Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Drugs Ther ; 36(1): 1-13, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-32648168

RESUMO

PURPOSE: Mitochondrial reactive oxygen species (ROS) production upon reperfusion of ischemic tissue initiates the ischemia/reperfusion (I/R) injury associated with heart attack. During ischemia, succinate accumulates and its oxidation upon reperfusion by succinate dehydrogenase (SDH) drives ROS production. Inhibition of succinate accumulation and/or oxidation by dimethyl malonate (DMM), a cell permeable prodrug of the SDH inhibitor malonate, can decrease I/R injury. However, DMM is hydrolysed slowly, requiring administration to the heart prior to ischemia, precluding its administration to patients at the point of reperfusion, for example at the same time as unblocking a coronary artery following a heart attack. To accelerate malonate delivery, here we developed more rapidly hydrolysable malonate esters. METHODS: We synthesised a series of malonate esters and assessed their uptake and hydrolysis by isolated mitochondria, C2C12 cells and in mice in vivo. In addition, we assessed protection against cardiac I/R injury by the esters using an in vivo mouse model of acute myocardial infarction. RESULTS: We found that the diacetoxymethyl malonate diester (MAM) most rapidly delivered large amounts of malonate to cells in vivo. Furthermore, MAM could inhibit mitochondrial ROS production from succinate oxidation and was protective against I/R injury in vivo when added at reperfusion. CONCLUSIONS: The rapidly hydrolysed malonate prodrug MAM can protect against cardiac I/R injury in a clinically relevant mouse model.


Assuntos
Cardiotônicos/farmacologia , Malonatos/farmacologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Cardiotônicos/síntese química , Cardiotônicos/química , Linhagem Celular , Modelos Animais de Doenças , Ésteres/química , Feminino , Humanos , Masculino , Malonatos/síntese química , Malonatos/química , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Pró-Fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Ácido Succínico/metabolismo
3.
J Biol Chem ; 293(25): 9869-9879, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29743240

RESUMO

The generation of mitochondrial superoxide (O2̇̄) by reverse electron transport (RET) at complex I causes oxidative damage in pathologies such as ischemia reperfusion injury, but also provides the precursor to H2O2 production in physiological mitochondrial redox signaling. Here, we quantified the factors that determine mitochondrial O2̇̄ production by RET in isolated heart mitochondria. Measuring mitochondrial H2O2 production at a range of proton-motive force (Δp) values and for several coenzyme Q (CoQ) and NADH pool redox states obtained with the uncoupler p-trifluoromethoxyphenylhydrazone, we show that O2̇̄ production by RET responds to changes in O2 concentration, the magnitude of Δp, and the redox states of the CoQ and NADH pools. Moreover, we determined how expressing the alternative oxidase from the tunicate Ciona intestinalis to oxidize the CoQ pool affected RET-mediated O2̇̄ production at complex I, underscoring the importance of the CoQ pool for mitochondrial O2̇̄ production by RET. An analysis of O2̇̄ production at complex I as a function of the thermodynamic forces driving RET at complex I revealed that many molecules that affect mitochondrial reactive oxygen species production do so by altering the overall thermodynamic driving forces of RET, rather than by directly acting on complex I. These findings clarify the factors controlling RET-mediated mitochondrial O2̇̄ production in both pathological and physiological conditions. We conclude that O2̇̄ production by RET is highly responsive to small changes in Δp and the CoQ redox state, indicating that complex I RET represents a major mode of mitochondrial redox signaling.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Superóxidos/metabolismo , Ubiquinona/metabolismo , Animais , Transporte de Elétrons , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação Oxidativa , Ratos , Ratos Wistar , Transdução de Sinais
4.
Free Radic Biol Med ; 108: 374-382, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28366801

RESUMO

The redox state of cysteine thiols is critical for protein function. Whereas cysteines play an important role in the maintenance of protein structure through the formation of internal disulfides, their nucleophilic thiol groups can become oxidatively modified in response to diverse redox challenges and thereby function in signalling and antioxidant defences. These oxidative modifications occur in response to a range of agents and stimuli, and can lead to the existence of multiple redox states for a given protein. To assess the role(s) of a protein in redox signalling and antioxidant defence, it is thus vital to be able to assess which of the multiple thiol redox states are present and to investigate how these alter under different conditions. While this can be done by a range of mass spectrometric-based methods, these are time-consuming, costly, and best suited to study abundant proteins or to perform an unbiased proteomic screen. One approach that can facilitate a targeted assessment of candidate proteins, as well as proteins that are low in abundance or proteomically challenging, is by electrophoretic mobility shift assays. Redox-modified cysteine residues are selectively tagged with a large group, such as a polyethylene glycol (PEG) polymer, and then the proteins are separated by electrophoresis followed by immunoblotting, which allows the inference of redox changes based on band shifts. However, the applicability of this method has been impaired by the difficulty of cleanly modifying protein thiols by large PEG reagents. To establish a more robust method for redox-selective PEGylation, we have utilised a Click chemistry approach, where free thiol groups are first labelled with a reagent modified to contain an alkyne moiety, which is subsequently Click-reacted with a PEG molecule containing a complementary azide function. This strategy can be adapted to study reversibly reduced or oxidised cysteines. Separation of the thiol labelling step from the PEG conjugation greatly facilitates the fidelity and flexibility of this approach. Here we show how the Click-PEGylation technique can be used to interrogate the redox state of proteins.


Assuntos
Cisteína/química , Polietilenoglicóis/metabolismo , Compostos de Sulfidrila/química , Animais , Catalase/química , Catalase/metabolismo , Bovinos , Dissulfetos/química , Eletroforese , Ensaio de Desvio de Mobilidade Eletroforética , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/química , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Oxirredução , Estresse Oxidativo , Polietilenoglicóis/química , Proteômica/métodos , Coelhos
5.
Acta Histochem ; 119(3): 315-326, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28314612

RESUMO

Mitochondria exist in a dynamic cycle of fusion and fission whose balance directly influences the morphology of the 'mitochondrial network', a term that encompasses the branched, reticular structure of fused mitochondria as well as the separate, punctate individual organelles within a eukaryotic cell. Over the past decade, the significance of the mitochondrial network has been increasingly appreciated, motivating the development of various approaches to analyze it. Here, we describe the Mitochondrial Network Analysis (MiNA) toolset, a relatively simple pair of macros making use of existing ImageJ plug-ins, allowing for semi-automated analysis of mitochondrial networks in cultured mammalian cells. MiNA is freely available at https://github.com/ScienceToolkit/MiNA. The tool incorporates optional preprocessing steps to enhance the quality of images before converting the images to binary and producing a morphological skeleton for calculating nine parameters to quantitatively capture the morphology of the mitochondrial network. The efficacy of the macro toolset is demonstrated using a sample set of images from SH-SY5Y, C2C12, and mouse embryo fibroblast (MEF) cell cultures treated under different conditions and exhibiting hyperfused, fused, and fragmented mitochondrial network morphologies.


Assuntos
Técnicas Citológicas/métodos , Fibroblastos/citologia , Mitocôndrias/ultraestrutura , Software , Animais , Técnicas de Cultura de Células , Linhagem Celular , Humanos , Camundongos , Coloração e Rotulagem
6.
Biochem Biophys Res Commun ; 485(2): 249-254, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28235489

RESUMO

Resveratrol (RES) is a plant-derived stilbene associated with a wide range of health benefits. Mitochondria are a key downstream target of RES, and in some cell types RES promotes mitochondrial biogenesis, altered cellular redox status, and a shift toward oxidative metabolism. Mitochondria exist as a dynamic network that continually remodels via fusion and fission processes, and the extent of fusion is related to cellular redox status and metabolism. We investigated RES's effects on mitochondrial network morphology in several cell lines using a quantitative approach to measure the extent of network fusion. 48 h continuous treatment with 10-20 µM RES stimulated mitochondrial fusion in C2C12 myoblasts, PC3 cancer cells, and mouse embryonic fibroblasts stimulated significant increases in fusion in all instances, resulting in larger and more highly branched mitochondrial networks. Mitofusin-2 (Mfn2) is a key protein facilitating mitochondrial fusion, and its expression was also stimulated by RES. Using Mfn2-null cells we demonstrated that RES's effects on mitochondrial fusion, cellular respiration rates, and cell growth are all dependent upon the presence of Mfn2. Taken together, these results demonstrate that Mfn2 and mitochondrial fusion are affected by RES in ways that appear to relate to RES's known effects on cellular metabolism and growth.


Assuntos
Antioxidantes/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Estilbenos/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , GTP Fosfo-Hidrolases/genética , Deleção de Genes , Humanos , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/genética , Resveratrol
7.
Cell Metab ; 23(2): 379-85, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26712463

RESUMO

The mitochondrial membrane potential (Δψm) is a major determinant and indicator of cell fate, but it is not possible to assess small changes in Δψm within cells or in vivo. To overcome this, we developed an approach that utilizes two mitochondria-targeted probes each containing a triphenylphosphonium (TPP) lipophilic cation that drives their accumulation in response to Δψm and the plasma membrane potential (Δψp). One probe contains an azido moiety and the other a cyclooctyne, which react together in a concentration-dependent manner by "click" chemistry to form MitoClick. As the mitochondrial accumulation of both probes depends exponentially on Δψm and Δψp, the rate of MitoClick formation is exquisitely sensitive to small changes in these potentials. MitoClick accumulation can then be quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). This approach enables assessment of subtle changes in membrane potentials within cells and in the mouse heart in vivo.


Assuntos
Química Click/métodos , Potencial da Membrana Mitocondrial , Espectrometria de Massas em Tandem/métodos , Animais , Linhagem Celular , Camundongos Endogâmicos C57BL , Sondas Moleculares/metabolismo
8.
Free Radic Biol Med ; 89: 668-78, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26453920

RESUMO

Mitochondrial oxidative damage contributes to a wide range of pathologies. One therapeutic strategy to treat these disorders is targeting antioxidants to mitochondria by conjugation to the lipophilic triphenylphosphonium (TPP) cation. To date only hydrophobic antioxidants have been targeted to mitochondria; however, extending this approach to hydrophilic antioxidants offers new therapeutic and research opportunities. Here we report the development and characterization of MitoC, a mitochondria-targeted version of the hydrophilic antioxidant ascorbate. We show that MitoC can be taken up by mitochondria, despite the polarity and acidity of ascorbate, by using a sufficiently hydrophobic link to the TPP moiety. MitoC reacts with a range of reactive species, and within mitochondria is rapidly recycled back to the active ascorbate moiety by the glutathione and thioredoxin systems. Because of this accumulation and recycling MitoC is an effective antioxidant against mitochondrial lipid peroxidation and also decreases aconitase inactivation by superoxide. These findings show that the incorporation of TPP function can be used to target polar and acidic compounds to mitochondria, opening up the delivery of a wide range of bioactive compounds. Furthermore, MitoC has therapeutic potential as a new mitochondria-targeted antioxidant, and is a useful tool to explore the role(s) of ascorbate within mitochondria.


Assuntos
Antioxidantes/química , Antioxidantes/farmacologia , Ácido Ascórbico/química , Ácido Ascórbico/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Mitocôndrias Hepáticas/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Feminino , Mitocôndrias Hepáticas/efeitos dos fármacos , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar
9.
Free Radic Biol Med ; 89: 883-94, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26454075

RESUMO

Superoxide is the proximal reactive oxygen species (ROS) produced by the mitochondrial respiratory chain and plays a major role in pathological oxidative stress and redox signaling. While there are tools to detect or decrease mitochondrial superoxide, none can rapidly and specifically increase superoxide production within the mitochondrial matrix. This lack impedes progress, making it challenging to assess accurately the roles of mitochondrial superoxide in cells and in vivo. To address this unmet need, we synthesized and characterized a mitochondria-targeted redox cycler, MitoParaquat (MitoPQ) that comprises a triphenylphosphonium lipophilic cation conjugated to the redox cycler paraquat. MitoPQ accumulates selectively in the mitochondrial matrix driven by the membrane potential. Within the matrix, MitoPQ produces superoxide by redox cycling at the flavin site of complex I, selectively increasing superoxide production within mitochondria. MitoPQ increased mitochondrial superoxide in isolated mitochondria and cells in culture ~a thousand-fold more effectively than untargeted paraquat. MitoPQ was also more toxic than paraquat in the isolated perfused heart and in Drosophila in vivo. MitoPQ enables the selective generation of superoxide within mitochondria and is a useful tool to investigate the many roles of mitochondrial superoxide in pathology and redox signaling in cells and in vivo.


Assuntos
Herbicidas/farmacologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Hepáticas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Paraquat/farmacologia , Superóxidos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Complexo I de Transporte de Elétrons , Feminino , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Oxirredução , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Nature ; 515(7527): 431-435, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25383517

RESUMO

Ischaemia-reperfusion injury occurs when the blood supply to an organ is disrupted and then restored, and underlies many disorders, notably heart attack and stroke. While reperfusion of ischaemic tissue is essential for survival, it also initiates oxidative damage, cell death and aberrant immune responses through the generation of mitochondrial reactive oxygen species (ROS). Although mitochondrial ROS production in ischaemia reperfusion is established, it has generally been considered a nonspecific response to reperfusion. Here we develop a comparative in vivo metabolomic analysis, and unexpectedly identify widely conserved metabolic pathways responsible for mitochondrial ROS production during ischaemia reperfusion. We show that selective accumulation of the citric acid cycle intermediate succinate is a universal metabolic signature of ischaemia in a range of tissues and is responsible for mitochondrial ROS production during reperfusion. Ischaemic succinate accumulation arises from reversal of succinate dehydrogenase, which in turn is driven by fumarate overflow from purine nucleotide breakdown and partial reversal of the malate/aspartate shuttle. After reperfusion, the accumulated succinate is rapidly re-oxidized by succinate dehydrogenase, driving extensive ROS generation by reverse electron transport at mitochondrial complex I. Decreasing ischaemic succinate accumulation by pharmacological inhibition is sufficient to ameliorate in vivo ischaemia-reperfusion injury in murine models of heart attack and stroke. Thus, we have identified a conserved metabolic response of tissues to ischaemia and reperfusion that unifies many hitherto unconnected aspects of ischaemia-reperfusion injury. Furthermore, these findings reveal a new pathway for metabolic control of ROS production in vivo, while demonstrating that inhibition of ischaemic succinate accumulation and its oxidation after subsequent reperfusion is a potential therapeutic target to decrease ischaemia-reperfusion injury in a range of pathologies.


Assuntos
Isquemia/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Ácido Succínico/metabolismo , Monofosfato de Adenosina/metabolismo , Animais , Ácido Aspártico/metabolismo , Ciclo do Ácido Cítrico , Modelos Animais de Doenças , Transporte de Elétrons , Complexo I de Transporte de Elétrons/metabolismo , Fumaratos/metabolismo , Isquemia/enzimologia , Malatos/metabolismo , Masculino , Metabolômica , Camundongos , Mitocôndrias/enzimologia , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/metabolismo , Miocárdio/citologia , Miocárdio/enzimologia , Miocárdio/metabolismo , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , NAD/metabolismo , Traumatismo por Reperfusão/enzimologia , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/metabolismo , Succinato Desidrogenase/metabolismo
11.
Aging Cell ; 13(5): 962-4, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25059507

RESUMO

Reduced signalling through the insulin/insulin-like growth factor-1 signalling (IIS) pathway is a highly conserved lifespan determinant in model organisms. The precise mechanism underlying the effects of the IIS on lifespan and health is currently unclear, although cellular stress resistance may be important. We have previously demonstrated that mice globally lacking insulin receptor substrate 1 (Irs1(-/-) ) are long-lived and enjoy a greater period of their life free from age-related pathology compared with wild-type (WT) controls. In this study, we show that primary dermal fibroblasts and primary myoblasts derived from Irs1(-/-) mice are no more resistant to a range of oxidant and nonoxidant chemical stressors than cells derived from WT mice.


Assuntos
Fibroblastos/metabolismo , Proteínas Substratos do Receptor de Insulina/deficiência , Estresse Fisiológico/fisiologia , Animais , Feminino , Fibroblastos/efeitos dos fármacos , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo/fisiologia
12.
Longev Healthspan ; 3(1): 4, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24690218

RESUMO

Since its inception more than four decades ago, the Mitochondrial Free Radical Theory of Aging (MFRTA) has served as a touchstone for research into the biology of aging. The MFRTA suggests that oxidative damage to cellular macromolecules caused by reactive oxygen species (ROS) originating from mitochondria accumulates in cells over an animal's lifespan and eventually leads to the dysfunction and failure that characterizes aging. A central prediction of the theory is that the ability to ameliorate or slow this process should be associated with a slowed rate of aging and thus increased lifespan. A vast pool of data bearing on this idea has now been published. ROS production, ROS neutralization and macromolecule repair have all been extensively studied in the context of longevity. We review experimental evidence from comparisons between naturally long- or short-lived animal species, from calorie restricted animals, and from genetically modified animals and weigh the strength of results supporting the MFRTA. Viewed as a whole, the data accumulated from these studies have too often failed to support the theory. Excellent, well controlled studies from the past decade in particular have isolated ROS as an experimental variable and have shown no relationship between its production or neutralization and aging or longevity. Instead, a role for mitochondrial ROS as intracellular messengers involved in the regulation of some basic cellular processes, such as proliferation, differentiation and death, has emerged. If mitochondrial ROS are involved in the aging process, it seems very likely it will be via highly specific and regulated cellular processes and not through indiscriminate oxidative damage to macromolecules.

13.
Phytochemistry ; 98: 164-73, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24361291

RESUMO

The mitochondrial antioxidant enzyme, Mn superoxide dismutase (MnSOD), has been shown to confer cytoprotection and to regulate cell cycle progression. Resveratrol, a phytoestrogen found in red wines and other foods, has been previously reported to increase MnSOD protein levels and activity both in vitro and in vivo. Numerous structural analogues of resveratrol produced via the same stilbene synthesis pathway (e.g. pterostilbene and piceid) and also present in foods and red wine may be capable of eliciting the same effects. Furthermore, in humans resveratrol is rapidly metabolized to resveratrol-4'-sulfate, resveratrol-3-glucuronide and other metabolites in vivo. Although these metabolites may accumulate to relatively high levels in plasma and tissues, little is known about their biological activities. Here the activities were compared of these stilbenes and stilbene metabolites in mammalian cells. Two key cellular activities associated with resveratrol were examined: inhibition of proliferative growth and increased stress resistance (important anti-cancer and cell protective activities, respectively). While resveratrol-4'-sulfate and resveratrol-3-glucuronide had no effect on either cell growth or stress resistance, both pterostilbene and piceid were at least as effective as resveratrol. Using pharmacological and genetic approaches, it was found that the effects of pterostilbene and piceid required an induction of the mitochondrial enzyme MnSOD and intact mitochondrial respiration. In addition, using estrogen receptor beta (ERbeta) knockout mouse myoblasts, it was demonstrated that the effects of stilbene compounds on cell growth and stress resistance all require ERbeta. Taken together, these results indicate that resveratrol, pterostilbene and piceid all activate the same mitochondrial response in mammalian cells, and therefore these latter two molecules might be as effective as resveratrol in eliciting positive health outcomes in vivo.


Assuntos
Inibidores Enzimáticos/farmacologia , Receptor beta de Estrogênio/antagonistas & inibidores , Glucosídeos/farmacologia , Estilbenos/farmacologia , Superóxido Dismutase/antagonistas & inibidores , Animais , Antioxidantes/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Receptor beta de Estrogênio/metabolismo , Glucosídeos/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Estresse Oxidativo/efeitos dos fármacos , Resveratrol , Estilbenos/química , Relação Estrutura-Atividade , Superóxido Dismutase/metabolismo
14.
Phytother Res ; 28(1): 120-31, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23526725

RESUMO

Phytoestrogens are of interest because of their reported beneficial effects on many human maladies including cancer, neurodegeneration, cardiovascular disease and diabetes. As data on phytoestrogens continues to accumulate, it is clear that there is significant overlap in the cellular effects elicited by these various compounds. Here, we show that one mechanism by which a number of phytoestrogens achieve their growth inhibitory and cytoprotective effects is via induction of the mitochondrial manganese superoxide dismutase (MnSOD). Eight phytoestrogens, including resveratrol, coumestrol, kaempferol, genistein, daidzein, apigenin, isoliquirtigenin and glycitin, were tested for their ability to induce MnSOD expression in mouse C2C12 and primary myoblasts. Five of these, resveratrol, coumestrol, kaempferol, genistein and daidzein, significantly increased MnSOD expression, slowed proliferative growth and enhanced stress resistance (hydrogen peroxide LD50) . When siRNA was used to prevent the MnSOD induction by genistein, coumestrol or daidzein, none of these compounds exerted any effect on proliferative growth, and only the effect of coumestrol on stress resistance persisted. The estrogen antagonist ICI182780 prevented the increased MnSOD expression and also the changes in cell growth and stress resistance, indicating that these effects are mediated by estrogen receptors (ER). The absence of effects of resveratrol or coumestrol, but not genistein, in ERß-null cells further indicated that this ER in particular is important in mediating these effects. Thus, an ER-mediated induction of MnSOD expression appears to underlie the growth inhibitory and cytoprotective activities of multiple phytoestrogens.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fitoestrógenos/farmacologia , Superóxido Dismutase/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Cumestrol/farmacologia , Citoproteção , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Genisteína/farmacologia , Isoflavonas/farmacologia , Quempferóis/farmacologia , Camundongos , Mioblastos/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Resveratrol , Estilbenos/farmacologia , Estresse Fisiológico
15.
Age (Dordr) ; 35(5): 1937-47, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22836712

RESUMO

Discovering key cellular and molecular traits that promote longevity is a major goal of aging and longevity research. One experimental strategy is to determine which traits have been selected during the evolution of longevity in naturally long-lived animal species. This comparative approach has been applied to lifespan research for nearly four decades, yielding hundreds of datasets describing aspects of cell and molecular biology hypothesized to relate to animal longevity. Here, we introduce a Comparative Cellular and Molecular Biology of Longevity Database, available at ( http://genomics.brocku.ca/ccmbl/ ), as a compendium of comparative cell and molecular data presented in the context of longevity. This open access database will facilitate the meta-analysis of amalgamated datasets using standardized maximum lifespan (MLSP) data (from AnAge). The first edition contains over 800 data records describing experimental measurements of cellular stress resistance, reactive oxygen species metabolism, membrane composition, protein homeostasis, and genome homeostasis as they relate to vertebrate species MLSP. The purpose of this review is to introduce the database and briefly demonstrate its use in the meta-analysis of combined datasets.


Assuntos
Biologia Celular , Longevidade , Biologia Molecular/métodos , Bases de Dados Factuais , Homeostase
16.
Artigo em Inglês | MEDLINE | ID: mdl-22708124

RESUMO

Within mammalian species, standard metabolic rate (SMR) increases disproportionately with body mass (Mb), such that the mass-specific SMR correlates negatively with Mb. This phenomenon can be explained in part by reduced cellular metabolic rates in larger species. To better understand the cause(s) of this cellular metabolic rate allometry we have used an ex vivo approach to isolate and identify potential contributors. Skeletal myoblasts from mammalian species ranging inMb from 30 g to over 300,000 g were isolated and differentiated into myotubes in vitro. Oxygen consumption rates, citrate synthase (CS) activity, and lactate dehydrogenase (LDH) activity were measured in myotubes under standardized conditions. No correlation of any of these parameters was observedwith speciesMb, suggesting that there is no genetic contribution to between-species differences in cellular metabolic rates. Myotubes were incubated in serum from species ranging from 30 g to 400,000 g to determine whether between-species differences in the levels of metabolically important hormones might produce allometric trends in the cultured cells. However, there was no observed effect of serum donor Mb on any of the metabolic characteristicsmeasured. Thus, there is no evidence for a relationship between skeletal muscle oxidative metabolism and Mb in an ex vivo model.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Mioblastos Esqueléticos/metabolismo , Animais , Metabolismo Basal , Células Cultivadas , Citrato (si)-Sintase/metabolismo , Metabolismo Energético , L-Lactato Desidrogenase/metabolismo , Mamíferos , Fibras Musculares Esqueléticas/enzimologia , Mioblastos Esqueléticos/enzimologia , Consumo de Oxigênio/fisiologia
17.
Free Radic Biol Med ; 50(7): 821-31, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21215799

RESUMO

trans-Resveratrol (RES) is one of a number of dietary polyphenols that have been reported to beneficially affect human physiology. Although numerous studies have attributed this to direct interactions between RES and histone deacetylases, recently the reliability of these results has been questioned. We have shown that the mitochondrial superoxide dismutase (MnSOD) is substantially upregulated in RES-treated cells. Here we explore the mechanisms underlying this, showing that two of RES's more interesting effects, inhibition of replication and enhancement of stress resistance, are mediated by MnSOD upregulation in three cell lines: MRC5 human lung fibroblasts, C2C12 mouse myoblasts, and SHSY5Y human neuroblastoma cells. When small interfering RNA was used to prevent induction of MnSOD expression, the effects of RES on population doubling time of cells in culture, and resistance to cell death after exposure to hydrogen peroxide or paraquat, were abolished. Interestingly, the RES-induced upregulation of MnSOD levels could be prevented by the estrogen receptor antagonist ICI 182780. RES's effects also could be reproduced using estradiol or the estrogen receptor-ß agonist diarylpropionitrile, but not using the estrogen receptor-α agonist propylpyrazole triol. Thus, we suggest that RES interacts with estrogen receptor-ß to induce the upregulation of MnSOD, which affects cell cycle progression and stress resistance. These results have important implications for our understanding of RES's biological activities and potential applications to human health.


Assuntos
Antioxidantes/farmacologia , Receptor beta de Estrogênio/antagonistas & inibidores , Estilbenos/farmacologia , Superóxido Dismutase/metabolismo , Regulação para Cima , Animais , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Nitrilas/farmacologia , Fenóis , Propionatos/farmacologia , Pirazóis/farmacologia , Resveratrol , Estresse Fisiológico/efeitos dos fármacos , Superóxido Dismutase/genética
18.
Mech Ageing Dev ; 131(4): 242-52, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20219522

RESUMO

Mitochondrial redox metabolism has long been considered to play important roles in mammalian aging and the development of age-related pathologies in the major oxidative organs. Both genetic and dietary manipulations of mitochondrial redox metabolism have been associated with the extension of lifespan. Here we provide a broad overview of the circumstantial evidence showing associations between mitochondrial reactive oxygen species (ROS) metabolism, aging and longevity. We address most aspects of mitochondrial ROS metabolism, from superoxide production, to ROS detoxification and the repair/removal of ROS-mediated macromolecular damage. Finally, we discuss the effects of dietary manipulations (e.g. caloric restriction, methionine restriction), dietary deficiencies (e.g. folate) and dietary supplementation (e.g. resveratrol) on mitochondrial ROS metabolism and lifespan.


Assuntos
Envelhecimento/metabolismo , Metionina/metabolismo , Metionina/farmacologia , Mitocôndrias/metabolismo , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Animais , Restrição Calórica , Dieta , Suplementos Nutricionais , Longevidade/efeitos dos fármacos , Longevidade/genética , Metionina/genética , Mitocôndrias/genética , Oxirredução , Ratos , Espécies Reativas de Oxigênio/metabolismo
19.
Molecules ; 15(3): 1196-212, 2010 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-20335973

RESUMO

Epidemiological evidence indicates that nutritionally-derived polyphenols such as resveratrol (RES) have neuroprotective properties. Administration of RES to culture media protects a wide variety of neuronal cell types from stress-induced death. Dietary supplementation of RES can ameliorate neuronal damage and death resulting from both acute and chronic stresses in rodents. The specific molecular mechanisms by which RES acts at the cellular level remain incompletely understood. However, many experimental data indicate that RES reduces or prevents the occurrence of oxidative damage. Here we discuss possible mechanisms by which RES might exert protection against oxidative damage and cell death. Evidence suggesting that RES's chemical antioxidant potential is not sufficient explanation for its effects is discussed. Putative biological activities, including interactions with estrogen receptors and sirtuins are critically discussed. We provide a synthesis of how RES's phytoestrogenic properties might mediate the neuronal stress resistance underlying its observed neuroprotective properties.


Assuntos
Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Camundongos , Ratos , Resveratrol , Estilbenos/farmacocinética , Superóxido Dismutase/metabolismo
20.
Curr Aging Sci ; 2(1): 12-27, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20021396

RESUMO

The causes of aging and determinants of maximum lifespan in animal species are multifaceted and complex. However, a wealth of experimental data suggests that mitochondria are involved both in the aging process and in regulating lifespan. Here we outline a somatic cell depletion (SCD) model to account for correlations between: (1) mitochondrial reactive oxygen species and lifespan; (2) mitochondrial antioxidant enzymes and lifespan; (3) mitochondrial DNA mutation and lifespan and (4) cellular stress resistance and lifespan. We examine the available data from within the framework of the SCD model, in which mitochondrial dysfunction leading to cell death and gradual loss of essential somatic cells eventually contributes to the decline in physiological performance that limits lifespan. This model is useful in explaining many of the mitochondrial manipulations that alter maximum lifespan in a variety of animal species; however, there are a number of caveats and critical experiments outstanding, and these are outlined in this review.


Assuntos
Apoptose/fisiologia , Longevidade/fisiologia , Mitocôndrias/fisiologia , Estresse Oxidativo/fisiologia , Envelhecimento/fisiologia , Animais , DNA Mitocondrial/genética , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...