Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 628
1.
J Biol Chem ; 298(7): 102075, 2022 07.
Article En | MEDLINE | ID: mdl-35643318

The ubiquinone (UQ) reduction step catalyzed by NADH-UQ oxidoreductase (mitochondrial respiratory complex I) is key to triggering proton translocation across the inner mitochondrial membrane. Structural studies have identified a long, narrow, UQ-accessing tunnel within the enzyme. We previously demonstrated that synthetic oversized UQs, which are unlikely to transit this narrow tunnel, are catalytically reduced by native complex I embedded in submitochondrial particles but not by the isolated enzyme. To explain this contradiction, we hypothesized that access of oversized UQs to the reaction site is obstructed in the isolated enzyme because their access route is altered following detergent solubilization from the inner mitochondrial membrane. In the present study, we investigated this using two pairs of photoreactive UQs (pUQm-1/pUQp-1 and pUQm-2/pUQp-2), with each pair having the same chemical properties except for a ∼1.0 Å difference in side-chain widths. Despite this subtle difference, reduction of the wider pUQs by the isolated complex was significantly slower than of the narrower pUQs, but both were similarly reduced by the native enzyme. In addition, photoaffinity-labeling experiments using the four [125I]pUQs demonstrated that their side chains predominantly label the ND1 subunit with both enzymes but at different regions around the tunnel. Finally, we show that the suppressive effects of different types of inhibitors on the labeling significantly changed depending on [125I]pUQs used, indicating that [125I]pUQs and these inhibitors do not necessarily share a common binding cavity. Altogether, we conclude that the reaction behaviors of pUQs cannot be simply explained by the canonical UQ tunnel model.


Electron Transport Complex I , Ubiquinone , Binding Sites , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Submitochondrial Particles/metabolism , Ubiquinone/metabolism
2.
Brief Bioinform ; 22(6)2021 11 05.
Article En | MEDLINE | ID: mdl-34337657

Mitochondria are membrane-bound organelles containing over 1000 different proteins involved in mitochondrial function, gene expression and metabolic processes. Accurate localization of those proteins in the mitochondrial compartments is critical to their operation. A few computational methods have been developed for predicting submitochondrial localization from the protein sequences. Unfortunately, most of these computational methods focus on employing biological features or evolutionary information to extract sequence features, which greatly limits the performance of subsequent identification. Moreover, the efficiency of most computational models is still under explored, especially the deep learning feature, which is promising but requires improvement. To address these limitations, we propose a novel computational method called iDeepSubMito to predict the location of mitochondrial proteins to the submitochondrial compartments. First, we adopted a coding scheme using the ProteinELMo to model the probability distribution over the protein sequences and then represent the protein sequences as continuous vectors. Then, we proposed and implemented convolutional neural network architecture based on the bidirectional LSTM with self-attention mechanism, to effectively explore the contextual information and protein sequence semantic features. To demonstrate the effectiveness of our proposed iDeepSubMito, we performed cross-validation on two datasets containing 424 proteins and 570 proteins respectively, and consisting of four different mitochondrial compartments (matrix, inner membrane, outer membrane and intermembrane regions). Experimental results revealed that our method outperformed other computational methods. In addition, we tested iDeepSubMito on the M187, M983 and MitoCarta3.0 to further verify the efficiency of our method. Finally, the motif analysis and the interpretability analysis were conducted to reveal novel insights into subcellular biological functions of mitochondrial proteins. iDeepSubMito source code is available on GitHub at https://github.com/houzl3416/iDeepSubMito.


Deep Learning , Mitochondrial Proteins/metabolism , Submitochondrial Particles/metabolism , Algorithms , Datasets as Topic , Neural Networks, Computer , Protein Transport
3.
Int J Mol Sci ; 21(6)2020 Mar 21.
Article En | MEDLINE | ID: mdl-32245189

This paper proposes the hypothesis that cytoplasmic organelles directly interact with each other and with gap junctions forming intracellular junctions. This hypothesis originated over four decades ago based on the observation that vesicles lining gap junctions of crayfish giant axons contain electron-opaque particles, similar in size to junctional innexons that often appear to directly interact with junctional innexons; similar particles were seen also in the outer membrane of crayfish mitochondria. Indeed, vertebrate connexins assembled into hexameric connexons are present not only in the membranes of the Golgi apparatus but also in those of the mitochondria and endoplasmic reticulum. It seems possible, therefore, that cytoplasmic organelles may be able to exchange small molecules with each other as well as with organelles of coupled cells via gap junctions.


Axons/metabolism , Connexins/metabolism , Cytoplasm/metabolism , Cytoplasmic Vesicles/metabolism , Gap Junctions/metabolism , Golgi Apparatus/metabolism , Mitochondria/metabolism , Animals , Astacoidea , Axons/ultrastructure , Biological Transport/physiology , Calmodulin/chemistry , Calmodulin/metabolism , Connexins/chemistry , Cytoplasmic Vesicles/ultrastructure , Endoplasmic Reticulum/metabolism , Gap Junctions/ultrastructure , Ion Channels/metabolism , Microscopy, Electron , Mitochondria/ultrastructure , Models, Chemical , Submitochondrial Particles/metabolism , Submitochondrial Particles/ultrastructure
4.
J Biol Chem ; 295(8): 2449-2463, 2020 02 21.
Article En | MEDLINE | ID: mdl-31953326

NADH-quinone oxidoreductase (complex I) couples electron transfer from NADH to quinone with proton translocation across the membrane. Quinone reduction is a key step for energy transmission from the site of quinone reduction to the remotely located proton-pumping machinery of the enzyme. Although structural biology studies have proposed the existence of a long and narrow quinone-access channel, the physiological relevance of this channel remains debatable. We investigated here whether complex I in bovine heart submitochondrial particles (SMPs) can catalytically reduce a series of oversized ubiquinones (OS-UQs), which are highly unlikely to transit the narrow channel because their side chain includes a bulky "block" that is ∼13 Šacross. We found that some OS-UQs function as efficient electron acceptors from complex I, accepting electrons with an efficiency comparable with ubiquinone-2. The catalytic reduction and proton translocation coupled with this reduction were completely inhibited by different quinone-site inhibitors, indicating that the reduction of OS-UQs takes place at the physiological reaction site for ubiquinone. Notably, the proton-translocating efficiencies of OS-UQs significantly varied depending on their side-chain structures, suggesting that the reaction characteristics of OS-UQs affect the predicted structural changes of the quinone reaction site required for triggering proton translocation. These results are difficult to reconcile with the current channel model; rather, the access path for ubiquinone may be open to allow OS-UQs to access the reaction site. Nevertheless, contrary to the observations in SMPs, OS-UQs were not catalytically reduced by isolated complex I reconstituted into liposomes. We discuss possible reasons for these contradictory results.


Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Molecular Probes/metabolism , Ubiquinone/chemistry , Ubiquinone/metabolism , Alkynes/metabolism , Animals , Cattle , Computer Simulation , Electron Transport , Membrane Potential, Mitochondrial , Mitochondrial Proteins/metabolism , Models, Molecular , NAD/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Protein Subunits/metabolism , Proteolipids/metabolism , Protons , Submitochondrial Particles/metabolism
5.
Mol Neurobiol ; 56(4): 2379-2393, 2019 Apr.
Article En | MEDLINE | ID: mdl-30027342

The present study evaluated the effects of AR-A014418 on behavioral and oxidative stress parameters of rats submitted to the animal model of mania induced by ouabain (OUA). Wistar rats were submitted to stereotaxic surgery and received a single intracerebroventricular (ICV) injection of artificial cerebrospinal fluid (aCSF), OUA, or AR-A014418. After 7 days, the animals were submitted to open-field test. After behavioral analysis, the brains were dissected in frontal cortex and hippocampus to the evaluation of oxidative stress. The OUA induced manic-like behavior in rats, which was reversed by AR-A014418 treatment. The ICV administration of OUA increases the levels of superoxide in submitochondrial particles, lipid hydroperoxide (LPH), 4-hydroxynonenal (4-HNE), 8-isoprostane, protein carbonyl, 3-nitrotyrosine, and activity of superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) in both structures evaluated. In general, the treatment with AR-A014418 reversed these effects of OUA on the submitochondrial particles, LPH, 4-HNE, 8-isoprostane, protein carbonyl, 3-nitrotyrosine levels, and SOD activity. Furthermore, the injection of OUA decreased the catalase activity, and AR-A014418 promoted an increase in activity of this enzyme in the brain structures. These results suggest that GSK-3ß inhibition can modulate manic-like behaviors. Also, it can be suggested that inhibition of GSK-3ß can be effective against oxidative stress. However, more studies are needed to better elucidate these mechanisms. Graphical Abstract The effects of AR-A014418 on the behavioral and oxidative stress parameters in the animal model of mania induced by ouabain. Superoxide = superoxide production in submitochondrial particles; LPH = lipid hydroperoxide; 4-HNE = 4-hydroxynonenal; SOD = superoxide dismutase; GPx = glutathione peroxidase; GR = glutathione reductase.


Behavior, Animal , Bipolar Disorder/enzymology , Bipolar Disorder/pathology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Oxidative Stress , Aldehydes/metabolism , Animals , Antioxidants/metabolism , Behavior, Animal/drug effects , Bipolar Disorder/physiopathology , Catalase/metabolism , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Disease Models, Animal , Glutathione Peroxidase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Lipid Peroxidation/drug effects , Male , Motor Activity/drug effects , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats, Wistar , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism , Thiazoles/administration & dosage , Thiazoles/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Urea/administration & dosage , Urea/analogs & derivatives , Urea/pharmacology
6.
Free Radic Biol Med ; 89: 602-13, 2015 Dec.
Article En | MEDLINE | ID: mdl-26456055

The effect of NO between cytochromes b and c of the mitochondrial respiratory chain were studied using submitochondrial particles (SMP) from bovine heart and GSNO and SPER-NO as NO sources. Succinate-cytochrome c reductase (complex II-III) activity (222 ± 4 nmol/min. mg protein) was inhibited by 51% in the presence of 500 µM GSNO and by 48% in the presence of 30 µM SPER-NO, in both cases at ~1.25 µM NO. Neither GSNO nor SPER-NO were able to inhibit succinate-Q reductase activity (complex II; 220 ± 9 nmol/min. mg protein), showing that NO affects complex III. Complex II-III activity was decreased (36%) when SMP were incubated with l-arginine and mtNOS cofactors, indicating that this effect is also produced by endogenous NO. GSNO (500 µM) reduced cytochrome b562 by 71%, in an [O2] independent manner. Hyperbolic increases in O2(•-) (up to 1.3 ± 0.1 nmol/min. mg protein) and H2O2 (up to 0.64 ± 0.05 nmol/min. mg protein) productions were observed with a maximal effect at 500 µM GSNO. The O2(•-)/H2O2 ratio was 1.98 in accordance with the stoichiometry of the O2(•-) disproportionation. Moreover, H2O2 production was increased by 72-74% when heart coupled mitochondria were exposed to 500 µM GSNO or 30 µM SPER-NO. SMP incubated in the presence of succinate showed an EPR signal (g=1.99) compatible with a stable semiquinone. This EPR signal was increased not only by antimycin but also by GSNO and SPER-NO. These signals were not modified under N2 atmosphere, indicating that they are not a consequence to the effect of NOx species on complex III area. These results show that NO interacts with ubiquinone-cytochrome b area producing antimycin-like effects. This behaviour comprises the inhibition of electron transfer, the interruption of the oxidation of cytochromes b, and the enhancement of [UQH(•)]ss which, in turn, leads to an increase in O2(•-) and H2O2 mitochondrial production rates.


Electron Transport Complex III/metabolism , Mitochondria, Heart/metabolism , Nitric Oxide/metabolism , Submitochondrial Particles/metabolism , Animals , Antimycin A/analogs & derivatives , Antimycin A/metabolism , Cattle , Electron Spin Resonance Spectroscopy , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Rats
7.
Arch Biochem Biophys ; 580: 75-83, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26116786

Manganese-induced toxicity has been recently associated with an increased ROS generation from mitochondrial complex II (succinate:ubiquinone oxidoreductase). To achieve a deeper mechanistic understanding how divalent manganese ions (Mn(2+)) could stimulate mitochondrial ROS production we performed investigations with bovine heart submitochondrial particles (SMP). In succinate fueled SMP, the Mn(2+) induced hydrogen peroxide (H2O2) production was blocked by the specific complex II ubiquinone binding site (IIQ) inhibitor atpenin A5 while a further downstream block at complex III increased the rate markedly. This suggests that site IIQ was the source of the reactive oxygen species. Moreover, Mn(2+) ions also accelerated the rate of superoxide dismutation, explaining the general increase in the measured rates of H2O2 production and an attenuation of direct superoxide detection.


Electron Transport Complex II/metabolism , Hydrogen Peroxide/metabolism , Manganese/pharmacology , Mitochondrial Membranes/drug effects , Submitochondrial Particles/drug effects , Animals , Cations, Divalent , Cattle , Electron Transport Complex III/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Pyridones/pharmacology , Reactive Oxygen Species/metabolism , Submitochondrial Particles/metabolism , Succinic Acid/metabolism , Succinic Acid/pharmacology , Superoxide Dismutase/metabolism , Ubiquinone/antagonists & inhibitors , Ubiquinone/metabolism
8.
PLoS One ; 10(6): e0129772, 2015.
Article En | MEDLINE | ID: mdl-26053025

ABCB10 (ATP binding cassette sub-family B10) is a mitochondrial inner-membrane ABC transporter. ABCB10 has been shown to protect the heart from the impact of ROS during ischemia-reperfusion and to allow for proper hemoglobin synthesis during erythroid development. ABC transporters are proteins that increase ATP binding and hydrolysis activity in the presence of the transported substrate. However, molecular entities transported by ABCB10 and its regulatory mechanisms are currently unknown. Here we characterized ATP binding and hydrolysis properties of ABCB10 by using the 8-azido-ATP photolabeling technique. This technique can identify potential ABCB10 regulators, transported substrates and amino-acidic residues required for ATP binding and hydrolysis. We confirmed that Gly497 and Lys498 in the Walker A motif, Glu624 in the Walker B motif and Gly602 in the C-Loop motif of ABCB10 are required for proper ATP binding and hydrolysis activity, as their mutation changed ABCB10 8-Azido-ATP photo-labeling. In addition, we show that the potential ABCB10 transported entity and heme precursor delta-aminolevulinic acid (dALA) does not alter 8-azido-ATP photo-labeling. In contrast, oxidized glutathione (GSSG) stimulates ATP hydrolysis without affecting ATP binding, whereas reduced glutathione (GSH) inhibits ATP binding and hydrolysis. Indeed, we detectABCB10 glutathionylation in Cys547 and show that it is one of the exposed cysteine residues within ABCB10 structure. In all, we characterize essential residues for ABCB10 ATPase activity and we provide evidence that supports the exclusion of dALA as a potential substrate directly transported by ABCB10. Last, we show the first molecular mechanism by which mitochondrial oxidative status, through GSH/GSSG, can regulate ABCB10.


ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Glutathione/metabolism , ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/analogs & derivatives , Aminolevulinic Acid/metabolism , Animals , Azides/metabolism , Biotin/metabolism , Conserved Sequence , Cysteine/metabolism , Glutathione Disulfide/metabolism , HEK293 Cells , Humans , Hydrolysis , Mice , Mitochondria/metabolism , Mutation/genetics , Protein Structure, Tertiary , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism
9.
Biochemistry ; 54(17): 2739-46, 2015 May 05.
Article En | MEDLINE | ID: mdl-25849763

Amilorides, well-known inhibitors of Na(+)/H(+) antiporters, were previously shown to inhibit bacterial and mitochondrial NADH-quinone oxidoreductase (complex I) but were markedly less active for complex I. Because membrane subunits ND2, ND4, and ND5 of bovine complex I are homologous to Na(+)/H(+) antiporters, amilorides have been thought to bind to any or all of the antiporter-like subunits; however, there is currently no direct experimental evidence that supports this notion. To identify the binding site of amilorides in bovine complex I, we synthesized two photoreactive amilorides (PRA1 and PRA2), which have a photoreactive azido (-N3) group and terminal alkyne (-C≡CH) group at the opposite ends of the molecules, respectively, and conducted photoaffinity labeling with bovine heart submitochondrial particles. The terminal alkyne group allows various molecular tags to covalently attach to it via Cu(+)-catalyzed click chemistry, thereby allowing purification and/or detection of the labeled peptides. Proteomic analyses revealed that PRA1 and PRA2 label none of the antiporter-like subunits; they specifically label the accessory subunit B14.5a and core subunit 49 kDa (N-terminal region of Thr25-Glu115), respectively. Suppressive effects of ordinary inhibitors (bullatacin, fenpyroximate, and quinazoline), which bind to the putative quinone binding pocket, on labeling were fairly different between the B14.5a and 49 kDa subunits probably because the binding positions of the three inhibitors differ within the pocket. The results of this study clearly demonstrate that amilorides inhibit complex I activity by occupying the quinone binding pocket rather than directly blocking translocation of protons through the antiporter-like subunits (ND2, ND4, and ND5). The accessory subunit B14.5a may be located adjacent to the N-terminal region of the 49 kDa subunits. The structural features of the quinone binding pocket in bovine complex I were discussed on the basis of these results.


Acid Sensing Ion Channel Blockers/metabolism , Amiloride/metabolism , Benzoquinones/metabolism , Electron Transport Complex I/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cattle , Electron Transport Complex I/chemistry , Molecular Sequence Data , Photoaffinity Labels , Submitochondrial Particles/metabolism
10.
Biochemistry ; 53(14): 2307-17, 2014 Apr 15.
Article En | MEDLINE | ID: mdl-24660830

The site-specific chemical modification of NADH-quinone oxidoreductase (complex I) by various functional probes such as fluorophores and microbeads, without affecting the enzyme activity, may allow single-molecule analyses of putative dynamic conformational changes in the enzyme. In an attempt to address this challenge, we performed site-specific alkynylation of complex I in bovine heart submitochondrial particles by means of a ligand-directed tosylate (LDT) chemistry strategy with synthetic acetogenin ligand 1, which has an alkynylated tosylate in the tail moiety, as a high-affinity ligand against the enzyme. The terminal alkyne was chosen as the tag to be incorporated into the enzyme because this functional group can serve as a "footing" for subsequent diverse chemical modifications via so-called click chemistry (i.e., azide-alkyne [3+2] cycloaddition in water). To identify the position alkynylated by ligand 1, fluorescent tetramethylrhodamine was covalently attached to the incorporated alkyne by click chemistry after the solubilization of complex I. Detailed proteomic analyses revealed that alkynylation occurred at Asp160 in the 49 kDa subunit, which may be located in the inner part of the putative quinone-binding cavity. The alkynylation was completely suppressed in the presence of an excess of other inhibitors such as bullatacin and quinazoline. While the reaction yield of the alkynylation step via LDT chemistry was estimated to be ~50%, the alkynylation unfortunately resulted in the almost complete inhibition of enzyme activity. Nevertheless, the results of this study demonstrate that complex I can be site-specifically alkynylated through LDT chemistry, providing a clue about the diverse chemical modifications of the enzyme in combination with click chemistry.


Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Submitochondrial Particles/metabolism , Tosyl Compounds/chemistry , Animals , Cattle , Chromatography, Liquid , Electron Transport Complex I/chemistry , Electrophoresis, Polyacrylamide Gel , Ligands , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Bioorg Med Chem ; 22(2): 684-91, 2014 Jan 15.
Article En | MEDLINE | ID: mdl-24393721

Tocopherols (TOH) are lipophilic antioxidants which require the phenolic OH group for their redox activity. In contrast, non-redox active esters of α-TOH with succinate (α-TOS) were shown to possess proapoptotic activity in cancer cells. It was suggested that this activity is mediated via mitochondrial inhibition with subsequent O2(-) production triggering apoptosis and that the modification of the linker between the succinate and the lipophilic chroman may modulate this activity. However, the specific mechanism and the influence of the linker are not clear yet on the level of the mitochondrial respiratory chain. Therefore, this study systematically compared the effects of α-TOH acetate (α-TOA), α-TOS and α-tocopheramine succinate (α-TNS) in cells and submitochondrial particles (SMP). The results showed that not all cancer cell lines are highly sensitive to α-TOS and α-TNS. In HeLa cells α-TNS did more effectively reduce cell viability than α-TOS. The complex I activity of SMP was little affected by α-TNS and α-TOS while the complex II activity was much more inhibited (IC50=42±8µM α-TOS, 106±8µM α-TNS, respectively) than by α-TOA (IC50 >1000µM). Also the complex III activity was inhibited by α-TNS (IC50=137±6µM) and α-TOS (IC50=315±23µM). Oxygen consumption of NADH- or succinate-respiring SMP, involving the whole electron transfer machinery, was dose-dependently decreased by α-TOS and α-TNS, but only marginal effects were observed in the presence of α-TOA. In contrast to the similar inhibition pattern of α-TOS and α-TNS, only α-TOS triggered O2(-) formation in succinate- and NADH-respiring SMP. Inhibitor studies excluded complex I as O2(-) source and suggested an involvement of complex III in O2(-) production. In cancer cells only α-TOS was reproducibly able to increase O2(-) levels above the background level but neither α-TNS nor α-TOA. Furthermore, the stability of α-TNS in liver homogenates was significantly lower than that of α-TOS. In conclusion, this suggests that α-TNS although it has a structure similar to α-TOS is not acting via the same mechanism and that for α-TOS not only complex II but also complex III interactions are involved.


Antineoplastic Agents/pharmacology , Mitochondria/drug effects , Succinates/pharmacology , Superoxides/metabolism , Vitamin E/analogs & derivatives , alpha-Tocopherol/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex II/metabolism , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Free Radicals/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Molecular Structure , Structure-Activity Relationship , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism , Succinates/chemistry , Succinates/metabolism , Tumor Cells, Cultured , Vitamin E/chemistry , Vitamin E/metabolism , Vitamin E/pharmacology , alpha-Tocopherol/chemistry , alpha-Tocopherol/metabolism
12.
Eur Biophys J ; 42(9): 709-20, 2013 Sep.
Article En | MEDLINE | ID: mdl-23903554

Physiological and pathological functions of mitochondria are highly dependent on the properties and regulation of mitochondrial ion channels. There is still no clear understanding of the molecular identity, regulation, and properties of anion mitochondrial channels. The inner membrane anion channel (IMAC) was assumed to be equivalent to mitochondrial centum picosiemens (mCS). However, the different properties of IMAC and mCS channels challenges this opinion. In our study, we characterized the single-channel anion selectivity and pH regulation of chloride channels from purified cardiac mitochondria. We observed that channel conductance decreased in the order: Cl⁻ > Br⁻ > I⁻ > chlorate ≈ formate > acetate, and that gluconate did not permeate under control conditions. The selectivity sequence was Br⁻ ≥ chlorate ≥ I⁻ ≥ Cl⁻ ≥ formate ≈ acetate. Measurement of the concentration dependence of chloride conductance revealed altered channel gating kinetics, which was demonstrated by prolonged mean open time value with increasing chloride concentration. The observed mitochondrial chloride channels were in many respects similar to those of mCS, but not those of IMAC. Surprisingly, we observed that acidic pH increased channel conductance and that an increase of pH from 7.4 to 8.5 reduced it. The gluconate current appeared and gradually increased when pH decreased from pH 7.0 to 5.6. Our results indicate that pH regulates the channel pore diameter in such a way that dilation increases with more acidic pH. We assume this newly observed pH-dependent anion channel property may be involved in pH regulation of anion distribution in different mitochondrial compartments.


Chloride Channels/chemistry , Chloride Channels/metabolism , Electrophysiological Phenomena , Mitochondria/metabolism , Animals , Electrophysiological Phenomena/drug effects , Gluconates/metabolism , Glycolates/pharmacology , Hydrogen-Ion Concentration , Magnesium/pharmacology , Male , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Porosity , Protein Conformation/drug effects , Rats , Rats, Wistar , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism , Substrate Specificity
13.
Acta Biotheor ; 61(2): 259-68, 2013 Jun.
Article En | MEDLINE | ID: mdl-23475502

The mitochondrion is a key organelle of eukaryotic cell that provides the energy for cellular activities. Correctly identifying submitochondria locations of proteins can provide plentiful information for understanding their functions. However, using web-experimental methods to recognize submitochondria locations of proteins are time-consuming and costly. Thus, it is highly desired to develop a bioinformatics method to predict the submitochondria locations of mitochondrion proteins. In this work, a novel method based on support vector machine was developed to predict the submitochondria locations of mitochondrion proteins by using over-represented tetrapeptides selected by using binomial distribution. A reliable and rigorous benchmark dataset including 495 mitochondrion proteins with sequence identity ≤25% was constructed for testing and evaluating the proposed model. Jackknife cross-validated results showed that the 91.1% of the 495 mitochondrion proteins can be correctly predicted. Subsequently, our model was estimated by three existing benchmark datasets. The overall accuracies are 94.0, 94.7 and 93.4%, respectively, suggesting that the proposed model is potentially useful in the realm of mitochondrion proteome research. Based on this model, we built a predictor called TetraMito which is freely available at http://lin.uestc.edu.cn/server/TetraMito.


Oligopeptides/metabolism , Submitochondrial Particles/metabolism , Models, Theoretical , Support Vector Machine
14.
PLoS One ; 8(3): e59989, 2013.
Article En | MEDLINE | ID: mdl-23527291

Trauma and sepsis can cause acute lung injury (ALI) and Acute Respiratory Distress Syndrome (ARDS) in part by triggering neutrophil (PMN)-mediated increases in endothelial cell (EC) permeability. We had shown that mitochondrial (mt) damage-associated molecular patterns (DAMPs) appear in the blood after injury or shock and activate human PMN. So we now hypothesized that mitochondrial DAMPs (MTD) like mitochondrial DNA (mtDNA) and peptides might play a role in increased EC permeability during systemic inflammation and proceeded to evaluate the underlying mechanisms. MtDNA induced changes in EC permeability occurred in two phases: a brief, PMN-independent 'spike' in permeability was followed by a prolonged PMN-dependent increase in permeability. Fragmented mitochondria (MTD) caused PMN-independent increase in EC permeability that were abolished with protease treatment. Exposure to mtDNA caused PMN-EC adherence by activating expression of adherence molecule expression in both cell types. Cellular activation was manifested as an increase in PMN calcium flux and EC MAPK phosphorylation. Permeability and PMN adherence were attenuated by endosomal TLR inhibitors. EC lacked formyl peptide receptors but were nonetheless activated by mt-proteins, showing that non-formylated mt-protein DAMPs can activate EC. Mitochondrial DAMPs can be released into the circulation by many processes that cause cell injury and lead to pathologic endothelial permeability. We show here that mitochondria contain multiple DAMP motifs that can act on EC and/or PMN via multiple pathways. This can enhance PMN adherence to EC, activate PMN-EC interactions and subsequently increase systemic endothelial permeability. Mitochondrial DAMPs may be important therapeutic targets in conditions where inflammation pathologically increases endothelial permeability.


Acute Lung Injury/physiopathology , Cell Membrane Permeability/physiology , Endothelial Cells/physiology , Neutrophils/metabolism , Respiratory Distress Syndrome/physiopathology , Submitochondrial Particles/metabolism , Blotting, Western , Cell Adhesion/physiology , Cell Culture Techniques , Cells, Cultured , DNA, Mitochondrial/metabolism , Humans , Microscopy, Confocal , Polymerase Chain Reaction , Submitochondrial Particles/pathology
15.
Arch Biochem Biophys ; 529(1): 45-54, 2013 Jan 01.
Article En | MEDLINE | ID: mdl-23142682

Peroxynitrite (ONOO(-)) formation in mitochondria may be favored due to the constant supply of superoxide radical (O(2)(∙-)) by the electron transport chain plus the facile diffusion of nitric oxide ((∙)NO) to this organelle. Herein, a model system of submitochondrial particles (SMP) in the presence of succinate plus the respiratory inhibitor antimycin A (to increase O(2)(∙-) rates) and the (∙)NO-donor NOC-7 was studied to directly establish and quantitate peroxynitrite by a multiplicity of methods including chemiluminescence, fluorescence and immunochemical analysis. While all the tested probes revealed peroxynitrite at near stoichiometric levels with respect to its precursor radicals, coumarin boronic acid (a probe that directly reacts with peroxynitrite) had the more straightforward oxidation profile from O(2)(∙-)-forming SMP as a function of the (∙)NO flux. Interestingly, immunospintrapping studies verified protein radical generation in SMP by peroxynitrite. Substrate-supplemented SMP also reduced Mn(III)porphyrins (MnP) to Mn(II)P under physiologically-relevant oxygen levels (3-30 µM); then, Mn(II)P were capable to reduce peroxynitrite and protect SMP from the inhibition of complex I-dependent oxygen consumption and protein radical formation and nitration of membranes. The data directly support the formation of peroxynitrite in mitochondria and demonstrate that MnP can undergo a catalytic redox cycle to neutralize peroxynitrite-dependent mitochondrial oxidative damage.


Antioxidants/chemistry , Manganese/chemistry , Metalloporphyrins/chemistry , Nitric Oxide/chemistry , Peroxynitrous Acid/metabolism , Submitochondrial Particles/metabolism , Animals , Antimycin A/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Catalysis , Electron Transport/drug effects , Hydrazines/pharmacology , Luminescent Measurements , Manganese/metabolism , Metalloporphyrins/metabolism , Metalloporphyrins/pharmacology , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , NAD/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Oxidation-Reduction , Oxygen/chemistry , Oxygen/metabolism , Peroxynitrous Acid/antagonists & inhibitors , Rats , Spectrometry, Fluorescence , Submitochondrial Particles/drug effects , Succinic Acid/metabolism , Superoxides/antagonists & inhibitors , Superoxides/metabolism
16.
J Cell Biol ; 199(2): 215-24, 2012 Oct 15.
Article En | MEDLINE | ID: mdl-23045551

Mclk1 (also known as Coq7) and Coq3 code for mitochondrial enzymes implicated in the biosynthetic pathway of ubiquinone (coenzyme Q or UQ). Mclk1(+/-) mice are long-lived but have dysfunctional mitochondria. This phenotype remains unexplained, as no changes in UQ content were observed in these mutants. By producing highly purified submitochondrial fractions, we report here that Mclk1(+/-) mice present a unique mitochondrial UQ profile that was characterized by decreased UQ levels in the inner membrane coupled with increased UQ in the outer membrane. Dietary-supplemented UQ(10) was actively incorporated in both mitochondrial membranes, and this was sufficient to reverse mutant mitochondrial phenotypes. Further, although homozygous Coq3 mutants die as embryos like Mclk1 homozygous null mice, Coq3(+/-) mice had a normal lifespan and were free of detectable defects in mitochondrial function or ubiquinone distribution. These findings indicate that MCLK1 regulates both UQ synthesis and distribution within mitochondrial membranes.


Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxygen Consumption/genetics , Ubiquinone/metabolism , Animals , Cell Respiration/genetics , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/deficiency , Mixed Function Oxygenases , Submitochondrial Particles/metabolism , Ubiquinone/administration & dosage , Ubiquinone/genetics
17.
Biochemistry ; 50(50): 10792-803, 2011 Dec 20.
Article En | MEDLINE | ID: mdl-22091587

The mitochondrial electron transport chain (ETC) is a major source of free radical production. However, due to the highly reactive nature of radical species and their short lifetimes, accurate detection and identification of these molecules in biological systems is challenging. The aim of this investigation was to determine the free radical species produced from the mitochondrial ETC by utilizing EPR spin-trapping techniques and the recently commercialized spin-trap, 5-(2,2-dimethyl-1,3-propoxycyclophosphoryl)-5-methyl-1-pyrroline N-oxide (CYPMPO). We demonstrate that this spin-trap has the preferential quality of having minimal mitochondrial toxicity at concentrations required for radical detection. In rat heart mitochondria and submitochondrial particles supplied with NADH, the major species detected under physiological pH was a carbon-centered radical adduct, indicated by markedly large hyperfine coupling constant with hydrogen (a(H) > 2.0 mT). In the presence of the ETC inhibitors, the carbon-centered radical formation was increased and exhibited NADH concentration dependency. The same carbon-centered radical could also be produced with the NAD biosynthesis precursor, nicotinamide mononucleotide, in the presence of a catalytic amount of NADH. The results support the conclusion that the observed species is a complex I derived NADH radical. The formation of the NADH radical could be blocked by hydroxyl radical scavengers but not SOD. In vitro experiments confirmed that an NADH-radical is readily formed by hydroxyl radical but not superoxide anion, further implicating hydroxyl radical as an upstream mediator of NADH radical production. These findings demonstrate the identification of a novel mitochondrial radical species with potential physiological significance and highlight the diverse mechanisms and sites of production within the ETC.


Electron Transport Chain Complex Proteins/metabolism , Mitochondria, Heart/metabolism , NAD/chemistry , NAD/metabolism , Spin Trapping , Animals , Biocatalysis/drug effects , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Electron Spin Resonance Spectroscopy , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Free Radicals/chemistry , Free Radicals/metabolism , Hydroxyl Radical/chemistry , Hydroxyl Radical/metabolism , Kinetics , Male , Mitochondria, Heart/drug effects , Nicotinamide Mononucleotide/chemistry , Nicotinamide Mononucleotide/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism , Uncoupling Agents/pharmacology
18.
Biochemistry ; 50(32): 6901-8, 2011 Aug 16.
Article En | MEDLINE | ID: mdl-21721533

Quinazolines are strong inhibitors of NADH-ubiquinone oxidoreductase (complex I) from bovine heart mitochondria. Using a photoreactive quinazoline, [(125)I]AzQ, and bovine heart submitochondrial particles (SMPs), we demonstrated previously that [(125)I]AzQ binds at the interface of the 49 kDa and ND1 subunits in complex I; it labeled a site in the N-terminal (Asp41-Arg63) region of the 49 kDa subunit, suggesting that this region contacts the ND1 subunit [Murai, M., et al. (2009) Biochemistry 48, 688-698]. The labeled region of ND1 could not be identified because it is highly hydrophobic, and the SMPs did not yield sufficient amounts of labeled protein. Here, we describe how photoaffinity labeling of isolated complex I by [(125)I]AzQ yielded sufficient material for identification of the labeled region of the ND1 subunit. The inhibition of the isolated enzyme by AzQ is comparable to that of SMPs. Our results reveal that the labeled site in ND1 is between Asp199 and Lys262, mostly likely in the third matrix loop that connects the fifth and sixth transmembrane helices. Thus, our results reveal new information about the interface between the hydrophilic and hydrophobic domains of complex I, a region that is thought to be important for ubiquinone reduction and energy transduction.


Electron Transport Complex I/metabolism , Animals , Binding Sites , Cattle , Electron Transport Complex I/chemistry , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Mitochondria, Heart/enzymology , Photoaffinity Labels , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Submitochondrial Particles/metabolism
19.
Biochemistry ; 49(23): 4794-803, 2010 Jun 15.
Article En | MEDLINE | ID: mdl-20459120

Biochemical characterization of the inhibition mechanism of Deltalac-acetogenins synthesized in our laboratory indicated that they are a new type of inhibitor of bovine heart mitochondrial NADH-ubiquinone oxidoreductase (complex I) [Murai, M., et al. (2006) Biochemistry 45, 9778-9787]. To identify the binding site of Deltalac-acetogenins with a photoaffinity labeling technique, we synthesized a photoreactive Deltalac-acetogenin ([(125)I]diazinylated Deltalac-acetogenin, [(125)I]DAA) which has a small photoreactive diazirine group attached to a pharmacophore, the bis-THF ring moiety. Characterization of the inhibitory effects of DAA on bovine complex I revealed unique features specific to, though not completely the same as those of, the original Deltalac-acetogenin. Using [(125)I]DAA, we carried out photoaffinity labeling with bovine heart submitochondrial particles. Analysis of the photo-cross-linked protein by Western blotting and immunoprecipitation revealed that [(125)I]DAA binds to the membrane subunit ND1 with high specificity. The photo-cross-linking to the ND1 subunit was suppressed by an exogenous short-chain ubiquinone (Q(2)) in a concentration-dependent manner. Careful examination of the fragmentation patterns of the cross-linked ND1 generated by limited proteolysis using lysylendopeptidase, endoprotease Asp-N, or trypsin and their changes in the presence of the original Deltalac-acetogenin strongly suggested that the cross-linked residues are located at two different sites in the third matrix-side loop connecting the fifth and sixth transmembrane helices.


Acetogenins/metabolism , Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Acetogenins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cattle , Diazomethane/chemistry , Diazomethane/metabolism , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/chemistry , Furans/chemistry , Furans/metabolism , Hydrophobic and Hydrophilic Interactions , Iodine Radioisotopes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mitochondria, Heart/enzymology , Molecular Sequence Data , NADH Dehydrogenase/antagonists & inhibitors , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Photoaffinity Labels/metabolism , Protein Structure, Tertiary , Submitochondrial Particles/enzymology , Submitochondrial Particles/metabolism
20.
Neuroscience ; 168(1): 2-10, 2010 Jun 16.
Article En | MEDLINE | ID: mdl-20359525

Pyrethroids are one of the most widely used class of insecticides and their toxicity is dominated by pharmacological actions upon the CNS. This study reports as the subchronic treatment (60 days) with permethrin (PERM) (1/10 of LD(50)) induced nuclear DNA damage in rat striatum cells. Comet assay outcomes showed that PERM produced single- and double-strand breaks in striatum cells, the DNA damage was not related to oxidation at pyrimidine and purine bases. Vitamin E (280 mg/kg body weight/day) and vitamin E+coenzyme Q(10) (10 mg/kg/3 ml) supplementation could protect PERM treated rats against nuclear DNA damage. With the aim to evaluate the cause of nuclear DNA damage observed in striatum of rat treated with PERM, in vitro studies on striatum submitochondrial particles (SMPs) and on striatum cells treated with 10 muM PERM alone or plus 16 or 32 nM GSH were performed. SMPs incubated with PERM showed a decrease in superoxide anion release from the electron transport chain by inhibition of mitochondrial complex I. The effect could be related to the decrease of membrane fluidity measured in the hydrophilic-hydrophobic region of the mitochondrial membrane. This result discarded the involvement of the mitochondrial reactive oxygen species in the nuclear DNA damage. On the contrary, GSH played a crucial role on striatum since it was able to protect the cells against nuclear DNA damage induced by PERM. In conclusion our outcomes suggested that nuclear DNA damage of striatum cells was directly related to GSH depletion due to PERM insecticide.


Cell Nucleus/metabolism , Corpus Striatum/drug effects , DNA Damage/drug effects , Glutathione/physiology , Insecticides/toxicity , Permethrin/toxicity , Animals , Corpus Striatum/metabolism , Electron Transport Complex I/antagonists & inhibitors , Male , Membrane Fluidity/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Rats , Rats, Wistar , Submitochondrial Particles/drug effects , Submitochondrial Particles/metabolism , Superoxides/metabolism
...