Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
PLoS One ; 11(8): e0161413, 2016.
Article in English | MEDLINE | ID: mdl-27537184

ABSTRACT

Methyltriphenylphosphonium (TPMP) salts have been widely used to measure the mitochondrial membrane potential and the triphenylphosphonium (TPP+) moiety has been attached to many bioactive compounds including antioxidants to target them into mitochondria thanks to their high affinity to accumulate in the mitochondrial matrix. The adverse effects of these compounds on cellular metabolism have been insufficiently studied and are still poorly understood. Micromolar concentrations of TPMP cause a progressive inhibition of cellular respiration in adherent cells without a marked effect on mitochondrial coupling. In permeabilized cells the inhibition was limited to NADH-linked respiration. We found a mixed inhibition of the Krebs cycle enzyme 2-oxoglutarate dehydrogenase complex (OGDHC) with an estimated IC50 3.93 [3.70-4.17] mM, which is pharmacologically plausible since it corresponds to micromolar extracellular concentrations. Increasing the lipophilic character of the used TPP+ compound further potentiates the inhibition of OGDHC activity. This effect of TPMP on the Krebs cycle ought to be taken into account when interpreting observations on cells and mitochondria in the presence of TPP+ derivatives. Compounds based on or similar to TPP+ derivatives may also be used to alter OGDHC activity for experimental or therapeutic purposes.


Subject(s)
Citric Acid Cycle/drug effects , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Membrane Potential, Mitochondrial/drug effects , Onium Compounds/pharmacology , Trityl Compounds/pharmacology , Animals , Cell Line , Citrate (si)-Synthase/drug effects , Citrate (si)-Synthase/metabolism , Glutamate Dehydrogenase/drug effects , Glutamate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/drug effects , Isocitrate Dehydrogenase/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Malate Dehydrogenase/drug effects , Malate Dehydrogenase/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/enzymology , Muscle, Skeletal/enzymology , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Wistar
2.
J Neurosci Res ; 94(8): 749-58, 2016 08.
Article in English | MEDLINE | ID: mdl-27027410

ABSTRACT

Pyruvate dehydrogenase complex (PDH) is a brain mitochondrial matrix enzyme. PDH impairment after stroke is particularly devastating given PDH's critical role in the link between anaerobic and aerobic metabolism. This study evaluates the restoration of oxidative metabolism and energy regulation with a therapeutic combination of normobaric oxygen (NBO) plus either therapeutic hypothermia (TH) or ethanol. Sprague-Dawley rats were subjected to middle cerebral artery occlusion with an autologous embolus. One hour after occlusion, tissue-type plasminogen activator (t-PA) was administered alone or with NBO (60%), EtOH (1.0 g/kg), or TH (33°C), either singly or in combination. Neurological deficit score and infarct volume were assessed 24 hr after t-PA-induced reperfusion. PDH activity and reactive oxygen species (ROS) levels were measured 3 and 24 hr after t-PA. Western blotting was used to detect PDH and pyruvate dehydrogenase kinase (PDK) protein expression. After t-PA in ischemic rats, NBO combined with TH or EtOH most effectively decreased infarct volume and neurological deficit. The combined therapies produced greater increases in PDH activity and protein expression as well as greater decreases in PDK expression. Compared with the monotherapeutic approaches, the combined therapies provided the most significant declines in ROS generation. Reperfusion with t-PA followed by 60% NBO improves the efficacy of EtOH or TH in neuroprotection by ameliorating oxidative injury and improving PDH regulation. Comparable neuroprotective effects were found when treating with either EtOH or TH, suggesting a similar mechanism of neuroprotection and the possibility of substituting EtOH for TH in clinical settings. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain Ischemia/therapy , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hypothermia, Induced/methods , Neuroprotection , Oxygen Inhalation Therapy/methods , Pyruvate Dehydrogenase Complex/metabolism , Thromboembolism/therapy , Animals , Brain Ischemia/enzymology , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Combined Modality Therapy , Male , Pyruvate Dehydrogenase Complex/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Thromboembolism/enzymology , Thrombolytic Therapy
3.
Am J Physiol Heart Circ Physiol ; 310(6): H667-80, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26747502

ABSTRACT

Mitochondrial dysfunction has been implicated as a cause of energy deprivation in heart failure (HF). Herein, we tested individual and combined effects of two pathogenic factors of nonischemic HF, inhibition of nitric oxide synthesis [with l-N(G)-nitroarginine methyl ester (l-NAME)] and hypertension [with angiotensin II (AngII)], on myocardial mitochondrial function, oxidative stress, and metabolic gene expression. l-NAME and AngII were administered individually and in combination to mice for 5 wk. Although all treatments increased blood pressure and reduced cardiac contractile function, the l-NAME + AngII group was associated with the most severe HF, as characterized by edema, hypertrophy, oxidative stress, increased expression of Nppa and Nppb, and decreased expression of Atp2a2 and Camk2b. l-NAME + AngII-treated mice exhibited robust deterioration of cardiac mitochondrial function, as observed by reduced respiratory control ratios in subsarcolemmal mitochondria and reduced state 3 levels in interfibrillar mitochondria for complex I but not for complex II substrates. Cardiac myofibrils showed reduced ADP-supported and oligomycin-inhibited oxygen consumption. Mitochondrial functional impairment was accompanied by reduced mitochondrial DNA content and activities of pyruvate dehydrogenase and complex I but increased H2O2 production and tissue protein carbonyls in hearts from AngII and l-NAME + AngII groups. Microarray analyses revealed the majority of the gene changes attributed to the l-NAME + AngII group. Pathway analyses indicated significant changes in metabolic pathways, such as oxidative phosphorylation, mitochondrial function, cardiac hypertrophy, and fatty acid metabolism in l-NAME + AngII hearts. We conclude that l-NAME + AngII is associated with impaired mitochondrial respiratory function and increased oxidative stress compared with either l-NAME or AngII alone, resulting in nonischemic HF.


Subject(s)
Angiotensin II/pharmacology , Enzyme Inhibitors/pharmacology , Heart Failure/etiology , Mitochondria, Heart/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Atrial Natriuretic Factor , Calcium-Calmodulin-Dependent Protein Kinase Type 2/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cardiomegaly , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Electron Transport Complex II/drug effects , Electron Transport Complex II/metabolism , Gene Expression/drug effects , Heart/drug effects , Hydrogen Peroxide/metabolism , Mice , Mitochondria, Heart/metabolism , Myocardium/metabolism , Myocardium/pathology , Natriuretic Peptide, Brain/drug effects , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, C-Type/drug effects , Natriuretic Peptide, C-Type/genetics , Protein Precursors/drug effects , Protein Precursors/genetics , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
4.
Am J Physiol Heart Circ Physiol ; 304(8): H1103-13, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23396452

ABSTRACT

The renin-angiotensin system (RAS) may alter cardiac energy metabolism in heart failure. Angiotensin II (ANG II), the main effector of the RAS in heart failure, has emerged as an important regulator of cardiac hypertrophy and energy metabolism. We studied the metabolic perturbations and insulin response in an ANG II-induced hypertrophy model. Ex vivo heart perfusion showed that hearts from ANG II-treated mice had a lower response to insulin with significantly reduced rates of glucose oxidation in association with increased pyruvate dehydrogenase kinase 4 (PDK4) levels. Palmitate oxidation rates were significantly reduced in response to insulin in vehicle-treated hearts but remained unaltered in ANG II-treated hearts. Furthermore, phosphorylation of Akt was also less response to insulin in ANG II-treated wild-type (WT) mice, suggestive of insulin resistance. We evaluated the role of PDK4 in the ANG II-induced pathology and showed that deletion of PDK4 prevented ANG II-induced diastolic dysfunction and normalized glucose oxidation to basal levels. ANG II-induced reduction in the levels of the deacetylase, SIRT3, was associated with increased acetylation of pyruvate dehydrogenase (PDH) and a reduced PDH activity. In conclusion, our findings show that a combination of insulin resistance and decrease in PDH activity are involved in ANG II-induced reduction in glucose oxidation, resulting in cardiac inefficiency. ANG II reduces PDH activity via acetylation of PDH complex, as well as increased phosphorylation in response to increased PDK4 levels.


Subject(s)
Angiotensin II/pharmacology , Cardiomegaly/metabolism , Energy Metabolism/drug effects , Heart Failure, Diastolic/metabolism , Insulin Resistance/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/diagnostic imaging , Disease Models, Animal , Echocardiography , Energy Metabolism/physiology , Fatty Acids/metabolism , Glucose/metabolism , Glucose Tolerance Test , Heart/drug effects , Heart/physiology , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Oxidation-Reduction , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Sirtuin 3/metabolism
5.
Alcohol Alcohol ; 46(3): 233-8, 2011.
Article in English | MEDLINE | ID: mdl-21349883

ABSTRACT

AIMS: To clarify the role of acetate in neurochemical mechanisms of the initial (inborn) tolerance to ethanol. METHODS: Rats with low and high inborn tolerance to hypnotic effect of ethanol were used. In the brain region homogenates (frontal and parietal cortex, hypothalamus, striatum, medulla oblongata) and brain cortex synaptosomes, the levels of acetate, acetyl-CoA, acetylcholine (AcH), the activity of pyruvate dehydrogenase (PDG) and acetyl-CoA synthetase were examined. RESULTS: It has been found that brain cortex of rats with high tolerance to hypnotic effect of ethanol have higher level of acetate and activity of acetyl-CoA synthetase, but lower level of acetyl-СCoA and activity of PDG. In brain cortex synaptosomes of tolerant rats, the pyruvate oxidation rate as well as the content of acetyl-CoA and AcH synthesis were lower when compared with intolerant animals. The addition of acetate into the medium significantly increased the AcH synthesis in synaptosomes of tolerant, but not of intolerant animals. Calcium ions stimulated the AcH release from synaptosomes twice as high in tolerant as in intolerant animals. Acetate eliminated the stimulating effect of calcium ions upon the release of AcH in synaptosomes of intolerant rats, but not in tolerant animals. As a result, the quantum release of AcH from synaptosomes in the presence of acetate was 6.5 times higher in tolerant when compared with intolerant rats. CONCLUSION: The brain cortex of rats with high inborn tolerance to hypnotic effect of ethanol can better utilize acetate for the acetyl-CoA and AcH synthesis, as well as being resistant to inhibitory effect of acetate to calcium-stimulated release of AcH. It indicates the metabolic and cholinergic mechanisms of the initial tolerance to ethanol.


Subject(s)
Acetates/metabolism , Adaptation, Physiological/genetics , Alcohol-Related Disorders/genetics , Central Nervous System Depressants/metabolism , Ethanol/metabolism , Synaptosomes/drug effects , Acetyl Coenzyme A/drug effects , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/physiology , Acetylcholine/analysis , Acetylcholine/genetics , Acetylcholine/physiology , Adaptation, Physiological/physiology , Alcohol-Related Disorders/metabolism , Animals , Brain/metabolism , Central Nervous System Depressants/pharmacology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Ethanol/pharmacology , Humans , Hypothalamus/metabolism , Male , Medulla Oblongata/metabolism , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/physiology , Rats , Rats, Wistar , Synaptosomes/enzymology
6.
FASEB J ; 24(2): 627-36, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19812372

ABSTRACT

The physiology of two metabolites of vitamin A is understood in substantial detail: retinaldehyde functions as the universal chromophore in the vertebrate and invertebrate eye; retinoic acid regulates a set of vertebrate transcription factors, the retinoic acid receptor superfamily. The third member of this retinoid triumvirate is retinol. While functioning as the precursor of retinaldehyde and retinoic acid, a growing body of evidence suggests a far more fundamental role for retinol in signal transduction. Here we show that retinol is essential for the metabolic fitness of mitochondria. When cells were deprived of retinol, respiration and ATP synthesis defaulted to basal levels. They recovered to significantly higher energy output as soon as retinol was restored to physiological concentration, without the need for metabolic conversion to other retinoids. Retinol emerged as an essential cofactor of protein kinase Cdelta (PKCdelta), without which this enzyme failed to be activated in mitochondria. Furthermore, retinol needed to physically bind PKCdelta, because mutation of the retinol binding site rendered PKCdelta unresponsive to Rol, while retaining responsiveness to phorbol ester. The PKCdelta/retinol complex signaled the pyruvate dehydrogenase complex for enhanced flux of pyruvate into the Krebs cycle. The baseline response was reduced in vitamin A-deficient lecithin:retinol acyl transferase-knockout mice, but this was corrected within 3 h by intraperitoneal injection of vitamin A; this suggests that vitamin A is physiologically important. These results illuminate a hitherto unsuspected role of vitamin A in mitochondrial bioenergetics of mammals, acting as a nutritional sensor. As such, retinol is of fundamental importance for energy homeostasis. The data provide a mechanistic explanation to the nearly 100-yr-old question of why vitamin A deficiency causes so many pathologies that are independent of retinoic acid action.


Subject(s)
Energy Metabolism/physiology , Mitochondria/metabolism , Protein Kinase C-delta/metabolism , Pyruvate Dehydrogenase Complex/physiology , Vitamin A/physiology , Animals , Homeostasis/drug effects , Homeostasis/physiology , Humans , Jurkat Cells , Male , Mice , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/drug effects , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pyruvate Dehydrogenase Complex/drug effects , Retinoids/pharmacology , Signal Transduction , Vitamin A Deficiency/metabolism
7.
Neurochem Int ; 56(1): 143-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19781588

ABSTRACT

Excessive accumulation of zinc in the brain is one of putative factors involved in pathomechanism of cholinergic encephalopathies. The aim of this work was to investigate whether short-term increase of zinc concentration in the extracellular space may affect energy and acetylcholine metabolism in SN56 cholinergic cells of septal origin. Short 30 min exposition of SN56 cells to increasing zinc levels caused greater loss of viability of differentiated (DC, [EC(0.4)] 0.09 mM) than nondifferentiated cells (NC, [EC(0.4)] 0.14 mM). Concentration-dependent accumulation of zinc displayed exponential non-saturable kinetics. Zinc accumulation caused the decrease of calcium accumulation in mitochondria and its increase in cytoplasmic compartment of SN56 cells. Significant inverse and direct correlations were found between zinc accumulation and calcium levels in mitochondrial (r=-0.96, p=0.028) and cytoplasmic (r=0.97, p=0.028) compartments of DC, respectively. Zinc exerted similar inhibition of pyruvate dehydrogenase, aconitase and isocitrate dehydrogenase both in NC and DC homogenates, at Ki values equal to about 0.07, 0.08 and 0.005 mM, respectively. On the other hand, ketoglutarate dehydrogenase activity in DC was inhibited by zinc (Ki 0.0005 mM) 8 times stronger that in NC (Ki 0.004 mM). Also zinc-evoked decreases in acetylcholine content and its release were significantly greater in DC than in NC. Same conditions caused suppression of cytoplasmic and mitochondrial content of acetyl-CoA, that positively correlated with inhibition of transmitter functions (r=0.995, p=005) and loss of cell viability (r=0.990, p=0.0006), respectively. Significant correlations were also found in zinc-challenged cells between pyruvate dehydrogenase activity and both mitochondrial acetyl-CoA content and cell viability. These data indicate that pyruvate dehydrogenase-dependent acetyl-CoA synthesis in neuronal mitochondria may be a primary target for short-term neurotoxic effects of zinc. In consequence, shortages of acetyl-CoA in the mitochondrial compartment would cause fast loss of functional and structural integrity of cholinergic neurons.


Subject(s)
Acetyl Coenzyme A/biosynthesis , Acetylcholine/metabolism , Cholinergic Fibers/drug effects , Energy Metabolism/drug effects , Neurons/drug effects , Zinc/toxicity , Aconitate Hydratase/drug effects , Aconitate Hydratase/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cholinergic Fibers/metabolism , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Extracellular Space/drug effects , Extracellular Space/physiology , Isocitrate Dehydrogenase/drug effects , Isocitrate Dehydrogenase/metabolism , Kinetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/metabolism , Neurotoxins/toxicity , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Time Factors
8.
J Trauma ; 67(3): 628-33, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19741411

ABSTRACT

BACKGROUND: The pyruvate dehydrogenase complex (PDC) catalyzes the conversion of pyruvate to acetyl CoA, effectively controlling the entrance of glycolysis products into aerobic metabolism. Because hyperlactatemia is one of the hallmarks of sepsis, we hyphothesized that gram-positive and negative bacterial toxin treatment will interfere with mRNA levels of regulatory enzymes of the PDC and overall enzyme activity in hepatocytes. METHODS: HEP G2 hepatocarcinoma cells were incubated for 24 hours in the presence of lipopolysaccaride (LPS) or lipoteichoic acid. Total RNA was then isolated and message RNA levels for both pyruvate dehydrogense kinase 4 and phosphatase 2 were determined by RTPCR. Amplified DNA fragments were visualized by ethidium bromide in agarose gels and densitometry of the bands was performed. Data were then normalized to the housekeeping gene, GAPDH. Enzyme activity was then determined by capturing intact PDC on nitrocellulose membranes then determining PDC-dependent production of NADH. RESULTS: LPS treatment led to a time dependent increase in PDK4 message while decreasing PDP2 levels. Enzyme activity, in these cells, also significantly decreased 24 hours after exposure to LPS. Cells cultured in the presence of lipoteichoic acid and insulin exhibited differing message ratios and activity levels when evaluated at 4 hours, but at 24 hours shifted to mimic those observed in LPS treated cells. CONCLUSION: This data may indicate that exposure to bacterial cell wall components and insulin could create cellular environments that result in a build-up of lactate.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/enzymology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Lipopolysaccharides/pharmacology , Pyruvate Dehydrogenase Complex/drug effects , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Culture Techniques , Cell Line, Tumor , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Protein Kinases/genetics , Protein Kinases/metabolism , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/genetics , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/metabolism , RNA, Messenger/metabolism
9.
Am J Respir Crit Care Med ; 177(10): 1090-4, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18263802

ABSTRACT

RATIONALE: Impaired skeletal muscle function contributes to exercise limitation in patients with chronic obstructive pulmonary disease (COPD). This is characterized by reduced mitochondrial adenosine triphosphate generation, and greater reliance on nonmitochondrial energy production. Dichloroacetate (DCA) infusion activates muscle pyruvate dehydrogenase complex (PDC) at rest, reducing inertia in mitochondrial energy delivery at the onset of exercise and diminishing anaerobic energy production. OBJECTIVES: This study aimed to determine whether DCA infusion enhanced mitochondrial energy delivery during symptom-limited maximal exercise, thereby reducing exercise-induced lactate and ammonia accumulation and, consequently, improving exercise performance in patients with COPD. METHODS: A randomized, double-blind crossover design was used. Eighteen subjects with COPD performed maximal cycle exercise after an intravenous infusion of DCA (50 mg/kg body mass) or saline (control). Exercise work output was determined, and blood lactate and ammonia concentrations were measured at rest, 1 and 2 minutes of exercise, peak exercise, and 2 minutes postexercise. MEASUREMENTS AND MAIN RESULTS: DCA infusion reduced peak blood lactate concentration by 20% (mean [SE]; difference, 0.48 [0.11] mmol/L, P < 0.001) and peak blood ammonia concentration by 15% (mean [SE]; difference, 14.2 [2.9] mumol/L, P < 0.001] compared with control. After DCA, peak exercise workload improved significantly by a mean (SE) of 8 (1) W (P < 0.001) and peak oxygen consumption by 1.2 (0.5) ml/kg/minute (P = 0.03) compared with control. CONCLUSIONS: We have shown that a pharmacologic intervention known to activate muscle PDC can reduce blood lactate and ammonia accumulation during exercise and improve maximal exercise performance in subjects with COPD. Skeletal muscle PDC activation may be a target for pharmacologic intervention in the management of exercise intolerance in COPD.


Subject(s)
Dichloroacetic Acid/pharmacology , Enzyme Activators/pharmacology , Exercise Tolerance/drug effects , Muscle, Skeletal/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pyruvate Dehydrogenase Complex/drug effects , Aged , Aged, 80 and over , Ammonia/metabolism , Cross-Over Studies , Double-Blind Method , Exercise Test , Exercise Tolerance/physiology , Female , Humans , Lactic Acid/metabolism , Male , Middle Aged , Muscle, Skeletal/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology
10.
J Neurochem ; 103(3): 972-83, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17662047

ABSTRACT

Zinc is a trace element necessary for proper development and function of brain cells. However, excessive accumulation of zinc exerts several cytotoxic effects in the brain. The aim of this work was to see whether cytotoxic effects of zinc are quantitatively correlated with changes in acetyl-CoA metabolism. The zinc levels up to 0.20 mmol/L caused concentration-dependent inhibition of pyruvate dehydrogenase (PDH) activity that correlated with the increase in trypan blue-positive fraction and the decrease in cultured cell number (r = 0.96, p = 0.0001). Chronic exposure of cells to 0.15 mmol/L zinc decreased choline acetyltransferase and aconitase activities, cytoplasmic acetyl-CoA and whole cell ATP level by 38%, 57%, 35%, and 62%, respectively but caused no change in mitochondrial acetyl-CoA level and activities of other enzymes of glycolytic and tricarboxylic acid cycle. dl-alpha-lipoamide when added simultaneously with zinc to cultured cells or their homogenates attenuated its chronic or acute suppressive effects. In homogenates of chronically Zn-treated cells, lipoamide overcame PDH but not aconitase inhibition. Presented data indicate that acute-transient elevation of zinc caused reversible inhibition of PDH, aconitase activities and acetyl-CoA metabolism, which when prolonged could lead to irreversible enzyme inactivation yielding decrease in cell viability and secondary suppression of their cholinergic phenotype.


Subject(s)
Acetyl Coenzyme A/drug effects , Acetylcholine/biosynthesis , Energy Metabolism/drug effects , Nerve Degeneration/chemically induced , Neurons/drug effects , Zinc/toxicity , Acetyl Coenzyme A/metabolism , Aconitate Hydratase/antagonists & inhibitors , Aconitate Hydratase/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Choline O-Acetyltransferase/antagonists & inhibitors , Choline O-Acetyltransferase/metabolism , Citric Acid Cycle/drug effects , Citric Acid Cycle/physiology , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/toxicity , Glycolysis/drug effects , Glycolysis/physiology , Mice , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neuroblastoma , Neurons/metabolism , Neurotoxins/toxicity , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Thioctic Acid/analogs & derivatives , Thioctic Acid/pharmacology
11.
Mol Genet Metab ; 89(1-2): 97-105, 2006.
Article in English | MEDLINE | ID: mdl-16765624

ABSTRACT

The pyruvate dehydrogenase complex (PDC) is integral to metabolism and energetics. Congenital PDC deficiency leads to lactic acidosis, neurological degeneration and early death. An investigational compound for such defects is dichloroacetate (DCA), which activates the PDC (inhibiting reversible phosphorylation of the E1alpha subunit) and decreases its turnover. Here, primary human fibroblast cultures from five healthy subjects and six patients with mutations in the PDC-E1 component were grown in media+/-DCA, exposed to media containing (13)C-labeled glucose, and studied (as cell extracts) by nuclear magnetic resonance (NMR) spectroscopy. Computer modeling of NMR-derived (13)C-glutamate isotopomeric patterns estimated relative carbon flow through TCA cycle-associated pathways and characterized effects of PDC deficiency on metabolism and energetics. Rates of glucose consumption (GCR) and lactate production (LPR) were measured. With the exception of one patient cell line expressing an unusual splicing mutation, PDC-deficient cells had significantly higher GCR, LPR and label-derived acetyl-CoA, indicative of increased glycolysis vs. controls. In all cells, DCA caused a major shift (40% decrease) from anaplerotic-related pathways (e.g., pyruvate carboxylase) toward flux through PDC. Ignoring the patient with the splicing mutation, DCA decreased average glycolysis (29%) in patient cells, but had no significant effect on control cells, and did not change LPR or the nucleoside triphosphate to diphosphate ratio (NTP/NDP) in either cell type. Maintenance of NTP despite reduced glycolysis indicates that DCA improves metabolic efficiency by increasing glucose oxidation. This study demonstrates that NMR spectroscopy provides insight into biochemical consequences of PDC deficiency and the mechanism of putative therapeutic agents.


Subject(s)
Glucose/metabolism , Magnetic Resonance Spectroscopy/methods , Mitochondria/metabolism , Pyruvate Dehydrogenase Complex Deficiency Disease/metabolism , Pyruvate Dehydrogenase Complex/analysis , Cells, Cultured , Dichloroacetic Acid/pharmacology , Energy Metabolism , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Infant , Male , Mitochondria/enzymology , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex Deficiency Disease/enzymology
12.
Can J Microbiol ; 51(10): 833-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16333342

ABSTRACT

Oxythiamine is an antivitamin derivative of thiamine that after phosphorylation to oxythiamine pyro phosphate can bind to the active centres of thiamine-dependent enzymes. In the present study, the effect of oxythiamine on the viability of Saccharomyces cerevisiae and the activity of thiamine pyrophosphate dependent enzymes in yeast cells has been investigated. We observed a decrease in pyruvate decarboxylase specific activity on both a control and an oxythiamine medium after the first 6 h of culture. The cytosolic enzymes transketolase and pyruvate decarboxylase decreased their specific activity in the presence of oxythiamine but only during the beginning of the cultivation. However, after 12 h of cultivation, oxythiamine-treated cells showed higher specific activity of cytosolic enzymes. More over, it was established by SDS-PAGE that the high specific activity of pyruvate decarboxylase was followed by an increase in the amount of the enzyme protein. In contrast, the mitochondrial enzymes, pyruvate dehydrogenase and 2-oxoglutarate dehydrogenase complexes, were inhibited by oxythiamine during the entire experiment. Our results suggest that the observed strong decrease in growth rate and viability of yeast on medium with oxythiamine may be due to stronger inhibition of mitochondrial pyruvate dehydrogenase than of cytosolic enzymes.


Subject(s)
Antimetabolites/pharmacology , Oxythiamine/pharmacology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/growth & development , Thiamine Pyrophosphate/metabolism , Colony Count, Microbial , Culture Media , Cytosol/enzymology , Ketoglutarate Dehydrogenase Complex/drug effects , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/enzymology , Pyruvate Decarboxylase/drug effects , Pyruvate Decarboxylase/metabolism , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Saccharomyces cerevisiae/drug effects , Transketolase/drug effects , Transketolase/metabolism
13.
J Neurochem ; 90(4): 952-61, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15287901

ABSTRACT

Nerve growth factor (NGF) is a peptide displaying multiple cholinotropic activities. The aim of this work was to explain mechanisms of the positive and negative effects of NGF on phenotypic properties and viability of cholinergic cells. To discriminate these effects we used two p75NTR receptor-positive lines of cholinergic neuroblastoma cells, SN56 and T17 that are devoid of or express high affinity NGF (TrkA) receptors, respectively. cAMP and retinoic acid caused differentiation of both cell lines. In addition to the morphologic maturation, the increase of choline acetyltransferase activity, acetylcholine, Ca and cytoplasmic acetyl-CoA levels and decrease of mitochondrial acetyl-CoA and cell viability were observed. NGF caused similar effects in non-differentiated T17 cells but had no influence on non-differentiated SN56 cells. On the contrary, in both cAMP/all-trans-retinoic acid (RA) differentiated cell lines, NGF resulted in a similar suppression of cholinergic phenotype along with an increase of mitochondrial acetyl-CoA and cell susceptibility to nitric oxide and amyloid-beta25-35. These effects of NGF were prevented by an antibody against the p75NTR receptor. Data indicate that: (i) positive cholinotrophic effects of NGF required activation of both TrkA and p75NTR receptors; (ii) cAMP/RA-evoked differentiation inhibited NGF effects mediated by TrkA receptors and activated its p75NTR-dependent suppressing influences and (iii) a differentiation-evoked decrease of mitochondrial acetyl-CoA and an elevation of mitochondrial Ca could augment impairment of cholinergic neurons by neurotoxic signals.


Subject(s)
Acetyl Coenzyme A/metabolism , Acetylcholine/metabolism , Choline O-Acetyltransferase/metabolism , Nerve Growth Factor/pharmacology , Neuroblastoma/metabolism , Neurons/drug effects , Neurons/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Antibodies/pharmacology , Bucladesine/pharmacology , Calcium/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Choline O-Acetyltransferase/drug effects , Cytoplasm/drug effects , Cytoplasm/enzymology , Enzyme Activation/drug effects , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Nitroprusside/pharmacology , Peptide Fragments/pharmacology , Pyruvate Dehydrogenase Complex/drug effects , Rats , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Tretinoin/pharmacology
14.
J Neurochem ; 88(6): 1352-60, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15009635

ABSTRACT

Altered energy metabolism, including reductions in activities of the key mitochondrial enzymes alpha-ketoglutarate dehydrogenase complex (KGDHC) and pyruvate dehydrogenase complex (PDHC), are characteristic of many neurodegenerative disorders including Alzheimer's Disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Dihydrolipoamide dehydrogenase is a critical subunit of KGDHC and PDHC. We tested whether mice that are deficient in dihydrolipoamide dehydrogenase (Dld+/-) show increased vulnerability to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), malonate and 3-nitropropionic acid (3-NP), which have been proposed for use in models of PD and HD. Administration of MPTP resulted in significantly greater depletion of tyrosine hydroxylase-positive neurons in the substantia nigra of Dld+/- mice than that seen in wild-type littermate controls. Striatal lesion volumes produced by malonate and 3-NP were significantly increased in Dld+/- mice. Studies of isolated brain mitochondria treated with 3-NP showed that both succinate-supported respiration and membrane potential were suppressed to a greater extent in Dld+/- mice. KGDHC activity was also found to be reduced in putamen from patients with HD. These findings provide further evidence that mitochondrial defects may contribute to the pathogenesis of neurodegenerative diseases.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Dihydrolipoamide Dehydrogenase/deficiency , Genetic Predisposition to Disease , Malonates , Neurodegenerative Diseases/enzymology , Propionates , Animals , Caudate Nucleus/drug effects , Caudate Nucleus/enzymology , Caudate Nucleus/pathology , Cell Count , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/pathology , Dihydrolipoamide Dehydrogenase/genetics , Disease Models, Animal , Heterozygote , Huntington Disease/chemically induced , Huntington Disease/enzymology , Huntington Disease/pathology , Ketoglutarate Dehydrogenase Complex/drug effects , Ketoglutarate Dehydrogenase Complex/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Nitro Compounds , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/pathology , Putamen/drug effects , Putamen/enzymology , Putamen/pathology , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Substantia Nigra/drug effects , Substantia Nigra/enzymology , Substantia Nigra/pathology
15.
Hum Mol Genet ; 13(3): 303-14, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14662656

ABSTRACT

Mitochondrial dysfunction, with an estimated incidence of 1 in 10 000 live births, is among the most common genetically determined conditions. Missense mutations in the human NDUFV1 gene, which encodes the 51 kDa active site subunit of the NADH-ubiquinone oxidoreductase or complex I, can lead to severe neurological disorders. Owing to the rare and often sporadic nature of mitochondrial disorders, the mechanisms of pathogenesis of most mutations remain poorly understood. We have generated transgenic strains of Caenorhabditis elegans that express disease-causing mutations in the nuo-1 gene, the C. elegans homolog of the NDUFV1 gene. The transgenic strains demonstrate hallmark features of complex I dysfunction such as lactic acidosis and decreased NADH-dependent mitochondrial respiration. They are also hypersensitive to exogenous oxidative stress, suggesting that cellular defense mechanisms against reactive oxygen species are already taxed by an endogenous stress. The lactic acidosis induced by the NDUFV1 mutations could be partially corrected with the vitamins riboflavin and thiamine or with sodium dichloroacetate, an activator of the pyruvate dehydrogenase complex, resulting in significant increases in animal fitness. Surprisingly, cytochrome c oxidase activity and protein levels were reduced, establishing a connection between complexes I and IV. Our results indicate that complex I mutations exert their pathogenic effects in multiple ways: by impeding the metabolism of NADH, by increasing the production of reactive oxygen species, and by interfering with the function or assembly of other mitochondrial respiratory chain components.


Subject(s)
Acidosis, Lactic/genetics , Electron Transport Complex IV/metabolism , Electron Transport Complex I/genetics , Oxidative Stress/genetics , Acidosis, Lactic/metabolism , Aging/genetics , Aging/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins , Electron Transport Complex I/metabolism , Enzyme Activators/pharmacology , Oxidative Stress/physiology , Pyruvate Dehydrogenase Complex/drug effects , Vitamins/pharmacology
16.
Ann Nutr Metab ; 46(3-4): 114-20, 2002.
Article in English | MEDLINE | ID: mdl-12169854

ABSTRACT

The aim of this work was to study the effect of the administration of cod liver oil on the non-oxidative and oxidative fate of glucose metabolism in the skeletal muscle of normal rats. To achieve this goal, the gastrocnemius was examined regarding glucose oxidation, glycogen synthase activity and glycogen storage both at baseline and during euglycemic hyperinsulinemic clamping. The results show that dietary fish oil decreases plasma insulin levels without alteration in glucose homeostasis (at baseline). In addition, the observed enhancement in whole body glucose utilization during clamping suggests an increased peripheral insulin sensitivity. Furthermore, under insulin-stimulated glucose disposal, an enhancement in the glycolytic pathway (increased levels of muscle glucose-6-phosphate and plasma lactate) rather than changes in the oxidation (pyruvate dehydrogenase complex) and storage components of glucose metabolism was observed in the skeletal muscle of rats fed dietary fish oil. These results coupled with the hypolipidemic effects of fish oil may have implications for the prevention and/or management of some pathological states manifested by insulin resistance with or without dyslipidemia.


Subject(s)
Dietary Fats, Unsaturated/pharmacology , Fish Oils/pharmacology , Glucose/metabolism , Insulin/metabolism , Muscle, Skeletal/physiology , Animals , Cod Liver Oil/pharmacology , Corn Oil/pharmacology , Fatty Acids/administration & dosage , Glucose Clamp Technique , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Synthase/drug effects , Male , Protein Kinases/drug effects , Protein Serine-Threonine Kinases , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/drug effects , Rats , Rats, Wistar , Reference Values , Triglycerides/metabolism
17.
J Neurosci Res ; 66(5): 803-11, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11746405

ABSTRACT

Nuclear magnetic resonance was used as the primary technique to investigate the effect of ethanol (40, 80, and 160 mM) on the levels of high-energy phosphates, glycolytic flux, anaplerotic and oxidative fluxes to the tricarboxylic acid (TCA) cycle, the contribution of the pentose phosphate pathway (PPP), and the uptake and release of amino acids on primary cultures of rat astrocytes. On line (31)P-NMR spectroscopy showed that long-term exposure to ethanol caused a drop in the levels of ATP and phosphocreatine. The ratio between the fluxes through the pyruvate dehydrogenase and pyruvate carboxylase reactions also decreased, whereas the glycolytic flux and the ratio between formation of lactate and glucose consumption increased when cells were exposed to acute doses of ethanol. Flux through the pentose phosphate pathway was not affected. The uptake of cysteine and the release of glutamine were stimulated by ethanol, whereas the release of methionine was inhibited. Moreover, the fractional enrichment in serine was enhanced. The changes in the amino acid metabolism are interpreted as a response to oxidative stress induced by ethanol.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Astrocytes/drug effects , Brain/drug effects , Energy Metabolism/drug effects , Ethanol/pharmacokinetics , Adenine Nucleotides/metabolism , Alcohol-Induced Disorders, Nervous System/physiopathology , Amino Acids/drug effects , Amino Acids/metabolism , Animals , Animals, Newborn , Astrocytes/diagnostic imaging , Brain/diagnostic imaging , Carbon Radioisotopes/pharmacokinetics , Cells, Cultured , Citric Acid Cycle/drug effects , Citric Acid Cycle/physiology , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Ethanol/metabolism , Glycolysis/drug effects , Glycolysis/physiology , Magnetic Resonance Spectroscopy , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/physiology , Phosphorus Radioisotopes/pharmacokinetics , Pyruvate Carboxylase/drug effects , Pyruvate Carboxylase/metabolism , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Radionuclide Imaging , Rats , Rats, Wistar
18.
J Am Coll Cardiol ; 36(4): 1378-85, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11028498

ABSTRACT

OBJECTIVES: We sought to determine whether improving coupling between glucose oxidation and glycolysis by stimulating glucose oxidation during reperfusion enhances postischemic recovery of hypertrophied hearts. BACKGROUND: Low rates of glucose oxidation and high glycolytic rates are associated with greater postischemic dysfunction of hypertrophied as compared with nonhypertrophied hearts. METHODS: Heart function, glycolysis and glucose oxidation were measured in isolated working control and hypertrophied rat hearts for 30 min before 20 min of global, no-flow ischemia and during 60 min of reperfusion. Selected control and hypertrophied hearts received 1.0 mmol/liter dichloroacetate (DCA), an activator of pyruvate dehydrogenase, at the time of reperfusion to stimulate glucose oxidation. RESULTS: In the absence of DCA, glycolysis was higher and glucose oxidation and recovery of function were lower in hypertrophied hearts than in control hearts during reperfusion. Dichloroacetate stimulated glucose oxidation during reperfusion approximately twofold in both groups, while significantly reducing glycolysis in hypertrophied hearts. It also improved function of both hypertrophied and control hearts. In the presence of DCA, recovery of function of hypertrophied hearts was comparable to or better than that of untreated control hearts. CONCLUSIONS: Dichloroacetate, given at the time of reperfusion, normalizes postischemic function of hypertrophied rat hearts and improves coupling between glucose oxidation and glycolysis by increasing glucose oxidation and decreasing glycolysis. These findings support the hypothesis that low glucose oxidation rates and high glycolytic rates contribute to the exaggerated postischemic dysfunction of hypertrophied hearts.


Subject(s)
Cardiomegaly/physiopathology , Dichloroacetic Acid/therapeutic use , Glucose/metabolism , Glycolysis/drug effects , Myocardial Reperfusion Injury/drug therapy , Ventricular Function/physiology , Animals , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Disease Models, Animal , Glycogen/metabolism , Glycolysis/physiology , In Vitro Techniques , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Oxidation-Reduction/drug effects , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Function/drug effects
19.
J Med Genet ; 37(9): 669-73, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10978358

ABSTRACT

The thiamine transporter gene SLC19A2 was recently found to be mutated in thiamine responsive megaloblastic anaemia with diabetes and deafness (TRMA, Rogers syndrome), an early onset autosomal recessive disorder. We now report a novel G1074A transition mutation in exon 4 of the SLC19A2 gene, predicting a Trp358 to ter change, in a girl with consanguineous parents. In addition to the typical triad of Rogers syndrome, the girl presented with short stature, hepatosplenomegaly, retinal degeneration, and a brain MRI lesion. Both muscle and skin biopsies were obtained before high dose thiamine supplementation. While no mitochondrial abnormalities were seen on morphological examination of muscle, biochemical analysis showed a severe deficiency of pyruvate dehydrogenase and complex I of the respiratory chain. In the patient's fibroblasts, the supplementation with high doses of thiamine resulted in restoration of complex I activity. In conclusion, we provide evidence that thiamine deficiency affects complex I activity. The clinical features of TRMA, resembling in part those found in typical mitochondrial disorders with complex I deficiency, may be caused by a secondary defect in mitochondrial energy production.


Subject(s)
Anemia, Megaloblastic/genetics , Carrier Proteins/genetics , Membrane Transport Proteins , NADH, NADPH Oxidoreductases/deficiency , Thiamine/therapeutic use , Adolescent , Anemia, Megaloblastic/drug therapy , Base Sequence , Consanguinity , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Electron Transport Complex I , Family Health , Female , Humans , Male , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/enzymology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Mutation , NADH, NADPH Oxidoreductases/drug effects , Pedigree , Point Mutation , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex Deficiency Disease
20.
Chem Res Toxicol ; 13(8): 749-60, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10956063

ABSTRACT

The major initial product of the oxidation of norepinephrine (NE) in the presence of L-cysteine is 5-S-cysteinylnorepinephrine which is then further easily oxidized to the dihydrobenzothiazine (DHBT) 7-(1-hydroxy-2-aminoethyl)-3,4-dihydro-5-hydroxy-2H-1, 4-benzothiazine-3-carboxylic acid (DHBT-NE-1). When incubated with intact rat brain mitochondria, DHBT-NE-1 evokes rapid inhibition of complex I respiration without affecting complex II respiration. DHBT-NE-1 also evokes time- and concentration-dependent irreversible inhibition of NADH-coenzyme Q(1) (CoQ(1)) reductase, the pyruvate dehydrogenase complex (PDHC), and alpha-ketoglutarate dehydrogenase (alpha-KGDH) when incubated with frozen and thawed rat brain mitochondria (mitochondrial membranes). The time dependence of the inhibition of NADH-CoQ(1) reductase, PDHC, and alpha-KGDH by DHBT-NE-1 appears to be related to its oxidation, catalyzed by an unknown component of the inner mitochondrial membrane, to electrophilic intermediates which bind covalently to active site cysteinyl residues of these enzyme complexes. The latter conclusion is based on the ability of glutathione to block inhibition of NADH-CoQ(1) reductase, PDHC, and alpha-KGDH by scavenging electrophilic intermediates, generated by the mitochondrial membrane-catalyzed oxidation of DHBT-NE-1, forming glutathionyl conjugates, several of which have been isolated and spectroscopically identified. The possible implications of these results to the degeneration of neuromelanin-pigmented noradrenergic neurons in the locus ceruleus in Parkinson's disease are discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Multienzyme Complexes/drug effects , Norepinephrine/analogs & derivatives , Parkinson Disease/metabolism , Thiazines/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/metabolism , Chromatography, High Pressure Liquid , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/drug effects , Enzyme Inhibitors/chemical synthesis , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/drug effects , Male , Mitochondria/enzymology , Multienzyme Complexes/antagonists & inhibitors , Norepinephrine/metabolism , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL