Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 204: 287-300, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37225107

ABSTRACT

Pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH) are vital sources of hydrogen peroxide (H2O2) and key sites for redox regulation. Here, we report KGDH is more sensitive to inhibition by S-nitroso-glutathione (GSNO) when compared to PDH and deactivation of both enzymes by nitro modification is affected by sex and diet. Liver mitochondria from male C57BL/6 N mice displayed a robust inhibition of H2O2 production after exposure to 500-2000 µM GSNO. H2O2 genesis by PDH was not significantly affected by GSNO. Purified KGDH of porcine heart origin displayed a ∼82% decrease in H2O2 generating activity at 500 µM GSNO, which was mirrored by a decrease in NADH production. By contrast, H2O2- and NADH-producing activity of purified PDH was only minimally affected by an incubation in 500 µM GSNO. Incubations in GSNO had no significant effect on the H2O2-generating activity of KGDH and PDH in female liver mitochondria when compared to samples collected from males, which was attributed to higher GSNO reductase (GSNOR) activity. High fat feeding augmented the GSNO-mediated inhibition of KGDH in liver mitochondria from male mice. Exposure of male mice to a HFD also resulted in a significant decrease in the GSNO-mediated inhibition of H2O2 genesis by PDH, an effect not observed in mice fed a control-matched diet (CD). Female mice displayed higher resistance to the GSNO-induced inhibition of H2O2 production, regardless of being fed a CD or HFD. However, exposure to a HFD did result in a small but significant decrease in H2O2 production by KGDH and PDH when female liver mitochondria were treated with GSNO. Although, the effect was less when compared to their male counterparts. Collectively, we show for the first time GSNO deactivates H2O2 production by α-keto acid dehydrogenases and we demonstrate that sex and diet are determinants for the nitro-inhibition of both KGDH and PDH.


Subject(s)
Hydrogen Peroxide , Ketoglutarate Dehydrogenase Complex , Animals , Female , Male , Mice , Diet , Glutathione , Ketoglutarate Dehydrogenase Complex/physiology , Mice, Inbred C57BL , NAD , Peroxides
2.
Biochim Biophys Acta Gen Subj ; 1861(8): 1960-1969, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28506882

ABSTRACT

Pyruvate dehydrogenase (PDHC) and α-ketoglutarate dehydrogenase complex (KGDHC) are important sources of reactive oxygen species (ROS). In addition, it has been found that mitochondria can also serve as sinks for cellular hydrogen peroxide (H2O2). However, the ROS forming and quenching capacity of liver mitochondria has never been thoroughly examined. Here, we show that mouse liver mitochondria use catalase, glutathione (GSH), and peroxiredoxin (PRX) systems to quench ROS. Incubation of mitochondria with catalase inhibitor 3-amino-1,2,4-triazole (triazole) induced a significant increase in pyruvate or α-ketoglutarate driven O2-/H2O2 formation. 1-Choro-2,4-dinitrobenzene (CDNB), which depletes glutathione (GSH), elicited a similar effect. Auranofin (AF), a thioredoxin reductase-2 (TR2) inhibitor which disables the PRX system, did not significantly change O2-/H2O2 formation. By contrast catalase, GSH, and PRX were all required to scavenging extramitochondrial H2O2. In this study, the ROS forming potential of PDHC, KGDHC, Complex I, and Complex III was also profiled. Titration of mitochondria with 3-methyl-2-oxovaleric acid (KMV), a specific inhibitor for O2-/H2O2 production by KGDHC, induced a ~86% and ~84% decrease in ROS production during α-ketoglutarate and pyruvate oxidation. Titration of myxothiazol, a Complex III inhibitor, decreased O2-/H2O2 formation by ~45%. Rotenone also lowered ROS production in mitochondria metabolizing pyruvate or α-ketoglutarate indicating that Complex I does not contribute to ROS production during forward electron transfer from NADH. Taken together, our results indicate that KGDHC and Complex III are high capacity sites for O2-/H2O2 production in mouse liver mitochondria. We also confirm that catalase plays a role in quenching either exogenous or intramitochondrial H2O2.


Subject(s)
Hydrogen Peroxide/metabolism , Mitochondria, Liver/metabolism , Superoxides/metabolism , Animals , Caprylates/pharmacology , Catalase/physiology , Electron Transport Complex III/physiology , Glutathione/metabolism , Ketoglutarate Dehydrogenase Complex/physiology , Male , Methacrylates/pharmacology , Mice , Mice, Inbred C57BL , Peroxiredoxins/physiology , Reactive Oxygen Species/metabolism , Sulfides/pharmacology , Thiazoles/pharmacology
3.
Free Radic Biol Med ; 97: 501-512, 2016 08.
Article in English | MEDLINE | ID: mdl-27394173

ABSTRACT

Pyruvate dehydrogenase (Pdh) and 2-oxoglutarate dehydrogenase (Ogdh) are vital for Krebs cycle metabolism and sources of reactive oxygen species (ROS). O2(·-)/H2O2 formation by Pdh and Ogdh from porcine heart were compared when operating under forward or reverse electron transfer conditions. Comparisons were also conducted with liver and cardiac mitochondria. During reverse electron transfer (RET) from NADH, purified Ogdh generated ~3-3.5× more O2(·-)/H2O2 in comparison to Pdh when metabolizing 0.5-10µM NADH. Under forward electron transfer (FET) conditions Ogdh generated ~2-4× more O2(·-)/H2O2 than Pdh. In both liver and cardiac mitochondria, Ogdh displayed significantly higher rates of ROS formation when compared to Pdh. Ogdh was also a significant source of ROS in liver mitochondria metabolizing 50µM and 500µM pyruvate or succinate. Finally, we also observed that DTT directly stimulated O2(·-)/H2O2 formation by purified Pdh and Ogdh and in cardiac or liver mitochondria in the absence of substrates and cofactors. Taken together, Ogdh is a more potent source of ROS than Pdh in liver and cardiac tissue. Ogdh is also an important ROS generator regardless of whether pyruvate or succinate serve as the sole source of carbon. Our observations provide insight into the ROS generating capacity of either complex in cardiac and liver tissue. The evidence presented herein also indicates DTT, a reductant that is routinely added to biological samples, should be avoided when assessing mitochondrial O2(·-)/H2O2 production.


Subject(s)
Hydrogen Peroxide/metabolism , Ketoglutarate Dehydrogenase Complex/physiology , Pyruvate Dehydrogenase Complex/physiology , Superoxides/metabolism , Animals , Male , Mice, Inbred C57BL , Mitochondria, Heart/enzymology , Mitochondria, Liver/enzymology , Succinic Acid/metabolism
4.
Free Radic Biol Med ; 91: 247-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26708453

ABSTRACT

In humans, mutations in dehydrogenase E1 and transketolase domain containing 1 (DHTKD1) are associated with neurological abnormalities and accumulation of 2-oxoadipate, 2-aminoadipate, and reactive oxygen species. The protein encoded by DHTKD1 has sequence and structural similarities to 2-oxoglutarate dehydrogenase, and the 2-oxoglutarate dehydrogenase complex can produce superoxide/H2O2 at high rates. The DHTKD1 enzyme is hypothesized to catalyze the oxidative decarboxylation of 2-oxoadipate, a shared intermediate of the degradative pathways for tryptophan, lysine and hydroxylysine. Here, we show that rat skeletal muscle mitochondria can produce superoxide/H2O2 at high rates when given 2-oxoadipate. We identify the putative mitochondrial 2-oxoadipate dehydrogenase complex as one of the sources and characterize the conditions that favor its superoxide/H2O2 production. Rates increased at higher NAD(P)H/NAD(P)(+) ratios and were higher at each NAD(P)H/NAD(P)(+) ratio when 2-oxoadipate was present, showing that superoxide/H2O2 was produced during the forward reaction from 2-oxoadipate, but not in the reverse reaction from NADH in the absence of 2-oxoadipate. The maximum capacity of the 2-oxoadipate dehydrogenase complex for production of superoxide/H2O2 is comparable to that of site IF of complex I, and seven, four and almost two-fold lower than the capacities of the 2-oxoglutarate, pyruvate and branched-chain 2-oxoacid dehydrogenase complexes, respectively. Regulation by ADP and ATP of H2O2 production driven by 2-oxoadipate was very different from that driven by 2-oxoglutarate, suggesting that site AF of the 2-oxoadipate dehydrogenase complex is a new source of superoxide/H2O2 associated with the NADH isopotential pool in mitochondria.


Subject(s)
Adipates/metabolism , Hydrogen Peroxide/metabolism , Ketoglutarate Dehydrogenase Complex/physiology , Mitochondria, Muscle/enzymology , Superoxides/metabolism , Animals , Female , Kinetics , Muscle, Skeletal/enzymology , Oxidation-Reduction , Rats, Wistar
5.
Biochemistry (Mosc) ; 81(12): 1498-1521, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28259128

ABSTRACT

2-Oxo acid dehydrogenase complexes are important metabolic checkpoints functioning at the intercept of sugar and amino acid degradation. This review presents a short summary of architectural, catalytic, and regulatory principles of the complexes structure and function, based on recent advances in studies of well-characterized family members. Special attention is given to use of synthetic phosphonate and phosphinate analogs of 2-oxo acids as selective and efficient inhibitors of the cognate complexes in biological systems of bacterial, plant, and animal origin. We summarize our own results concerning the application of synthetic analogs of 2-oxo acids in situ and in vivo to reveal functional interactions between 2-oxo acid dehydrogenase complexes and other components of metabolic networks specific to different cells and tissues. Based on our study of glutamate excitotoxicity in cultured neurons, we show how a modulation of metabolism by specific inhibition of its key reaction may be employed to correct pathologies. This approach is further developed in our study on the action of the phosphonate analog of 2-oxoglutarate in animals. The study revealed that upregulation of 2-oxoglutarate dehydrogenase complex is involved in animal stress response and may provide increased resistance to damaging effects, underlying so-called preconditioning. The presented analysis of published data suggests synthetic inhibitors of metabolic checkpoints as promising tools to solve modern challenges of systems biology, metabolic engineering, and medicine.


Subject(s)
Enzyme Inhibitors/chemistry , Ketoglutarate Dehydrogenase Complex/chemistry , Ketoglutaric Acids/chemistry , Organophosphonates/chemistry , Phosphinic Acids/chemistry , Animals , Humans , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Ketoglutarate Dehydrogenase Complex/physiology , Kinetics , Mitochondria/enzymology
6.
Metab Brain Dis ; 29(4): 1061-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24929329

ABSTRACT

Thiamine deficiency (TD) is the underlying cause of Wernicke's encephalopathy (WE), an acute neurological disorder characterized by structural damage to key periventricular structures in the brain. Increasing evidence suggests these focal histological lesions may be representative of a gliopathy in which astrocyte-related changes are a major feature of the disorder. These changes include a loss of the glutamate transporters GLT-1 and GLAST concomitant with elevated interstitial glutamate levels, lowered brain pH associated with increased lactate production, decreased levels of GFAP, reduction in the levels of glutamine synthetase, swelling, alterations in levels of aquaporin-4, and disruption of the blood-brain barrier. This review focusses on how these manifestations contribute to the pathophysiology of TD and possibly WE.


Subject(s)
Astrocytes/physiology , Thiamine Deficiency/physiopathology , Amino Acid Transport System X-AG/physiology , Animals , Biological Transport , Blood-Brain Barrier , Brain/pathology , Disease Models, Animal , Excitatory Amino Acid Transporter 2/physiology , Glutamic Acid/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Pyrithiamine/toxicity , Thiamine Deficiency/chemically induced , Thiamine Deficiency/metabolism , Wernicke Encephalopathy/etiology , Wernicke Encephalopathy/metabolism , Wernicke Encephalopathy/physiopathology
7.
Eukaryot Cell ; 12(1): 78-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23125353

ABSTRACT

The dihydrolipoyl succinyltransferase (E2) of the multisubunit α-ketoglutarate dehydrogenase complex (α-KD) is an essential Krebs cycle enzyme commonly found in the matrices of mitochondria. African trypanosomes developmentally regulate mitochondrial carbohydrate metabolism and lack a functional Krebs cycle in the bloodstream of mammals. We found that despite the absence of a functional α-KD, bloodstream form (BF) trypanosomes express α-KDE2, which localized to the mitochondrial matrix and inner membrane. Furthermore, α-KDE2 fractionated with the mitochondrial genome, the kinetoplast DNA (kDNA), in a complex with the flagellum. A role for α-KDE2 in kDNA maintenance was revealed in α-KDE2 RNA interference (RNAi) knockdowns. Following RNAi induction, bloodstream trypanosomes showed pronounced growth reduction and often failed to equally distribute kDNA to daughter cells, resulting in accumulation of cells devoid of kDNA (dyskinetoplastic) or containing two kinetoplasts. Dyskinetoplastic trypanosomes lacked mitochondrial membrane potential and contained mitochondria of substantially reduced volume. These results indicate that α-KDE2 is bifunctional, both as a metabolic enzyme and as a mitochondrial inheritance factor necessary for the distribution of kDNA networks to daughter cells at cytokinesis.


Subject(s)
Citric Acid Cycle , DNA, Kinetoplast/genetics , Ketoglutarate Dehydrogenase Complex/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/enzymology , Cells, Cultured , Cytokinesis , DNA Replication , Enzyme Stability , Flagella/metabolism , Gene Expression , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/physiology , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Mitochondria/genetics , Protein Binding , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/physiology , RNA Interference , Trypanosoma brucei brucei/cytology , Trypanosoma brucei brucei/growth & development
8.
FEMS Microbiol Lett ; 303(1): 33-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20041955

ABSTRACT

Persisters are a small population of slowly growing or nongrowing bacteria that are phenotypically resistant to antibiotics, but the mechanisms involved are not well understood. The aim of this study is to determine new mechanisms underlying antibiotic-tolerant persisters. The Escherichia coli deletion mutant library was screened to identify mutants that had a defect in persister survival after exposure to ampicillin for 24 h or 5 days. The identified mutants and the parent strain were subjected to minimum inhibitory concentration (MIC) and minimum bactericidal tests and antibiotic or stress conditions in exposure assays. sucB and ubiF mutants deficient in energy production were identified from the mutant screens to have defective persister survival as demonstrated by higher susceptibility to various antibiotics, including ampicillin, norfloxacin, tetracycline and gentamicin, and different stresses such as oxidative stress, acid pH and weak acid compared with the parent strain. In addition, both sucB and ubiF had a twofold lower MIC than the parent strain. The above sucB and ubiF mutant phenotypes could be complemented by their respective functional genes. Defective energy production through mutations in sucB and ubiF affects persister survival and could serve as new drug targets for persister bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/physiology , Escherichia coli/physiology , Ketoglutarate Dehydrogenase Complex/physiology , Microbial Viability , Mixed Function Oxygenases/physiology , Stress, Physiological , Acids/pharmacology , Ampicillin/pharmacology , Escherichia coli/drug effects , Escherichia coli Proteins/genetics , Gene Deletion , Genetic Complementation Test , Ketoglutarate Dehydrogenase Complex/genetics , Microbial Sensitivity Tests , Mixed Function Oxygenases/genetics , Oxidative Stress
9.
PLoS Pathog ; 5(11): e1000662, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19936047

ABSTRACT

Aerobic organisms have a tricarboxylic acid (TCA) cycle that is functionally distinct from those found in anaerobic organisms. Previous reports indicate that the aerobic pathogen Mycobacterium tuberculosis lacks detectable alpha-ketoglutarate (KG) dehydrogenase activity and drives a variant TCA cycle in which succinyl-CoA is replaced by succinic semialdehyde. Here, we show that M. tuberculosis expresses a CoA-dependent KG dehydrogenase activity, albeit one that is typically found in anaerobic bacteria. Unlike most enzymes of this family, the M. tuberculosis KG: ferredoxin oxidoreductase (KOR) is extremely stable under aerobic conditions. This activity is absent in a mutant strain deleted for genes encoding a previously uncharacterized oxidoreductase, and this strain is impaired for aerobic growth in the absence of sufficient amounts of CO(2). Interestingly, inhibition of the glyoxylate shunt or exclusion of exogenous fatty acids alleviates this growth defect, indicating the presence of an alternate pathway that operates in the absence of beta-oxidation. Simultaneous disruption of KOR and the first enzyme of the succinic semialdehyde pathway (KG decarboxylase; KGD) results in strict dependence upon the glyoxylate shunt for growth, demonstrating that KG decarboxylase is also functional in M. tuberculosis intermediary metabolism. These observations demonstrate that unlike most organisms M. tuberculosis utilizes two distinct TCA pathways from KG, one that functions concurrently with beta-oxidation (KOR-dependent), and one that functions in the absence of beta-oxidation (KGD-dependent). As these pathways are regulated by metabolic cues, we predict that their differential utilization provides an advantage for growth in different environments within the host.


Subject(s)
Citric Acid Cycle/physiology , Ketoglutarate Dehydrogenase Complex/physiology , Mycobacterium tuberculosis/physiology , Pyruvate Synthase/physiology , Anaerobiosis/physiology , Oxidation-Reduction
10.
Biochim Biophys Acta ; 1787(11): 1309-16, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19413950

ABSTRACT

Studies in Bristol in the 1960s and 1970s, led to the recognition that four mitochondrial dehydrogenases are activated by calcium ions. These are FAD-glycerol phosphate dehydrogenase, pyruvate dehydrogenase, NAD-isocitrate dehydrogenase and oxoglutarate dehydrogenase. FAD-glycerol phosphate dehydrogenase is located on the outer surface of the inner mitochondrial membrane and is influenced by changes in cytoplasmic calcium ion concentration. The other three enzymes are located within mitochondria and are regulated by changes in mitochondrial matrix calcium ion concentration. These and subsequent studies on purified enzymes, mitochondria and intact cell preparations have led to the widely accepted view that the activation of these enzymes is important in the stimulation of the respiratory chain and hence ATP supply under conditions of increased ATP demand in many stimulated mammalian cells. The effects of calcium ions on FAD-isocitrate dehydrogenase involve binding to an EF-hand binding motif within this enzyme but the binding sites involved in the effects of calcium ions on the three intramitochondrial dehydrogenases remain to be fully established. It is also emphasised in this article that these three dehydrogenases appear only to be regulated by calcium ions in vertebrates and that this raises some interesting and potentially important developmental issues.


Subject(s)
Calcium/physiology , Glycerolphosphate Dehydrogenase/physiology , Isocitrate Dehydrogenase/physiology , Ketoglutarate Dehydrogenase Complex/physiology , Mitochondria/enzymology , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/physiology , Animals , Enzyme Activation , Humans
11.
Neuroscience ; 158(2): 610-6, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-18996448

ABSTRACT

The alpha-ketoglutarate dehydrogenase complex (KGDHC) which catalyzes the conversion of alpha-ketoglutarate to succinyl-CoA and NADH in mitochondria, is known to generate O(2).- in vitro. To find out if KGDHC contributes to neuronal reactive oxygen species (ROS) increase in situ, we investigated whether the specific inhibitors of cellular KGDHC, succinyl phosphonate (SP) and the SP triethyl ester (TESP), might affect the glutamate-induced ROS production in cultured hippocampal neurons from rats. The concentration-dependent decrease in the mitochondrial potential of the glutamate-overstimulated neurons in the presence of SP or TESP indicated that under the conditions inducing neuronal ROS generation, the inhibitors are delivered to mitochondria, and their subsequent inhibition of KGDHC decreases the mitochondrial potential. The production of O(2).- was detected by reaction with hydroethidine. The distribution of the resulting fluorescence of DNA-ethidium coincided with that of the mitochondrial marker Mitotracker, pointing to the mitochondrial origin of the hydroethidine-detected ROS in response to glutamate (100 microM). At 200 microM, both TESP and SP administered together with glutamate, inhibited the glutamate-induced ROS production by about 20%, with the inhibition increasing to 44% at 500 microM TESP. The decrease in neuronal ROS by specific inhibitors of KGDHC demonstrates that KGDHC is a source of ROS in cultured neurons responding to glutamate. However, increasing the concentration of the strongest KGDHC inhibitor SP to 500 microM even increased the ROS production compared with glutamate alone, presumably due to secondary effects arising upon the strong KGDHC inhibition. Our work extends the current understanding of the glutamate-induced ROS generation in neurons, shedding light on the pathological mechanisms of the KGDHC involvement in glutamate neurotoxicity. In conclusion, potent KGDHC inhibitors are promising diagnostic tools for in situ study of neurodegenerative mechanisms.


Subject(s)
Hippocampus/cytology , Ketoglutarate Dehydrogenase Complex/physiology , Neurons/drug effects , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Cells, Cultured , Complex Mixtures/pharmacology , DNA , Dose-Response Relationship, Drug , Drug Interactions , Ethidium/analogs & derivatives , Membrane Potential, Mitochondrial/drug effects , Organophosphonates/pharmacology , Rats , Succinates/pharmacology , Time Factors
12.
Plant Physiol ; 148(4): 1782-96, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18842826

ABSTRACT

The 2-oxoglutarate dehydrogenase complex constitutes a mitochondrially localized tricarboxylic acid cycle multienzyme system responsible for the conversion of 2-oxoglutarate to succinyl-coenzyme A concomitant with NAD(+) reduction. Although regulatory mechanisms of plant enzyme complexes have been characterized in vitro, little is known concerning their role in plant metabolism in situ. This issue has recently been addressed at the cellular level in nonplant systems via the use of specific phosphonate inhibitors of the enzyme. Here, we describe the application of these inhibitors for the functional analysis of the potato (Solanum tuberosum) tuber 2-oxoglutarate dehydrogenase complex. In vitro experiments revealed that succinyl phosphonate (SP) and a carboxy ethyl ester of SP are slow-binding inhibitors of the 2-oxoglutarate dehydrogenase complex, displaying greater inhibitory effects than a diethyl ester of SP, a phosphono ethyl ester of SP, or a triethyl ester of SP. Incubation of potato tuber slices with the inhibitors revealed that they were adequately taken up by the tissue and produced the anticipated effects on the in situ enzyme activity. In order to assess the metabolic consequences of the 2-oxoglutarate dehydrogenase complex inhibition, we evaluated the levels of a broad range of primary metabolites using an established gas chromatography-mass spectrometry method. We additionally analyzed the rate of respiration in both tuber discs and isolated mitochondria. Finally, we evaluated the metabolic fate of radiolabeled acetate, 2-oxoglutarate or glucose, and (13)C-labeled pyruvate and glutamate following incubation of tuber discs in the presence or absence of either SP or the carboxy ethyl ester of SP. The data obtained are discussed in the context of the roles of the 2-oxoglutarate dehydrogenase complex in respiration and carbon-nitrogen interactions.


Subject(s)
Ketoglutarate Dehydrogenase Complex/physiology , Nitrogen/metabolism , Plant Proteins/physiology , Solanum tuberosum/enzymology , Carbon Dioxide/metabolism , Enzyme Inhibitors/pharmacology , Esters/pharmacology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Organophosphonates/pharmacology , Oxygen/metabolism , Plant Proteins/antagonists & inhibitors , Plant Tubers/drug effects , Plant Tubers/enzymology , Plant Tubers/metabolism , Solanum tuberosum/drug effects , Solanum tuberosum/metabolism , Succinates/pharmacology
13.
Biochim Biophys Acta ; 1782(4): 229-38, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18206986

ABSTRACT

Measures in autopsied brains from Alzheimer's Disease (AD) patients reveal a decrease in the activity of alpha-ketoglutarate dehydrogenase complex (KGDHC) and an increase in malate dehydrogenase (MDH) activity. The present experiments tested whether both changes could be caused by the common oxidant H(2)O(2) and to probe the mechanism underlying these changes. Since the response to H(2)O(2) is modified by the level of the E2k subunit of KGDHC, the interaction of MDH and KGDHC was studied in cells with varying levels of E2k. In cells with only 23% of normal E2k protein levels, one-hour treatment with H(2)O(2) decreased KGDHC and increased MDH activity as well as the mRNA level for both cytosolic and mitochondrial MDH. The increase in MDH did not occur in cells with 100% or 46% of normal E2k. Longer treatments with H(2)O(2) inhibited the activity of both enzymes. Glutathione is a major regulator of cellular redox state and can modify enzyme activities. H(2)O(2) converts reduced glutathione (GSH) to oxidized glutathione (GSSG), which reacts with protein thiols. Treatment of purified KGDHC with GSSG leads to glutathionylation of all three KGDHC subunits. Thus, cellular glutathione level was manipulated by two means to determine the effect on KGDHC and MDH activities. Both buthionine sulfoximine (BSO), which inhibits glutathione synthesis without altering redox state, and H(2)O(2) diminished glutathione to a similar level after 24 h. However, H(2)O(2), but not BSO, reduced KGDHC and MDH activities, and the reduction was greater in the E2k-23 line. These findings suggest that the E2k may mediate diverse responses of KGDHC and MDH to oxidants. In addition, the differential response of activities to BSO and H(2)O(2) together with the in vitro interaction of KGDHC with GSSG suggests that glutathionylation is one possible mechanism underlying oxidative stress-induced inhibition of the TCA cycle enzymes.


Subject(s)
Alzheimer Disease/enzymology , Ketoglutarate Dehydrogenase Complex/physiology , Buthionine Sulfoximine/pharmacology , Cell Line , Glutathione/physiology , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Malate Dehydrogenase/metabolism , Mitochondrial Proteins , Models, Biological , Oxidative Stress , Protein Subunits/metabolism , Reactive Oxygen Species/metabolism
14.
Philos Trans R Soc Lond B Biol Sci ; 360(1464): 2335-45, 2005 Dec 29.
Article in English | MEDLINE | ID: mdl-16321804

ABSTRACT

Alpha-ketoglutarate dehydrogenase (alpha-KGDH) is a highly regulated enzyme, which could determine the metabolic flux through the Krebs cycle. It catalyses the conversion of alpha-ketoglutarate to succinyl-CoA and produces NADH directly providing electrons for the respiratory chain. alpha-KGDH is sensitive to reactive oxygen species (ROS) and inhibition of this enzyme could be critical in the metabolic deficiency induced by oxidative stress. Aconitase in the Krebs cycle is more vulnerable than alpha-KGDH to ROS but as long as alpha-KGDH is functional NADH generation in the Krebs cycle is maintained. NADH supply to the respiratory chain is limited only when alpha-KGDH is also inhibited by ROS. In addition being a key target, alpha-KGDH is able to generate ROS during its catalytic function, which is regulated by the NADH/NAD+ ratio. The pathological relevance of these two features of alpha-KGDH is discussed in this review, particularly in relation to neurodegeneration, as an impaired function of this enzyme has been found to be characteristic for several neurodegenerative diseases.


Subject(s)
Citric Acid Cycle/physiology , Ketoglutarate Dehydrogenase Complex/metabolism , Mitochondria/metabolism , NAD/metabolism , Neurodegenerative Diseases/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Humans , Ketoglutarate Dehydrogenase Complex/physiology
15.
Rejuvenation Res ; 8(1): 13-7, 2005.
Article in English | MEDLINE | ID: mdl-15798370

ABSTRACT

The vicious cycle theory postulates that typical mitochondrial DNA (mtDNA) mutations cause their host mitochondria to generate more superoxide and other reactive oxygen species (ROS) than do normal mitochondria, thereby promoting the occurrence of additional mtDNA mutations at an ever-accelerating rate. However, nearly all the loss-of-function mtDNA mutations seen in vivo are large deletions, which (as the original statement of the theory indeed noted, though this has been widely overlooked) should not trigger a vicious cycle because they will prevent the assembly of the potentially superoxide-generating enzyme complexes. Consistent with this is the observation that each cell exhibiting loss of mtDNA-encoded function in vivo contains copies of a single, evidently clonally expanded, mutant mtDNA species, whereas the vicious cycle theory predicts a spectrum of mutant forms in each cell. Two recent papers, however, unveil a way in which mtDNA mutations could indeed promote ROS production of their host mitochondria. MtDNA mutations probably shift the intramitochondrial NAD(+)/NADH redox couple towards NADH, and this is now shown in vitro to cause ROS production by alpha-ketoglutarate dehydrogenase, an essential enzyme of the TCA cycle. This does not revive the vicious cycle theory, but it has complex implications for the two most plausible more recent theories, known as "survival of the slowest" and "crippled mitochondria." It may also prove to explain other recent observations in mitochondrially mutant cells in vivo.


Subject(s)
DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Mutation/physiology , Reactive Oxygen Species/metabolism , Cell Proliferation , DNA Damage/physiology , Humans , Ketoglutarate Dehydrogenase Complex/physiology , Mutation/genetics
16.
J Biol Chem ; 280(12): 10888-96, 2005 Mar 25.
Article in English | MEDLINE | ID: mdl-15649899

ABSTRACT

The activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC) declines in brains of patients with several neurodegenerative diseases. KGDHC consists of multiple copies of E1k, E2k, and E3. E1k and E2k are unique to KGDHC and may have functions independent of the complex. The present study tested the consequences of different levels of diminished E2k mRNA on protein levels of the subunits, KGDHC activity, and physiological responses. Human embryonic kidney cells were stably transfected with an E2k sense or antisense expression vector. Sense control (E2k-mRNA-100) was compared with two clones in which the mRNA was reduced to 67% of control (E2k-mRNA-67) or to 30% of control (E2k-mRNA-30). The levels of the E2k protein in clones paralleled the reduction in mRNA, and E3 proteins were unaltered. Unexpectedly, the clone with the greatest reduction in E2k protein (E2k-mRNA-30) had a 40% increase in E1k protein. The activity of the complex was only 52% of normal in E2k-mRNA-67 clone, but was near normal (90%) in E2k-mRNA-30 clone. Subsequent experiments tested whether the physiological consequences of a reduction in E2k mRNA correlated more closely to E2k protein or to KGDHC activity. Growth rate, increased DCF-detectable reactive oxygen species, and cell death in response to added oxidant were proportional to E2k proteins, but not complex activity. These results were not predicted because subunits unique to KGDHC have never been manipulated in mammalian cells. These results suggest that in addition to its essential role in metabolism, the E2k component of KGDHC may have other novel roles.


Subject(s)
Acyltransferases/physiology , Ketoglutarate Dehydrogenase Complex/physiology , Cell Line , Cell Proliferation , Cell Survival , Humans , Ketoglutarate Dehydrogenase Complex/chemistry , NAD/metabolism , Protein Subunits , RNA, Antisense/physiology , Reactive Oxygen Species
17.
J Biol Chem ; 279(51): 53782-8, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15466852

ABSTRACT

The 2-oxoglutarate dehydrogenase complex (OGHDC) (also known as the alpha-ketoglutarate dehydrogenase complex) is a rate-limiting enzyme in the mitochondrial Krebs cycle. Here we report that the RING finger ubiquitin-protein isopeptide ligase Siah2 binds to and targets OGDHC-E2 for ubiquitination-dependent degradation. OGDHC-E2 expression and activity are elevated in Siah2(-/-) cells compared with Siah2(+)(/)(+) cells. Deletion of the mitochondrial targeting sequence of OGDHC-E2 results in its cytoplasmic localization and rapid proteasome-dependent degradation in Siah2(+)(/)(+) but not in Siah2(-/-) cells. Significantly, because of its overexpression or disruption of the mitochondrial membrane potential, the release of OGDHC-E2 from mitochondria to the cytoplasm also results in its concomitant degradation. The role of the Siah family of ligases in the regulation of OGDHC-E2 stability is expected to take place under pathological conditions in which the levels of OGDHC-E2 are altered.


Subject(s)
Ketoglutarate Dehydrogenase Complex/physiology , Transcription Factors/physiology , Ubiquitin-Protein Ligases/chemistry , Animals , Blotting, Western , Cell Line , Cytoplasm/metabolism , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Immunoprecipitation , Ketoglutarate Dehydrogenase Complex/chemistry , Kinetics , Membrane Potentials , Mice , Microscopy, Fluorescence , Mitochondria/metabolism , Nuclear Proteins , Plasmids/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Transcription Factors/metabolism , Transfection , Transgenes , Ubiquitin/metabolism
18.
J Ind Microbiol Biotechnol ; 31(7): 295-300, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15257440

ABSTRACT

The efficiency of carbon conversion to biomass and desirable end products in industrial fermentations is diminished by the diversion of carbon to acetate and lactate excretions. In this study, the use of prototrophic and mutant strains of Escherichia coli, as well as enzyme active site directed inhibitors, revealed that flux to acetate excretion is physiologically advantageous to the organism as it facilitates a faster growth rate (mu) and permits growth to high cell densities. Moreover, the abolition of flux to acetate excretion was balanced by the excretion of lactate as well as 2-oxoglutarate, isocitrate and citrate, suggesting a 'bottle-neck' effect at the level of 2-oxoglutarate in the Krebs cycle. It is proposed that the acetate excreting enzymes, phosphotransacetylase and acetate kinase, constitute an anaplerotic loop or by-pass, the primary function of which is to replenish the Krebs cycle with reduced CoA, thus relieving the bottle-neck effect at the level of 2-oxoglutarate dehydrogenase. Furthermore, flux to lactate excretion plays a central role in regenerating proton gradient and maintaining the redox balance within the cell. The long-held view that flux to acetate and lactate excretions is merely a function of an 'over-flow' in central metabolism should, therefore, be re-evaluated.


Subject(s)
Acetates/metabolism , Escherichia coli/metabolism , Lactic Acid/metabolism , Acetate Kinase/physiology , Citric Acid/metabolism , Coenzyme A/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Escherichia coli/growth & development , Gluconates/metabolism , Glucose/metabolism , Isocitrates/metabolism , Ketoglutarate Dehydrogenase Complex/physiology , Ketoglutaric Acids/metabolism , Oxidation-Reduction , Phosphate Acetyltransferase/physiology , Pyruvates/pharmacology , Pyruvic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...