Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38260287

ABSTRACT

Background: Cardiac risk rises during acute SARS-CoV-2 infection and in long COVID syndrome in humans, but the mechanisms behind COVID-19-linked arrhythmias are unknown. This study explores the acute and long term effects of SARS-CoV-2 on the cardiac conduction system (CCS) in a hamster model of COVID-19. Methods: Radiotelemetry in conscious animals was used to non-invasively record electrocardiograms and subpleural pressures after intranasal SARS-CoV-2 infection. Cardiac cytokines, interferon-stimulated gene expression, and macrophage infiltration of the CCS, were assessed at 4 days and 4 weeks post-infection. A double-stranded RNA mimetic, polyinosinic:polycytidylic acid (PIC), was used in vivo and in vitro to activate viral pattern recognition receptors in the absence of SARS-CoV-2 infection. Results: COVID-19 induced pronounced tachypnea and severe cardiac conduction system (CCS) dysfunction, spanning from bradycardia to persistent atrioventricular block, although no viral protein expression was detected in the heart. Arrhythmias developed rapidly, partially reversed, and then redeveloped after the pulmonary infection was resolved, indicating persistent CCS injury. Increased cardiac cytokines, interferon-stimulated gene expression, and macrophage remodeling in the CCS accompanied the electrophysiological abnormalities. Interestingly, the arrhythmia phenotype was reproduced by cardiac injection of PIC in the absence of virus, indicating that innate immune activation was sufficient to drive the response. PIC also strongly induced cytokine secretion and robust interferon signaling in hearts, human iPSC-derived cardiomyocytes (hiPSC-CMs), and engineered heart tissues, accompanied by alterations in electrical and Ca 2+ handling properties. Importantly, the pulmonary and cardiac effects of COVID-19 were blunted by in vivo inhibition of JAK/STAT signaling or by a mitochondrially-targeted antioxidant. Conclusions: The findings indicate that long term dysfunction and immune cell remodeling of the CCS is induced by COVID-19, arising indirectly from oxidative stress and excessive activation of cardiac innate immune responses during infection, with implications for long COVID Syndrome.

2.
J Biol Chem ; 299(6): 104708, 2023 06.
Article in English | MEDLINE | ID: mdl-37061004

ABSTRACT

Physiologic Ca2+ entry via the Mitochondrial Calcium Uniporter (MCU) participates in energetic adaption to workload but may also contribute to cell death during ischemia/reperfusion (I/R) injury. The MCU has been identified as the primary mode of Ca2+ import into mitochondria. Several groups have tested the hypothesis that Ca2+ import via MCU is detrimental during I/R injury using genetically-engineered mouse models, yet the results from these studies are inconclusive. Furthermore, mitochondria exhibit unstable or oscillatory membrane potentials (ΔΨm) when subjected to stress, such as during I/R, but it is unclear if the primary trigger is an excess influx of mitochondrial Ca2+ (mCa2+), reactive oxygen species (ROS) accumulation, or other factors. Here, we critically examine whether MCU-mediated mitochondrial Ca2+ uptake during I/R is involved in ΔΨm instability, or sustained mitochondrial depolarization, during reperfusion by acutely knocking out MCU in neonatal mouse ventricular myocyte (NMVM) monolayers subjected to simulated I/R. Unexpectedly, we find that MCU knockout does not significantly alter mCa2+ import during I/R, nor does it affect ΔΨm recovery during reperfusion. In contrast, blocking the mitochondrial sodium-calcium exchanger (mNCE) suppressed the mCa2+ increase during Ischemia but did not affect ΔΨm recovery or the frequency of ΔΨm oscillations during reperfusion, indicating that mitochondrial ΔΨm instability on reperfusion is not triggered by mCa2+. Interestingly, inhibition of mitochondrial electron transport or supplementation with antioxidants stabilized I/R-induced ΔΨm oscillations. The findings are consistent with mCa2+ overload being mediated by reverse-mode mNCE activity and supporting ROS-induced ROS release as the primary trigger of ΔΨm instability during reperfusion injury.


Subject(s)
Mitochondria, Heart , Reperfusion Injury , Mice , Animals , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial , Mitochondria, Heart/metabolism , Ischemia/metabolism , Reperfusion Injury/metabolism , Reperfusion , Calcium/metabolism
3.
J Biol Chem ; 299(3): 102907, 2023 03.
Article in English | MEDLINE | ID: mdl-36642184

ABSTRACT

The dynamic cycling of O-linked GlcNAc (O-GlcNAc) on and off Ser/Thr residues of intracellular proteins, termed O-GlcNAcylation, is mediated by the conserved enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase. O-GlcNAc cycling is important in homeostatic and stress responses, and its perturbation sensitizes the heart to ischemic and other injuries. Despite considerable progress, many molecular pathways impacted by O-GlcNAcylation in the heart remain unclear. The mitogen-activated protein kinase (MAPK) pathway is a central signaling cascade that coordinates developmental, physiological, and pathological responses in the heart. The developmental or adaptive arm of MAPK signaling is primarily mediated by Erk kinases, while the pathophysiologic arm is mediated by p38 and Jnk kinases. Here, we examine whether O-GlcNAcylation affects MAPK signaling in cardiac myocytes, focusing on Erk1/2 and p38 in basal and hypertrophic conditions induced by phenylephrine. Using metabolic labeling of glycans coupled with alkyne-azide "click" chemistry, we found that Erk1/2 and p38 are O-GlcNAcylated. Supporting the regulation of p38 by O-GlcNAcylation, the OGT inhibitor, OSMI-1, triggers the phosphorylation of p38, an event that involves the NOX2-Ask1-MKK3/6 signaling axis and also the noncanonical activator Tab1. Additionally, OGT inhibition blocks the phenylephrine-induced phosphorylation of Erk1/2. Consistent with perturbed MAPK signaling, OSMI-1-treated cardiomyocytes have a blunted hypertrophic response to phenylephrine, decreased expression of cTnT (key component of the contractile apparatus), and increased expression of maladaptive natriuretic factors Anp and Bnp. Collectively, these studies highlight new roles for O-GlcNAcylation in maintaining a balanced activity of Erk1/2 and p38 MAPKs during hypertrophic growth responses in cardiomyocytes.


Subject(s)
Myocytes, Cardiac , Signal Transduction , Humans , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Phosphorylation , Hypertrophy/metabolism , Proteins/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Acetylglucosamine/metabolism
4.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34618683

ABSTRACT

Central obesity with cardiometabolic syndrome (CMS) is a major global contributor to human disease, and effective therapies are needed. Here, we show that cyclic GMP-selective phosphodiesterase 9A inhibition (PDE9-I) in both male and ovariectomized female mice suppresses preestablished severe diet-induced obesity/CMS with or without superimposed mild cardiac pressure load. PDE9-I reduces total body, inguinal, hepatic, and myocardial fat; stimulates mitochondrial activity in brown and white fat; and improves CMS, without significantly altering activity or food intake. PDE9 localized at mitochondria, and its inhibition in vitro stimulated lipolysis in a PPARα-dependent manner and increased mitochondrial respiration in both adipocytes and myocytes. PPARα upregulation was required to achieve the lipolytic, antiobesity, and metabolic effects of PDE9-I. All these PDE9-I-induced changes were not observed in obese/CMS nonovariectomized females, indicating a strong sexual dimorphism. We found that PPARα chromatin binding was reoriented away from fat metabolism-regulating genes when stimulated in the presence of coactivated estrogen receptor-α, and this may underlie the dimorphism. These findings have translational relevance given that PDE9-I is already being studied in humans for indications including heart failure, and efficacy against obesity/CMS would enhance its therapeutic utility.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adipose Tissue/embryology , Metabolic Syndrome/enzymology , Obesity/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Female , Male , Metabolic Syndrome/genetics , Mice , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/genetics , Obesity/genetics , PPAR alpha/genetics , PPAR alpha/metabolism
5.
Biophys J ; 120(16): 3261-3271, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34297964

ABSTRACT

Mitochondria exhibit unstable inner membrane potentials (ΔΨm) when subjected to stress, such as during ischemia/reperfusion (I/R). Understanding the mechanism of ΔΨm instability involves characterizing and quantifying this phenomenon in an unbiased and reproducible manner. Here, we describe a simple analytical workflow called "MitoWave" that combines wavelet transform methods and image segmentation to unravel dynamic ΔΨm changes in the cardiac mitochondrial network during I/R. In vitro ischemia was affected by placing a glass coverslip on a monolayer of neonatal mouse ventricular myocytes for 1 h and removing the coverslip to allow for reperfusion, revealing complex oscillatory ΔΨm. MitoWave analysis was then used to identify individual mitochondrial clusters within the cells and track their intrinsic oscillation frequencies over the course of reperfusion. Responses segregated into five typical behaviors were quantified by MitoWave that were corroborated by visual inspection of the time series. Statistical analysis of the distribution of oscillating mitochondrial clusters during reperfusion showed significant differences between the five different outcomes. Features such as the time point of ΔΨm depolarization during I/R, area of mitochondrial clusters, and time-resolved frequency components during reperfusion were determined per cell and per mitochondrial cluster. Mitochondria from neonatal mouse ventricular myocytes subjected to I/R oscillate in the frequency range of 8.6-45 mHz, with a mean of 8.73 ± 4.35 mHz. Oscillating clusters had smaller areas ranging from 49.8 ± 1.2 µm2, whereas nonoscillating clusters had larger areas 66 ± 1.5 µm2. A negative correlation between frequency and mitochondrial cluster area was observed. We also observed that late ΔΨm loss during ischemia correlated with early ΔΨm stabilization after oscillation on reperfusion. Thus, MitoWave analysis provides a semiautomated method to quantify complex time-resolved mitochondrial behavior in an easy-to-follow workflow, enabling unbiased, reproducible quantitation of complex nonstationary cellular phenomena.


Subject(s)
Ischemia , Myocytes, Cardiac , Animals , Ischemia/metabolism , Membrane Potential, Mitochondrial , Mice , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Reperfusion , Spatio-Temporal Analysis
6.
J Mol Cell Cardiol ; 151: 126-134, 2021 02.
Article in English | MEDLINE | ID: mdl-33290770

ABSTRACT

Ca2+ serves as a ubiquitous second messenger mediating a variety of cellular processes including electrical excitation, contraction, gene expression, secretion, cell death and others. The identification of the molecular components of the mitochondrial Ca2+ influx and efflux pathways has created a resurgent interest in the regulation of mitochondrial Ca2+ balance and its physiological and pathophysiological roles. While the pace of discovery has quickened with the availability of new cellular and animal models, many fundamental questions remain to be answered regarding the regulation and functional impact of mitochondrial Ca2+ in health and disease. This review highlights several experimental observations pertaining to key aspects of mitochondrial Ca2+ homeostasis that remain enigmatic, particularly whether mitochondrial Ca2+ signaling is depressed or excessive in heart failure, which will determine the optimal approach to therapeutic intervention.


Subject(s)
Calcium/metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Animals , Heart Failure/physiopathology , Humans , Ion Transport , Oxidation-Reduction , Reactive Oxygen Species/metabolism
7.
J Mol Cell Cardiol ; 144: 87-96, 2020 07.
Article in English | MEDLINE | ID: mdl-32442661

ABSTRACT

AIMS: In cardiomyocytes, there is microRNA (miR) in the mitochondria that originates from the nuclear genome and matures in the cytoplasm before translocating into the mitochondria. Overexpression of one such miR, miR-181c, can lead to heart failure by stimulating reactive oxygen species (ROS) production and increasing mitochondrial calcium level ([Ca2+]m). Mitochondrial calcium uptake 1 protein (MICU1), a regulatory protein in the mitochondrial calcium uniporter complex, plays an important role in regulating [Ca2+]m. Obesity results in miR-181c overexpression and a decrease in MICU1. We hypothesize that lowering miR-181c would protect against obesity-induced cardiac dysfunction. METHODS AND RESULTS: We used an in vivo mouse model of high-fat diet (HFD) for 18 weeks and induced high lipid load in H9c2 cells with oleate-conjugated bovine serum albumin in vitro. We tested the cardioprotective role of lowering miR-181c by using miR-181c/d-/- mice (in vivo) and AntagomiR against miR-181c (in vitro). HFD significantly upregulated heart levels of miR-181c and led to cardiac hypertrophy in wild-type mice, but not in miR-181c/d-/- mice. HFD also increased ROS production and pyruvate dehydrogenase activity (a surrogate for [Ca2+]m), but the increases were alleviated in miR-181c/d-/- mice. Moreover, miR-181c/d-/- mice fed a HFD had higher levels of MICU1 than did wild-type mice fed a HFD, attenuating the rise in [Ca2+]m. Overexpression of miR-181c in neonatal ventricular cardiomyocytes (NMVM) caused increased ROS production, which oxidized transcription factor Sp1 and led to a loss of Sp1, thereby slowing MICU1 transcription. Hence, miR-181c increases [Ca2+]m through Sp1 oxidation and downregulation of MICU1, suggesting that the cardioprotective effect of miR-181c/d-/- results from inhibition of Sp1 oxidation. CONCLUSION: This study has identified a unique nuclear-mitochondrial communication mechanism in the heart orchestrated by miR-181c. Obesity-induced overexpression of miR-181c increases [Ca2+]m via downregulation of MICU1 and leads to cardiac injury. A strategy to inhibit miR-181c in cardiomyocytes can preserve cardiac function during obesity by improving mitochondrial function. Altering miR-181c expression may provide a pharmacologic approach to improve cardiomyopathy in individuals with obesity/type 2 diabetes.


Subject(s)
Cell Nucleus/metabolism , MicroRNAs/genetics , Mitochondria, Heart/metabolism , Obesity/genetics , Obesity/metabolism , Ventricular Dysfunction/etiology , Ventricular Dysfunction/metabolism , Animals , Biomarkers , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/metabolism , Obesity/complications , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/metabolism , Ventricular Dysfunction/physiopathology
8.
J Mol Cell Cardiol ; 139: 176-189, 2020 02.
Article in English | MEDLINE | ID: mdl-32004507

ABSTRACT

The renal-outer-medullary­potassium (ROMK) channel, mutated in Bartter's syndrome, regulates ion exchange in kidney, but its extra-renal functions remain unknown. Additionally, ROMK was postulated to be the pore-forming subunit of the mitochondrial ATP-sensitive K+ channel (mitoKATP), a mediator of cardioprotection. Using global and cardiomyocyte-specific knockout mice (ROMK-GKO and ROMK-CKO respectively), we characterize the effects of ROMK knockout on mitochondrial ion handling, the response to pharmacological KATP channel modulators, and ischemia/reperfusion (I/R) injury. Mitochondria from ROMK-GKO hearts exhibited a lower threshold for Ca2+-triggered permeability transition pore (mPTP) opening but normal matrix volume changes during oxidative phosphorylation. Isolated perfused ROMK-GKO hearts exhibited impaired functional recovery and increased infarct size when I/R was preceded by an ischemic preconditioning (IPC) protocol. Because ROMK-GKO mice exhibited severe renal defects and cardiac remodeling, we further characterized ROMK-CKO hearts to avoid confounding systemic effects. Mitochondria from ROMK-CKO hearts had unchanged matrix volume responses during oxidative phosphorylation and still swelled upon addition of a mitoKATP opener, but exhibited a lower threshold for mPTP opening, similar to GKO mitochondria. Nevertheless, I/R induced damage was not exacerbated in ROMK-CKO hearts, either ex vivo or in vivo. Lastly, we examined the response of ROMK-CKO hearts to ex vivo I/R injury with or without IPC and found that IPC still protected these hearts, suggesting that cardiomyocyte ROMK does not participate significantly in the cardioprotective pathway elicited by IPC. Collectively, our findings from these novel strains of mice suggest that cardiomyocyte ROMK is not a central mediator of mitoKATP function, although it can affect mPTP activation threshold.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels/metabolism , Animals , Animals, Newborn , CRISPR-Cas Systems/genetics , Calcium/metabolism , Electrophysiological Phenomena , Gene Editing , Gene Knockout Techniques , Hemodynamics , Ischemic Preconditioning, Myocardial , Mice, Knockout , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Specificity , Perfusion , Phenotype , Potassium Channels, Inwardly Rectifying/metabolism
9.
PLoS One ; 9(3): e91005, 2014.
Article in English | MEDLINE | ID: mdl-24614678

ABSTRACT

Reliable and valid biomarkers of ageing (BoA) are needed to understand mechanisms, test interventions and predict the timing of adverse health events associated with ageing. Since increased reactive oxygen species (ROS) production and mitochondrial dysfunction are consequences of cellular senescence and may contribute causally to the ageing of organisms, we focused on these parameters as candidate BoA. Superoxide levels, mitochondrial mass and mitochondrial membrane potential in human peripheral blood mononuclear cells (PBMCs) and subpopulations (lymphocytes and monocytes) were measured in participants from the Newcastle 85+ study, a population-based study of the very old (aged 85 years and older). The intra- and inter-assay precision expressed as coefficient of variation (CV) for all parameters was acceptable (3% to 12% and 5 to 22% respectively). All parameters were stable in the short-term (1 week interval) in a sample of control individuals in the PBMCs and lymphocyte subpopulation, however they were unstable in the monocyte subpopulation; this rendered monocytes unreliable for further analysis. There was a significant association between superoxide levels and mitochondrial mass (positive in lymphocytes, p = 0.01) and between superoxide levels and mitochondrial membrane potential (negative in PBMCs, p = 0.01; positive in lymphocytes, p = 0.05). There were also significant associations between superoxide levels and mitochondrial parameters with other markers of oxidative stress-induced cellular senescence (p≤0.04), however some were in the opposite direction to expected. No associations were found between the measured parameters and age-related outcomes, including cognitive impairment, disability, co-morbidity and survival - questioning the validity of these parameters as candidate BoA in the very old.


Subject(s)
Aging/metabolism , Biomarkers/metabolism , Leukocytes/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Aged, 80 and over , Cell Survival , Cellular Senescence , Humans , Leukocytes, Mononuclear/metabolism , Membrane Potential, Mitochondrial , Oxidative Stress , Reproducibility of Results , Superoxides/metabolism
10.
PLoS One ; 9(2): e89938, 2014.
Article in English | MEDLINE | ID: mdl-24587137

ABSTRACT

Mitochondrial sn-glycerol 3-phosphate dehydrogenase (mGPDH) is a ubiquinone-linked enzyme in the mitochondrial inner membrane best characterized as part of the glycerol phosphate shuttle that transfers reducing equivalents from cytosolic NADH into the mitochondrial electron transport chain. Despite the widespread expression of mGPDH and the availability of mGPDH-null mice, the physiological role of this enzyme remains poorly defined in many tissues, likely because of compensatory pathways for cytosolic regeneration of NAD⁺ and mechanisms for glycerol phosphate metabolism. Here we describe a novel class of cell-permeant small-molecule inhibitors of mGPDH (iGP) discovered through small-molecule screening. Structure-activity analysis identified a core benzimidazole-phenyl-succinamide structure as being essential to inhibition of mGPDH while modifications to the benzimidazole ring system modulated both potency and off-target effects. Live-cell imaging provided evidence that iGPs penetrate cellular membranes. Two compounds (iGP-1 and iGP-5) were characterized further to determine potency and selectivity and found to be mixed inhibitors with IC50 and K(i) values between ∼1-15 µM. These novel mGPDH inhibitors are unique tools to investigate the role of glycerol 3-phosphate metabolism in both isolated and intact systems.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glycerol-3-Phosphate Dehydrogenase (NAD+)/antagonists & inhibitors , Mitochondrial Membranes/metabolism , Amides/chemistry , Amides/metabolism , Animals , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Fluorescence , Inhibitory Concentration 50 , Mice , Models, Biological , Molecular Structure , Muscle, Skeletal/cytology , Structure-Activity Relationship , Succinates/chemistry , Succinates/metabolism
11.
Free Radic Biol Med ; 65: 1047-1059, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994103

ABSTRACT

Mitochondrial production of reactive oxygen species is often considered an unavoidable consequence of aerobic metabolism and currently cannot be manipulated without perturbing oxidative phosphorylation. Antioxidants are widely used to suppress effects of reactive oxygen species after formation, but they can never fully prevent immediate effects at the sites of production. To identify site-selective inhibitors of mitochondrial superoxide/H2O2 production that do not interfere with mitochondrial energy metabolism, we developed a robust small-molecule screen and secondary profiling strategy. We describe the discovery and characterization of a compound (N-cyclohexyl-4-(4-nitrophenoxy)benzenesulfonamide; CN-POBS) that selectively inhibits superoxide/H2O2 production from the ubiquinone-binding site of complex I (site I(Q)) with no effects on superoxide/H2O2 production from other sites or on oxidative phosphorylation. Structure/activity studies identified a core structure that is important for potency and selectivity for site I(Q). By employing CN-POBS in mitochondria respiring on NADH-generating substrates, we show that site I(Q) does not produce significant amounts of superoxide/H2O2 during forward electron transport on glutamate plus malate. Our screening platform promises to facilitate further discovery of direct modulators of mitochondrially derived oxidative damage and advance our ability to understand and manipulate mitochondrial reactive oxygen species production under both normal and pathological conditions.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria, Muscle/metabolism , Reactive Oxygen Species/metabolism , Animals , Binding Sites , Female , High-Throughput Screening Assays , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Muscle/drug effects , Oxidation-Reduction , Rats, Wistar , Ubiquinone/metabolism
12.
Neurobiol Aging ; 34(12): 2889.e1-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23639206

ABSTRACT

Inherited genetic variation of mitochondrial DNA (mtDNA) could account for the missing heritability of human longevity and healthy aging. Here, we show no robust association between common genetic variants of mtDNA and frailty (an "unhealthy aging" phenotype) or mortality in 700, more than 85-year-old, participants of the Newcastle 85+ study. Conflicting data from different populations underscore our conclusion that there is currently no compelling link between inherited mtDNA variants and aging.


Subject(s)
Aging/genetics , DNA, Mitochondrial/genetics , Haplotypes/genetics , Longevity/genetics , Aged, 80 and over , Cohort Studies , Frail Elderly , Genetic Variation , Humans
13.
PLoS One ; 6(7): e21746, 2011.
Article in English | MEDLINE | ID: mdl-21799747

ABSTRACT

Recently developed technologies have enabled multi-well measurement of O(2) consumption, facilitating the rate of mitochondrial research, particularly regarding the mechanism of action of drugs and proteins that modulate metabolism. Among these technologies, the Seahorse XF24 Analyzer was designed for use with intact cells attached in a monolayer to a multi-well tissue culture plate. In order to have a high throughput assay system in which both energy demand and substrate availability can be tightly controlled, we have developed a protocol to expand the application of the XF24 Analyzer to include isolated mitochondria. Acquisition of optimal rates requires assay conditions that are unexpectedly distinct from those of conventional polarography. The optimized conditions, derived from experiments with isolated mouse liver mitochondria, allow multi-well assessment of rates of respiration and proton production by mitochondria attached to the bottom of the XF assay plate, and require extremely small quantities of material (1-10 µg of mitochondrial protein per well). Sequential measurement of basal, State 3, State 4, and uncoupler-stimulated respiration can be made in each well through additions of reagents from the injection ports. We describe optimization and validation of this technique using isolated mouse liver and rat heart mitochondria, and apply the approach to discover that inclusion of phosphatase inhibitors in the preparation of the heart mitochondria results in a specific decrease in rates of Complex I-dependent respiration. We believe this new technique will be particularly useful for drug screening and for generating previously unobtainable respiratory data on small mitochondrial samples.


Subject(s)
Cytological Techniques/methods , Mitochondria/metabolism , Animals , Cell Respiration , Female , Male , Mice , Oxygen Consumption , Rats , Reproducibility of Results , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...