecular mechanisms of motor neuron death are multi-factorial and remain unclear. Here we examined DNA damage, p53 activity and apoptosis in SH-SY5Y human neuroblastoma cells transfected to achieve low-level expression of either wild-type or mutant Gly(93)-->Ala (G93A) SOD1, typical of FALS. DNA damage was investigated by evaluating the levels of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGuo) and DNA strand breaks. Significantly higher levels of DNA damage, increased p53 activity, and a greater percentage of apoptotic cells were observed in SH-SY5Y cells transfected with G93A SOD1 when compared to cells overexpressing wild-type SOD1 and untransfected cells. Western blot, FACS, and confocal microscopy analysis demonstrated that G93A SOD1 is present in the nucleus in association with DNA. Nuclear G93A SOD1 has identical superoxide dismutase activity but displays increased peroxidase activity when compared to wild-type SOD1. These results indicate that the G93A mutant SOD1 association with DNA might induce DNA damage and trigger the apoptotic response by activating p53. This toxic activity of mutant SOD1 in the nucleus may play an important role in the complex mechanisms associated with motor neuron death observed in ALS pathogenesis.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Apoptose , Cromatina/metabolismo , Dano ao DNA , Neuroblastoma/metabolismo , Superóxido Dismutase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Esclerose Lateral Amiotrófica/patologia , Núcleo Celular/enzimologia , Núcleo Celular/patologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Humanos , Técnicas Imunoenzimáticas , Peroxidação de Lipídeos , Superóxido Dismutase-1 , Células Tumorais Cultivadas
15.
Aging Cell ; 7(4): 552-60, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18505478

RESUMO

Caloric restriction is the most effective non-genetic intervention to enhance lifespan known to date. A major research interest has been the development of therapeutic strategies capable of promoting the beneficial results of this dietary regimen. In this sense, we propose that compounds that decrease the efficiency of energy conversion, such as mitochondrial uncouplers, can be caloric restriction mimetics. Treatment of mice with low doses of the protonophore 2,4-dinitrophenol promotes enhanced tissue respiratory rates, improved serological glucose, triglyceride and insulin levels, decrease of reactive oxygen species levels and tissue DNA and protein oxidation, as well as reduced body weight. Importantly, 2,4-dinitrophenol-treated animals also presented enhanced longevity. Our results demonstrate that mild mitochondrial uncoupling is a highly effective in vivo antioxidant strategy, and describe the first therapeutic intervention capable of effectively reproducing the physiological, metabolic and lifespan effects of caloric restriction in healthy mammals.


Assuntos
2,4-Dinitrofenol/farmacologia , Metabolismo Energético/efeitos dos fármacos , Longevidade/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Desacopladores/farmacologia , Animais , Glicemia/metabolismo , Respiração Celular/efeitos dos fármacos , Feminino , Insulina/sangue , Camundongos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA