Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Biophys J ; 122(8): 1459-1469, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36905121

ABSTRACT

Mitochondrial inner membrane potentials in cardiomyocytes may oscillate in cycles of depolarization/repolarization when the mitochondrial network is exposed to metabolic or oxidative stress. The frequencies of such oscillations are dynamically changing while clusters of weakly coupled mitochondrial oscillators adjust to a common phase and frequency. Across the cardiac myocyte, the averaged signal of the mitochondrial population follows self-similar or fractal dynamics; however, fractal properties of individual mitochondrial oscillators have not yet been examined. We show that the largest synchronously oscillating cluster exhibits a fractal dimension, D, that is indicative of self-similar behavior with D=1.27±0.11, in contrast to the remaining network mitochondria whose fractal dimension is close to that of Brownian noise, D=1.58±0.10. We further demonstrate that fractal behavior is correlated with local coupling mechanisms, whereas it is only weakly linked to measures of functional connections between mitochondria. Our findings suggest that individual mitochondrial fractal dimensions may serve as a simple measure of local mitochondrial coupling.


Subject(s)
Fractals , Mitochondria , Oxidative Stress , Membrane Potential, Mitochondrial , Mitochondrial Membranes
2.
J Mol Cell Cardiol ; 165: 9-18, 2022 04.
Article in English | MEDLINE | ID: mdl-34954465

ABSTRACT

ATP synthase (F1Fo) is a rotary molecular engine that harnesses energy from electrochemical-gradients across the inner mitochondrial membrane for ATP synthesis. Despite the accepted tenet that F1Fo transports exclusively H+, our laboratory has demonstrated that, in addition to H+, F1Fo ATP synthase transports a significant fraction of ΔΨm-driven charge as K+ to synthesize ATP. Herein, we utilize a computational modeling approach as a proof of principle of the feasibility of the core mechanism underlying the enhanced ATP synthesis, and to explore its bioenergetic consequences. A minimal model comprising the 'core' mechanism constituted by ATP synthase, driven by both proton (PMF) and potassium motive force (KMF), respiratory chain, adenine nucleotide translocator, Pi carrier, and K+/H+ exchanger (KHEmito) was able to simulate enhanced ATP synthesis and respiratory fluxes determined experimentally with isolated heart mitochondria. This capacity of F1Fo ATP synthase confers mitochondria with a significant energetic advantage compared to K+ transport through a channel not linked to oxidative phosphorylation (OxPhos). The K+-cycling mechanism requires a KHEmito that exchanges matrix K+ for intermembrane space H+, leaving PMF as the overall driving energy of OxPhos, in full agreement with the standard chemiosmotic mechanism. Experimental data of state 4➔3 energetic transitions, mimicking low to high energy demand, could be reproduced by an integrated computational model of mitochondrial function that incorporates the 'core' mechanism. Model simulations display similar behavior compared to the experimentally observed changes in ΔΨm, mitochondrial K+ uptake, matrix volume, respiration, and ATP synthesis during the energetic transitions at physiological pH and K+ concentration. The model also explores the role played by KHEmito in modulating the energetic performance of mitochondria. The results obtained support the available experimental evidence on ATP synthesis driven by K+ and H+ transport through the F1Fo ATP synthase.


Subject(s)
Mitochondrial Membranes , Potassium/metabolism , Protons , Adenosine Triphosphate , Computer Simulation , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism
3.
J Mol Cell Cardiol ; 151: 113-125, 2021 02.
Article in English | MEDLINE | ID: mdl-33301801

ABSTRACT

Heart failure (HF) is a progressive, debilitating condition characterized, in part, by altered ionic equilibria, increased ROS production and impaired cellular energy metabolism, contributing to variable profiles of systolic and diastolic dysfunction with significant functional limitations and risk of premature death. We summarize current knowledge concerning changes of intracellular Na+ and Ca2+ control mechanisms during the disease progression and their consequences on mitochondrial Ca2+ homeostasis and the shift in redox balance. Absent existing biological data, our computational modeling studies advance a new 'in silico' analysis to reconcile existing opposing views, based on different experimental HF models, regarding variations in mitochondrial Ca2+ concentration that participate in triggering and perpetuating oxidative stress in the failing heart and their impact on cardiac energetics. In agreement with our hypothesis and the literature, model simulations demonstrate the possibility that the heart's redox status together with cytoplasmic Na+ concentrations act as regulators of mitochondrial Ca2+ levels in HF and of the bioenergetics response that will ultimately drive ATP supply and oxidative stress. The resulting model predictions propose future directions to study the evolution of HF as well as other types of heart disease, and to develop novel testable mechanistic hypotheses that may lead to improved therapeutics.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans , Oxidation-Reduction , Oxidative Stress , Sodium/metabolism
4.
J Physiol ; 598(7): 1393-1415, 2020 04.
Article in English | MEDLINE | ID: mdl-30462352

ABSTRACT

KEY POINTS: Hearts from type 2 diabetic animals display perturbations in excitation-contraction coupling, impairing myocyte contractility and delaying relaxation, along with altered substrate consumption patterns. Under high glucose and ß-adrenergic stimulation conditions, palmitate can, at least in part, offset left ventricle (LV) dysfunction in hearts from diabetic mice, improving contractility and relaxation while restoring coronary perfusion pressure. Fluxome calculations of central catabolism in diabetic hearts show that, in the presence of palmitate, there is a metabolic remodelling involving tricarboxylic acid cycle, polyol and pentose phosphate pathways, leading to improved redox balance in cytoplasmic and mitochondrial compartments. Under high glucose and increased energy demand, the metabolic/fluxomic redirection leading to restored redox balance imparted by palmitate helps explain maintained LV function and may contribute to designing novel therapeutic approaches to prevent cardiac dysfunction in diabetic patients. ABSTRACT: Type-2 diabetes (T2DM) leads to reduced myocardial performance, and eventually heart failure. Excessive accumulation of lipids and glucose is central to T2DM cardiomyopathy. Previous data showed that palmitate (Palm) or glutathione preserved heart mitochondrial energy/redox balance under excess glucose, rescuing ß-adrenergic-stimulated cardiac excitation-contraction coupling. However, the mechanisms underlying the accompanying improved contractile performance have been largely ignored. Herein we explore in intact heart under substrate excess the metabolic remodelling associated with cardiac function in diabetic db/db mice subjected to stress given by ß-adrenergic stimulation with isoproterenol and high glucose compared to their non-diabetic controls (+/+, WT) under euglycaemic conditions. When perfused with Palm, T2DM hearts exhibited improved contractility/relaxation compared to WT, accompanied by extensive metabolic remodelling as demonstrated by metabolomics-fluxomics combined with bioinformatics and computational modelling. The T2DM heart metabolome showed significant differences in the abundance of metabolites in pathways related to glucose, lipids and redox metabolism. Using a validated computational model of heart's central catabolism, comprising glucose and fatty acid (FA) oxidation in cytoplasmic and mitochondrial compartments, we estimated that fluxes through glucose degradation pathways are ∼2-fold lower in heart from T2DM vs. WT under all conditions studied. Palm addition elicits improvement of the redox status via enhanced ß-oxidation and decreased glucose uptake, leading to flux-redirection away from redox-consuming pathways (e.g. polyol) while maintaining the flux through redox-generating pathways together with glucose-FA 'shared fuelling' of oxidative phosphorylation. Thus, available FAs such as Palm may help improve function via enhanced redox balance in T2DM hearts during peaks of hyperglycaemia and increased workload.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Heart , Humans , Mice , Myocardium/metabolism , Oxidation-Reduction
5.
Biochem J ; 476(21): 3109-3124, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31689353

ABSTRACT

Although the discovery of glycogen in the liver, attributed to Claude Bernard, happened more than 160 years ago, the mechanism involved in the initiation of glucose polymerization remained unknown. The discovery of glycogenin at the core of glycogen's structure and the initiation of its glucopolymerization is among one of the most exciting and relatively recent findings in Biochemistry. This review focuses on the initial steps leading to the seminal discoveries of proteoglycogen and glycogenin at the beginning of the 1980s, which paved the way for subsequent foundational breakthroughs that propelled forward this new research field. We also explore the current, as well as potential, impact this research field is having on human health and disease from the perspective of glycogen storage diseases. Important new questions arising from recent studies, their links to basic mechanisms involved in the de novo glycogen biogenesis, and the pervading presence of glycogenin across the evolutionary scale, fueled by high throughput -omics technologies, are also addressed.


Subject(s)
Glucosyltransferases/metabolism , Glycogen/metabolism , Glycoproteins/metabolism , Animals , Glucose/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glycogen/chemistry , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/genetics , Glycogen Storage Disease/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Humans , Liver/enzymology , Liver/metabolism , Polymerization
6.
FASEB J ; 32(7): 3844-3858, 2018 07.
Article in English | MEDLINE | ID: mdl-29485903

ABSTRACT

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.


Subject(s)
Food Deprivation , Physical Conditioning, Animal/methods , Physical Endurance , Animals , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Organelle Biogenesis
7.
PLoS Comput Biol ; 13(6): e1005588, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28598967

ABSTRACT

Lipids are main fuels for cellular energy and mitochondria their major oxidation site. Yet unknown is to what extent the fuel role of lipids is influenced by their uncoupling effects, and how this affects mitochondrial energetics, redox balance and the emission of reactive oxygen species (ROS). Employing a combined experimental-computational approach, we comparatively analyze ß-oxidation of palmitoyl CoA (PCoA) in isolated heart mitochondria from Sham and streptozotocin (STZ)-induced type 1 diabetic (T1DM) guinea pigs (GPs). Parallel high throughput measurements of the rates of oxygen consumption (VO2) and hydrogen peroxide (H2O2) emission as a function of PCoA concentration, in the presence of L-carnitine and malate, were performed. We found that PCoA concentration < 200 nmol/mg mito protein resulted in low H2O2 emission flux, increasing thereafter in Sham and T1DM GPs under both states 4 and 3 respiration with diabetic mitochondria releasing higher amounts of ROS. Respiratory uncoupling and ROS excess occurred at PCoA > 600 nmol/mg mito prot, in both control and diabetic animals. Also, for the first time, we show that an integrated two compartment mitochondrial model of ß-oxidation of long-chain fatty acids and main energy-redox processes is able to simulate the relationship between VO2 and H2O2 emission as a function of lipid concentration. Model and experimental results indicate that PCoA oxidation and its concentration-dependent uncoupling effect, together with a partial lipid-dependent decrease in the rate of superoxide generation, modulate H2O2 emission as a function of VO2. Results indicate that keeping low levels of intracellular lipid is crucial for mitochondria and cells to maintain ROS within physiological levels compatible with signaling and reliable energy supply.


Subject(s)
Diabetes Mellitus/metabolism , Lipid Metabolism , Mitochondria, Heart/metabolism , Models, Cardiovascular , Palmitoyl Coenzyme A/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Respiration , Cells, Cultured , Computer Simulation , Electron Transport , Guinea Pigs , Hydrogen Peroxide/metabolism , Male , Metabolism , Oxidation-Reduction , Oxygen/metabolism
8.
Adv Exp Med Biol ; 982: 1-24, 2017.
Article in English | MEDLINE | ID: mdl-28551779

ABSTRACT

The spatio-temporal organization of mitochondria in cardiac myocytes facilitates myocyte-wide, cluster-bound, mitochondrial inner membrane potential oscillatory depolarizations, commonly triggered by metabolic or oxidative stressors. Local intermitochondrial coupling can be mediated by reactive oxygen species (ROS) that activate inner membrane pores to initiate a ROS-induced-ROS-release process that produces synchronized limit cycle oscillations of mitochondrial clusters within the whole mitochondrial network. The network's dynamic organization, structure and function can be assessed by quantifying dynamic local coupling constants and dynamic functional clustering coefficients, both providing information about the network's response to external stimuli. In addition to its special organization, the mitochondrial network of cardiac myocytes exhibits substrate-sensitive coupling constants and clustering coefficients. The myocyte's ability to form functional clusters of synchronously oscillating mitochondria is sensitive to conditions such as substrate availability (e.g., glucose, pyruvate, ß-hydroxybutyrate), antioxidant status, respiratory chain activity, or history of oxidative challenge (e.g., ischemia-reperfusion). This underscores the relevance of quantitative methods to characterize the network's functional status as a way to assess the myocyte's resilience to pathological stressors.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Animals , Humans , Mitochondrial Dynamics , Models, Cardiovascular , Periodicity , Stochastic Processes , Time Factors
9.
Clin Sci (Lond) ; 130(15): 1285-305, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27358026

ABSTRACT

Food nutrients and metabolic supply-demand dynamics constitute environmental factors that interact with our genome influencing health and disease states. These gene-environment interactions converge at the metabolic-epigenome-genome axis to regulate gene expression and phenotypic outcomes. Mounting evidence indicates that nutrients and lifestyle strongly influence genome-metabolic functional interactions determining disease via altered epigenetic regulation. The mitochondrial network is a central player of the metabolic-epigenome-genome axis, regulating the level of key metabolites [NAD(+), AcCoA (acetyl CoA), ATP] acting as substrates/cofactors for acetyl transferases, kinases (e.g. protein kinase A) and deacetylases (e.g. sirtuins, SIRTs). The chromatin, an assembly of DNA and nucleoproteins, regulates the transcriptional process, acting at the epigenomic interface between metabolism and the genome. Within this framework, we review existing evidence showing that preservation of mitochondrial network function is directly involved in decreasing the rate of damage accumulation thus slowing aging and improving healthspan.


Subject(s)
Aging/metabolism , Energy Metabolism , Epigenesis, Genetic , Genome, Human , Health Status , Mitochondria/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Aging/genetics , Aging/pathology , Animals , Child , Child, Preschool , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Gene Expression Regulation , Gene-Environment Interaction , Humans , Infant , Infant, Newborn , Life Style , Longevity , Middle Aged , Mitochondria/genetics , Mitochondria/pathology , Mutation , Nutritional Status , Young Adult
10.
Biophys J ; 108(1): 163-72, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25564863

ABSTRACT

We describe a believed-novel procedure for translating metabolite profiles (metabolome) into the set of metabolic fluxes (fluxome) from which they originated. Methodologically, computational modeling is integrated with an analytical platform comprising linear optimization, continuation and dynamic analyses, and metabolic control. The procedure was tested with metabolite profiles obtained from ex vivo mice Langendorff-heart preparations perfused with glucose. The metabolic profiles were analyzed using a detailed kinetic model of the glucose catabolic pathways including glycolysis, pentose phosphate (PP), glycogenolysis, and polyols to translate the glucose metabolome of the heart into the fluxome. After optimization, the ability of the model to simulate the initial metabolite profile was confirmed, and metabolic fluxes as well as the structure of control and regulation of the glucose catabolic network could be calculated. We show that the step catalyzed by phosphofructokinase together with ATP demand and glycogenolysis exert the highest control on the glycolytic flux. The negative flux control exerted by phosphofructokinase on the PP and polyol pathways revealed that the extent of glycolytic flux directly affects flux redirection through these pathways, i.e., the higher the glycolytic flux the lower the PP and polyols. This believed-novel methodological approach represents a step forward that may help in designing therapeutic strategies targeted to diagnose, prevent, and treat metabolic diseases.


Subject(s)
Computer Simulation , Glucose/metabolism , Metabolome/physiology , Models, Biological , Myocardium/metabolism , Adenosine Triphosphatases/metabolism , Animals , Glycogenolysis , Glycolysis , Kinetics , Linear Models , Mice, Inbred C57BL , Mice, Transgenic , NAD/metabolism , NADP/metabolism , Pentose Phosphate Pathway , Polymers/metabolism , Tissue Culture Techniques
11.
Biophys J ; 108(8): 1922-33, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902432

ABSTRACT

Oscillatory behavior of mitochondrial inner membrane potential (ΔΨm) is commonly observed in cells subjected to oxidative or metabolic stress. In cardiac myocytes, the activation of inner membrane pores by reactive oxygen species (ROS) is a major factor mediating intermitochondrial coupling, and ROS-induced ROS release has been shown to underlie propagated waves of ΔΨm depolarization as well as synchronized limit cycle oscillations of ΔΨm in the network. The functional impact of ΔΨm instability on cardiac electrophysiology, Ca(2+) handling, and even cell survival, is strongly affected by the extent of such intermitochondrial coupling. Here, we employ a recently developed wavelet-based analytical approach to examine how different substrates affect mitochondrial coupling in cardiac cells, and we also determine the oscillatory coupling properties of mitochondria in ventricular cells in intact perfused hearts. The results show that the frequency of ΔΨm oscillations varies inversely with the size of the oscillating mitochondrial cluster, and depends on the strength of local intermitochondrial coupling. Time-varying coupling constants could be quantitatively determined by applying a stochastic phase model based on extension of the well-known Kuramoto model for networks of coupled oscillators. Cluster size-frequency relationships varied with different substrates, as did mitochondrial coupling constants, which were significantly larger for glucose (7.78 × 10(-2) ± 0.98 × 10(-2) s(-1)) and pyruvate (7.49 × 10(-2) ± 1.65 × 10(-2) s(-1)) than lactate (4.83 × 10(-2) ± 1.25 × 10(-2) s(-1)) or ß-hydroxybutyrate (4.11 × 10(-2) ± 0.62 × 10(-2) s(-1)). The findings indicate that mitochondrial spatiotemporal coupling and oscillatory behavior is influenced by substrate selection, perhaps through differing effects on ROS/redox balance. In particular, glucose-perfusion generates strong intermitochondrial coupling and temporal oscillatory stability. Pathological changes in specific catabolic pathways, which are known to occur during the progression of cardiovascular disease, could therefore contribute to altered sensitivity of the mitochondrial network to oxidative stress and emergent ΔΨm instability, ultimately scaling to produce organ level dysfunction.


Subject(s)
Membrane Potential, Mitochondrial , Mitochondria, Heart/metabolism , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Glucose/metabolism , Guinea Pigs , Heart Ventricles/cytology , Heart Ventricles/metabolism , Lactic Acid/metabolism , Myocytes, Cardiac/physiology , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
12.
Biochim Biophys Acta ; 1837(2): 287-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269780

ABSTRACT

The Redox-Optimized ROS Balance [R-ORB] hypothesis postulates that the redox environment [RE] is the main intermediary between mitochondrial respiration and reactive oxygen species [ROS]. According to R-ORB, ROS emission levels will attain a minimum vs. RE when respiratory rate (VO2) reaches a maximum following ADP stimulation, a tenet that we test herein in isolated heart mitochondria under forward electron transport [FET]. ROS emission increased two-fold as a function of changes in the RE (~400 to ~900mV·mM) in state 4 respiration elicited by increasing glutamate/malate (G/M). In G/M energized mitochondria, ROS emission decreases two-fold for RE ~500 to ~300mV·mM in state 3 respiration at increasing ADP. Stressed mitochondria released higher ROS, that was only weakly dependent on RE under state 3. As a function of VO2, the ROS dependence on RE was strong between ~550 and ~350mV·mM, when VO2 is maximal, primarily due to changes in glutathione redox potential. A similar dependence was observed with stressed mitochondria, but over a significantly more oxidized RE and ~3-fold higher ROS emission overall, as compared with non-stressed controls. We conclude that under non-stressful conditions mitochondrial ROS efflux decreases when the RE becomes less reduced within a range in which VO2 is maximal. These results agree with the R-ORB postulate that mitochondria minimize ROS emission as they maximize VO2 and ATP synthesis. This relationship is altered quantitatively, but not qualitatively, by oxidative stress although stressed mitochondria exhibit diminished energetic performance and increased ROS release.


Subject(s)
Mitochondria, Heart/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Animals , Cell Respiration , Energy Metabolism , Guinea Pigs , Hydrogen Peroxide/metabolism , Light , Membrane Potential, Mitochondrial , Oxidation-Reduction , Oxidative Stress , Scattering, Radiation , Substrate Specificity , Time Factors
13.
Am J Physiol Heart Circ Physiol ; 309(8): H1271-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26254336

ABSTRACT

Levels of the HER2/ErbB2 protein in the heart are upregulated in some women during breast cancer therapy, and these women are at high risk for developing heart dysfunction after sequential treatment with anti-ErbB2/trastuzumab or doxorubicin. Doxorubicin is known to increase oxidative stress in the heart, and thus we considered the possibility that ErbB2 protein influences the status of cardiac antioxidant defenses in cardiomyocytes. In this study, we measured reactive oxygen species (ROS) in cardiac mitochondria and whole hearts from mice with cardiac-specific overexpression of ErbB2 (ErbB2(tg)) and found that, compared with control mice, high levels of ErbB2 in myocardium result in lower levels of ROS in mitochondria (P = 0.0075) and whole hearts (P = 0.0381). Neonatal cardiomyocytes isolated from ErbB2(tg) hearts have lower ROS levels and less cellular death (P < 0.0001) following doxorubicin treatment. Analyzing antioxidant enzyme levels and activities, we found that ErbB2(tg) hearts have increased levels of glutathione peroxidase 1 (GPx1) protein (P < 0.0001) and GPx activity (P = 0.0031) in addition to increased levels of two known GPx activators, c-Abl (P = 0.0284) and Arg (P < 0.0001). Interestingly, although mitochondrial ROS emission is reduced in the ErbB2(tg) hearts, oxygen consumption rates and complex I activity are similar to control littermates. Compared with these in vivo studies, H9c2 cells transfected with ErbB2 showed less cellular toxicity and produced less ROS (P < 0.0001) after doxorubicin treatment but upregulated GR activity (P = 0.0237) instead of GPx. Our study shows that ErbB2-dependent signaling contributes to antioxidant defenses and suggests a novel mechanism by which anticancer therapies involving ErbB2 antagonists can harm myocardial structure and function.


Subject(s)
Antineoplastic Agents/toxicity , Antioxidants/metabolism , Doxorubicin/toxicity , Glutathione Peroxidase/metabolism , Heart Diseases/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Line , Dose-Response Relationship, Drug , Glutathione Reductase/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/genetics , Heart Diseases/pathology , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Rats , Receptor, ErbB-2/genetics , Glutathione Peroxidase GPX1
14.
Am J Physiol Heart Circ Physiol ; 308(4): H291-302, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25485897

ABSTRACT

Hearts from type 2 diabetic (T2DM) subjects are chronically subjected to hyperglycemia and hyperlipidemia, both thought to contribute to oxidizing conditions and contractile dysfunction. How redox alterations and contractility interrelate, ultimately diminishing T2DM heart function, remains poorly understood. Herein we tested whether the fatty acid palmitate (Palm), in addition to its energetic contribution, rescues function by improving redox [glutathione (GSH), NAD(P)H, less oxidative stress] in T2DM rat heart trabeculae subjected to high glucose. Using cardiac trabeculae from Zucker Diabetic Fatty (ZDF) rats, we assessed the impact of low glucose (EG) and high glucose (HG), in absence or presence of Palm or insulin, on force development, energetics, and redox responses. We found that in EG ZDF and lean trabeculae displayed similar contractile work, yield of contractile work (Ycw), representing the ratio of force time integral over rate of O2 consumption. Conversely, HG had a negative impact on Ycw, whereas Palm, but not insulin, completely prevented contractile loss. This effect was associated with higher GSH, less oxidative stress, and augmented matrix GSH/thioredoxin (Trx) in ZDF mitochondria. Restoration of myocardial redox with GSH ethyl ester also rescued ZDF contractile function in HG, independently from Palm. These results support the idea that maintained redox balance, via increased GSH and Trx antioxidant activities to resist oxidative stress, is an essential protective response of the diabetic heart to keep contractile function.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Myocardial Contraction , Myocardium/metabolism , Oxidative Stress , Animals , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/physiopathology , Glutathione/metabolism , Heart/drug effects , Heart/physiopathology , Insulin/blood , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Oxygen Consumption , Palmitates/blood , Palmitates/pharmacology , Rats , Rats, Zucker , Thioredoxins/metabolism
15.
Clin Sci (Lond) ; 129(7): 561-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26186741

ABSTRACT

In Type I diabetic (T1DM) patients, both peaks of hyperglycaemia and increased sympathetic tone probably contribute to impair systolic and diastolic function. However, how these stressors eventually alter cardiac function during T1DM is not fully understood. In the present study, we hypothesized that impaired mitochondrial energy supply and excess reactive oxygen species (ROS) emission is centrally involved in T1DM cardiac dysfunction due to metabolic/redox stress and aimed to determine the mitochondrial sites implicated in these alterations. To this end, we used isolated myocytes and mitochondria from Sham and streptozotocin (STZ)-induced T1DM guinea pigs (GPs), untreated or treated with insulin. Relative to controls, T1DM myocytes exhibited higher oxidative stress when challenged with high glucose (HG) combined with ß-adrenergic stimulation [via isoprenaline (isoproterenol) (ISO)], leading to contraction/relaxation deficits. T1DM mitochondria had decreased respiration with complex II and IV substrates and markedly lower ADP phosphorylation rates and higher H2O2 emission when challenged with oxidants to mimic the more oxidized redox milieu present in HG + ISO-treated cardiomyocytes. Since in T1DM hearts insulin-sensitivity is preserved and a glucose-to-fatty acid (FA) shift occurs, we next tested whether insulin therapy or acute palmitate (Palm) infusion prevents HG + ISO-induced cardiac dysfunction. We found that insulin rescued proper cardiac redox balance, but not mitochondrial respiration or contractile performance. Conversely, Palm restored redox balance and preserved myocyte function. Thus, stressors such as peaks of HG and adrenergic hyperactivity impair mitochondrial respiration, hampering energy supply while exacerbating ROS emission. Our study suggests that an ideal therapeutic measure to treat metabolically/redox-challenged T1DM hearts should concomitantly correct energetic and redox abnormalities to fully maintain cardiac function.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Hydrogen Peroxide/chemistry , Mitochondria/metabolism , Animals , Blood Glucose/metabolism , Calcium/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Guinea Pigs , Insulin/metabolism , Male , Microscopy, Fluorescence , Mitochondria, Heart/metabolism , Muscle Cells/cytology , Muscle Contraction , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Oxygen/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Receptors, Adrenergic, beta/metabolism , Sarcomeres/metabolism
16.
J Pharmacol Exp Ther ; 349(1): 21-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24431470

ABSTRACT

The effect of inhalational anesthetics on myocardial contraction and energetics in type 2 diabetes mellitus is unknown. We investigated the effect of isoflurane (ISO) on force and intracellular Ca(2+) transient (iCa), myocardial oxygen consumption (MVo(2)), and energetics/redox behavior in trabecular muscles from Zucker diabetic fatty (ZDF) rats. At baseline, force and corresponding iCa were lower in ZDF trabeculae than in controls. ISO decreased force in both groups in a dose-dependent manner. ISO did not affect iCa amplitude in controls, but ISO > 1.5% significantly reduced iCa amplitude in ZDF trabeculae. ISO-induced force depression fully recovered as a result of increased iCa when external Ca(2+) was raised in controls. However, both force and iCa remained low in ZDF muscle at elevated external Ca(2+). In controls, force, iCa, and MVo(2) increased when stimulation frequency was increased from 0.5 to 1.5 Hz. ZDF muscles, however, exhibited blunted responses in force and iCa and decreased MVo(2). Oxidative stress levels were unchanged in control muscles but increased significantly in ZDF muscles after exposure to ISO. Finally, the depressive effect of ISO was prevented by 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (Tempol) in ZDF muscles. These findings suggest that ISO dose-dependently attenuates force in control and ZDF muscles with differential effect on iCa. The mechanism of force depression by ISO in controls is mainly decreased myofilament Ca(2+) sensitivity, whereas in ZDF muscles the ISO-induced decrease in contraction is due to worsening oxidative stress, which inhibits iCa and force development.


Subject(s)
Anesthetics, Inhalation/adverse effects , Diabetes Mellitus, Experimental/metabolism , Energy Metabolism/drug effects , Isoflurane/adverse effects , Myocardium/metabolism , Obesity/metabolism , Oxidative Stress/drug effects , Anesthetics, Inhalation/administration & dosage , Animals , Calcium/metabolism , Diabetes Mellitus, Experimental/complications , Dose-Response Relationship, Drug , Isoflurane/administration & dosage , Obesity/complications , Oxygen Consumption/drug effects , Rats , Rats, Zucker
17.
Biophys J ; 104(2): 332-43, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23442855

ABSTRACT

To understand the mechanisms involved in the control and regulation of mitochondrial reactive oxygen species (ROS) levels, a two-compartment computational mitochondrial energetic-redox (ME-R) model accounting for energetic, redox, and ROS metabolisms is presented. The ME-R model incorporates four main redox couples (NADH/NAD(+), NADPH/NADP(+), GSH/GSSG, Trx(SH)(2)/TrxSS). Scavenging systems-glutathione, thioredoxin, superoxide dismutase, catalase-are distributed in mitochondrial matrix and extra-matrix compartments, and transport between compartments of ROS species (superoxide: O(2)(⋅-), hydrogen peroxide: H(2)O(2)), and GSH is also taken into account. Model simulations are compared with experimental data obtained from isolated heart mitochondria. The ME-R model is able to simulate: i), the shape and order of magnitude of H(2)O(2) emission and dose-response kinetics observed after treatment with inhibitors of the GSH or Trx scavenging systems and ii), steady and transient behavior of ΔΨ(m) and NADH after single or repetitive pulses of substrate- or uncoupler-elicited energetic-redox transitions. The dynamics of the redox environment in both compartments is analyzed with the model following substrate addition. The ME-R model represents a useful computational tool for exploring ROS dynamics, the role of compartmentation in the modulation of the redox environment, and how redox regulation participates in the control of mitochondrial function.


Subject(s)
Cell Compartmentation , Energy Metabolism , Metabolic Networks and Pathways , Mitochondria, Heart/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Cell Respiration , Computer Simulation , Glutathione/metabolism , Guinea Pigs , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial , NAD/metabolism , Oxidation-Reduction , Reproducibility of Results , Substrate Specificity , Thioredoxins/metabolism , Time Factors
18.
Am J Physiol Heart Circ Physiol ; 304(7): H916-26, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23376824

ABSTRACT

Chronic hyperglycemia in type-1 diabetes mellitus is associated with oxidative stress (OS) and sudden death. Mechanistic links remain unclear. We investigated changes in electrophysiological (EP) properties in a model of chronic hyperglycemia before and after challenge with OS by GSH oxidation and tested reversibility of EP remodeling by insulin. Guinea pigs survived for 1 mo following streptozotocin (STZ) or saline (sham) injection. A treatment group received daily insulin for 2 wk to reverse STZ-induced hyperglycemia (STZ + Ins). EP properties were measured using high-resolution optical action potential mapping before and after challenge of hearts with diamide. Despite elevation of glucose levels in STZ compared with sham-operated (P = 0.004) and STZ + Ins (P = 0.002) animals, average action potential duration (APD) and arrhythmia propensity were not altered at baseline. Diamide promoted early (<10 min) formation of arrhythmic triggers reflected by a higher arrhythmia scoring index in STZ (P = 0.045) and STZ + Ins (P = 0.033) hearts compared with sham-operated hearts. APD heterogeneity underwent a more pronounced increase in response to diamide in STZ and STZ + Ins hearts compared with sham-operated hearts. Within 30 min, diamide resulted in spontaneous incidence of ventricular tachycardia and ventricular fibrillation (VT/VF) in 3/6, 2/5, 1/5, and 0/4 STZ, STZ + Ins, sham-operated, and normal hearts, respectively. Hearts prone to VT/VF exhibited greater APD heterogeneity (P = 0.010) compared with their VT/VF-free counterparts. Finally, altered EP properties in STZ were not rescued by insulin. In conclusion, GSH oxidation enhances APD heterogeneity and increases arrhythmia scoring index in a guinea pig model of chronic hyperglycemia. Despite normalization of glycemic levels by insulin, these proarrhythmic properties are not reversed, suggesting the importance of targeting antioxidant defenses for arrhythmia suppression.


Subject(s)
Glutathione/metabolism , Hyperglycemia/complications , Oxidative Stress , Tachycardia, Ventricular/metabolism , Ventricular Fibrillation/metabolism , Action Potentials/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Blood Glucose/metabolism , Diamide/pharmacology , Guinea Pigs , Heart/physiopathology , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Insulin/pharmacology , Oxidation-Reduction , Propensity Score , Streptozocin/pharmacology , Tachycardia, Ventricular/etiology , Ventricular Fibrillation/etiology , Ventricular Remodeling/drug effects
19.
Proc Natl Acad Sci U S A ; 107(32): 14315-20, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20656937

ABSTRACT

Mitochondrial networks in cardiac myocytes under oxidative stress show collective (cluster) behavior through synchronization of their inner membrane potentials (DeltaPsi(m)). However, it is unclear whether the oscillation frequency and coupling strength between individual mitochondria affect the size of the cluster and vice versa. We used the wavelet transform and developed advanced signal processing tools that allowed us to capture individual mitochondrial DeltaPsi(m) oscillations in cardiac myocytes and examine their dynamic spatio-temporal properties. Heterogeneous frequency behavior prompted us to sort mitochondria according to their frequencies. Signal analysis of the mitochondrial network showed an inverse relationship between cluster size and cluster frequency as well as between cluster amplitude and cluster size. High cross-correlation coefficients between neighboring mitochondria clustered longitudinally along the myocyte striations, indicated anisotropic communication between mitochondria. Isochronal mapping of the onset of myocyte-wide DeltaPsi(m) depolarization further exemplified heterogeneous DeltaPsi(m) among mitochondria. Taken together, the results suggest that frequency and amplitude modulation of clusters of synchronized mitochondria arises by means of strong changes in local coupling between neighboring mitochondria.


Subject(s)
Biological Clocks , Mitochondria/physiology , Myocytes, Cardiac/cytology , Animals , Guinea Pigs , Mitochondria/ultrastructure , Myocytes, Cardiac/physiology , Oxidative Stress
20.
Lab Anim (NY) ; 52(12): 289-314, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38017182

ABSTRACT

The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.


Subject(s)
Artificial Intelligence , Big Data , Animals , Geroscience , Computational Biology/methods , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL