Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Biol Chem ; 299(8): 105001, 2023 08.
Article in English | MEDLINE | ID: mdl-37394006

ABSTRACT

NADH-ubiquinone (UQ) oxidoreductase (complex I) couples electron transfer from NADH to UQ with proton translocation in its membrane part. The UQ reduction step is key to triggering proton translocation. Structural studies have identified a long, narrow, tunnel-like cavity within complex I, through which UQ may access a deep reaction site. To elucidate the physiological relevance of this UQ-accessing tunnel, we previously investigated whether a series of oversized UQs (OS-UQs), whose tail moiety is too large to enter and transit the narrow tunnel, can be catalytically reduced by complex I using the native enzyme in bovine heart submitochondrial particles (SMPs) and the isolated enzyme reconstituted into liposomes. Nevertheless, the physiological relevance remained unclear because some amphiphilic OS-UQs were reduced in SMPs but not in proteoliposomes, and investigation of extremely hydrophobic OS-UQs was not possible in SMPs. To uniformly assess the electron transfer activities of all OS-UQs with the native complex I, here we present a new assay system using SMPs, which were fused with liposomes incorporating OS-UQ and supplemented with a parasitic quinol oxidase to recycle reduced OS-UQ. In this system, all OS-UQs tested were reduced by the native enzyme, and the reduction was coupled with proton translocation. This finding does not support the canonical tunnel model. We propose that the UQ reaction cavity is flexibly open in the native enzyme to allow OS-UQs to access the reaction site, but their access is obstructed in the isolated enzyme as the cavity is altered by detergent-solubilizing from the mitochondrial membrane.


Subject(s)
Electron Transport Complex I , Ubiquinone , Animals , Cattle , Ubiquinone/metabolism , Electron Transport Complex I/metabolism , Mitochondrial Membranes/metabolism , NAD/metabolism , Protons , Liposomes
2.
J Biol Chem ; 298(7): 102075, 2022 07.
Article in English | MEDLINE | ID: mdl-35643318

ABSTRACT

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.


Subject(s)
Electron Transport Complex I , Ubiquinone , Binding Sites , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Submitochondrial Particles/metabolism , Ubiquinone/metabolism
3.
BMC Immunol ; 24(1): 34, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37752417

ABSTRACT

BACKGROUND: Rapid and accurate diagnosis of individuals with SARS-CoV-2 infection is an effective way to prevent and control the spread of COVID-19. Although the detection of SARS-CoV-2 viral RNA by RT-qPCR is the gold standard for COVID-19 testing, the use of antigen-detecting rapid diagnostic tests (Ag-RDTs) is emerging as a complementary surveillance tool as Omicron case numbers skyrocket worldwide. However, the results from Ag-RDTs are less accurate in individuals with low viral loads. RESULTS: To develop a highly sensitive and accurate Ag-RDT, 90 monoclonal antibodies were raised from guinea pigs immunized with SARS CoV-2 nucleocapsid protein (CoV-2-NP). By applying a capture antibody recognizing the structural epitope of the N-terminal domain of CoV-2-NP and a detection antibody recognizing the C-terminal tail of CoV-2-NP to an automated chemiluminescence flow-through membrane immunoassay device, we developed a novel Ag-RDT, CoV-2-POCube. The CoV-2-POCube exclusively recognizes CoV-2-NP variants but not the nucleocapsid proteins of other human coronaviruses. The CoV-2-POCube achieved a limit of detection sensitivity of 0.20 ~ 0.66 pg/mL of CoV-2-NPs, demonstrating more than 100 times greater sensitivity than commercially available SARS-CoV-2 Ag-RDTs. CONCLUSIONS: CoV-2-POCube has high analytical sensitivity and can detect SARS-CoV-2 variants in 15 min without observing the high-dose hook effect, thus meeting the need for early SARS-CoV-2 diagnosis with lower viral load. CoV-2-POCube is a promising alternative to currently available diagnostic devices for faster clinical decision making in individuals with suspected COVID-19 in resource-limited settings.


Subject(s)
Antibodies, Monoclonal , COVID-19 , Humans , Animals , Guinea Pigs , SARS-CoV-2 , COVID-19 Testing , COVID-19/diagnosis , Sensitivity and Specificity , Immunoassay
4.
J Biol Chem ; 297(5): 101173, 2021 11.
Article in English | MEDLINE | ID: mdl-34499926

ABSTRACT

Bisphenol A and its derivatives are recognized as endocrine disruptors based on their complex effects on estrogen receptor (ER) signaling. While the effects of bisphenol derivatives on ERα have been thoroughly evaluated, how these chemicals affect ERß signaling is less well understood. Herein, we sought to identify novel ERß ligands using a radioligand competitive binding assay to screen a chemical library of bisphenol derivatives. Many of the compounds identified showed intriguing dual activities as both ERα agonists and ERß antagonists. Docking simulations of these compounds and ERß suggested that they bound not only to the canonical binding site of ERß but also to the coactivator binding site located on the surface of the receptor, suggesting that they act as coactivator-binding inhibitors (CBIs). Receptor-ligand binding experiments using WT and mutated ERß support the presence of a second ligand-interaction position at the coactivator-binding site in ERß, and direct binding experiments of ERß and a coactivator peptide confirmed that these compounds act as CBIs. Our study is the first to propose that bisphenol derivatives act as CBIs, presenting critical insight for the future development of ER signaling-based drugs and their potential to function as endocrine disruptors.


Subject(s)
Benzhydryl Compounds , Estrogen Receptor beta , Phenols , Signal Transduction/drug effects , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , HeLa Cells , Humans , Mutation , Phenols/chemistry , Phenols/pharmacology , Protein Binding , Signal Transduction/genetics
5.
Biochemistry ; 60(10): 813-824, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33650850

ABSTRACT

The ubiquinone reduction step in NADH-ubiquinone oxidoreductase (complex I) is the key to triggering proton translocation in its membrane part. Although the existence of a long and narrow quinone-access channel has been identified, it remains debatable whether the channel model can account for binding of various ligands (ubiquinones and inhibitors) to the enzyme. We previously proposed that the matrix-side interfacial region of the 49 kDa, ND1, PSST, and 39 kDa subunits, which is covered by a loop connecting transmembrane helices (TMHs) 1 and 2 of ND3, may be the area for entry of some bulky ligands into the quinone reaction cavity. However, this proposition lacks direct evidence that the cavity is accessible from the putative matrix-side region, which allows ligands to pass. To address this, we examined whether Cys39 of ND3 and Asp160 of 49 kDa can be specifically cross-linked by bifunctional cross-linkers (tetrazine-maleimide hybrid, named TMBC). On the basis of the structural models of complex I, such dual cross-linking is unexpected because ND3 Cys39 and 49 kDa Asp160 are located on the TMH1-2 loop and deep inside the channel, respectively, and hence, they are physically separated by peptide chains forming the channel wall. However, three TMBCs with different spacer lengths did cross-link the two residues, resulting in the formation of new cross-linked ND3/49 kDa subunits. Chemical modification of either ND3 Cys39 or 49 kDa Asp160 blocked the dual cross-linking, ensuring the specificity of the cross-linking. Altogether, this study provides direct evidence that the quinone reaction cavity is indeed accessible from the proposed matrix-side region covered by the ND3 TMH1-2 loop.


Subject(s)
Cross-Linking Reagents/chemistry , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , Ubiquinone/metabolism , Animals , Binding Sites , Catalytic Domain , Cattle , Electron Transport , Ligands , Protein Conformation , Protein Subunits , Protons
6.
J Biol Chem ; 295(21): 7481-7491, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32295842

ABSTRACT

The small molecule IACS-010759 has been reported to potently inhibit the proliferation of glycolysis-deficient hypoxic tumor cells by interfering with the functions of mitochondrial NADH-ubiquinone oxidoreductase (complex I) without exhibiting cytotoxicity at tolerated doses in normal cells. Considering the significant cytotoxicity of conventional quinone-site inhibitors of complex I, such as piericidin and acetogenin families, we hypothesized that the mechanism of action of IACS-010759 on complex I differs from that of other known quinone-site inhibitors. To test this possibility, here we investigated IACS-010759's mechanism in bovine heart submitochondrial particles. We found that IACS-010759, like known quinone-site inhibitors, suppresses chemical modification by the tosyl reagent AL1 of Asp160 in the 49-kDa subunit, located deep in the interior of a previously proposed quinone-access channel. However, contrary to the other inhibitors, IACS-010759 direction-dependently inhibited forward and reverse electron transfer and did not suppress binding of the quinazoline-type inhibitor [125I]AzQ to the N terminus of the 49-kDa subunit. Photoaffinity labeling experiments revealed that the photoreactive derivative [125I]IACS-010759-PD1 binds to the middle of the membrane subunit ND1 and that inhibitors that bind to the 49-kDa or PSST subunit cannot suppress the binding. We conclude that IACS-010759's binding location in complex I differs from that of any other known inhibitor of the enzyme. Our findings, along with those from previous study, reveal that the mechanisms of action of complex I inhibitors with widely different chemical properties are more diverse than can be accounted for by the quinone-access channel model proposed by structural biology studies.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Glycolysis/drug effects , Mitochondria, Heart/enzymology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/metabolism , Oxadiazoles/pharmacology , Piperidines/pharmacology , Animals , Cattle , Cell Hypoxia/drug effects , Electron Transport Complex I/metabolism , Humans , Mitochondria, Heart/pathology , Neoplasm Proteins/metabolism , Neoplasms/pathology
7.
J Biol Chem ; 295(36): 12739-12754, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32690607

ABSTRACT

The Na+-pumping NADH-ubiquinone (UQ) oxidoreductase (Na+-NQR) is present in the respiratory chain of many pathogenic bacteria and is thought to be a promising antibiotic target. Whereas many details of Na+-NQR structure and function are known, the mechanisms of action of potent inhibitors is not well-understood; elucidating the mechanisms would not only advance drug design strategies but might also provide insights on a terminal electron transfer from riboflavin to UQ. To this end, we performed photoaffinity labeling experiments using photoreactive derivatives of two known inhibitors, aurachin and korormicin, on isolated Vibrio cholerae Na+-NQR. The inhibitors labeled the cytoplasmic surface domain of the NqrB subunit including a protruding N-terminal stretch, which may be critical to regulate the UQ reaction in the adjacent NqrA subunit. The labeling was blocked by short-chain UQs such as ubiquinone-2. The photolabile group (2-aryl-5-carboxytetrazole (ACT)) of these inhibitors reacts with nucleophilic amino acids, so we tested mutations of nucleophilic residues in the labeled region of NqrB, such as Asp49 and Asp52 (to Ala), and observed moderate decreases in labeling yields, suggesting that these residues are involved in the interaction with ACT. We conclude that the inhibitors interfere with the UQ reaction in two ways: the first is blocking structural rearrangements at the cytoplasmic interface between NqrA and NqrB, and the second is the direct obstruction of UQ binding at this interfacial area. Unusual competitive behavior between the photoreactive inhibitors and various competitors corroborates our previous proposition that there may be two inhibitor binding sites in Na+-NQR.


Subject(s)
Bacterial Proteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Ubiquinone/metabolism , Vibrio cholerae/metabolism , Bacterial Proteins/genetics , NADH, NADPH Oxidoreductases/genetics , Ubiquinone/genetics , Vibrio cholerae/genetics
8.
J Biol Chem ; 295(8): 2449-2463, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31953326

ABSTRACT

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.


Subject(s)
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
9.
Biosci Biotechnol Biochem ; 85(12): 2368-2377, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34625801

ABSTRACT

The mitochondrial machineries presiding over ATP synthesis via oxidative phosphorylation are promising druggable targets. Fusaramin, a 3-acyl tetramic acid isolated from Fusarium concentricum FKI-7550, is an inhibitor of oxidative phosphorylation in Saccharomyces cerevisiae mitochondria, although its target has yet to be identified. Fusaramin significantly interfered with [3H]ADP uptake by yeast mitochondria at the concentration range inhibiting oxidative phosphorylation. A photoreactive fusaramin derivative (pFS-5) specifically labeled voltage-dependent anion channel 1 (VDAC1), which facilitates trafficking of ADP/ATP across the outer mitochondrial membrane. These results strongly suggest that the inhibition of oxidative phosphorylation by fusaramin is predominantly attributable to the impairment of VDAC1 functions. Fusaramin also inhibited FoF1-ATP synthase and ubiquinol-cytochrome c oxidoreductase (complex III) at concentrations higher than those required for the VDAC inhibition. Considering that other tetramic acid derivatives are reported to inhibit FoF1-ATP synthase and complex III, natural tetramic acids were found to elicit multiple inhibitory actions against mitochondrial machineries.


Subject(s)
Oxidative Phosphorylation
10.
Org Biomol Chem ; 17(2): 234-239, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30556075

ABSTRACT

Use of the ku70-deficient strain of Coprinopsis cinerea enabled confirmation within the native context of the central role the sesquiterpene synthase Cop6 plays in lagopodin biosynthesis. Furthermore, yeast in vivo bioconversion and in vitro assays of two cytochrome P450 monooxygenases Cox1 and Cox2 allowed elucidation of the network of oxidation steps that build structural complexity onto the α-cuprenene framework during the biosynthesis of lagopodins. Three new compounds were identified as intermediates formed by the redox enzymes.


Subject(s)
Coprinus/enzymology , Coprinus/metabolism , Sesquiterpenes/metabolism , Biosynthetic Pathways , Coprinus/chemistry , Cytochrome P-450 Enzyme System/metabolism , Fungal Proteins/metabolism , Ligases/metabolism , Oxidation-Reduction , Quinones/chemistry , Quinones/metabolism , Sesquiterpenes/chemistry
11.
Biochemistry ; 56(32): 4279-4287, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28707880

ABSTRACT

We previously showed that a bulky ring-strained cycloalkyne possessing a rhodamine fluorophore directly reacts (via strain-promoted click chemistry) with the azido group incorporated (via ligand-directed tosyl chemistry) into Asp160 in the 49 kDa subunit of complex I in bovine heart submitochondrial particles [Masuya, T., et al. (2014) Biochemistry 53, 7816-7823]. This two-step conjugation may be a promising technique for specific chemical modifications of the quinone-access channel in complex I by various molecular probes, which would lead to new methodologies for studying the enzyme. However, because the reactivities of ring-strained cycloalkynes are generally high, they also react with other nucleophilic amino acids in mitochondrial proteins, resulting in significant undesired side reactions. To minimize side reactions and achieve precise pinpoint chemical modification of 49 kDa Asp160, we investigated an optimal pair of chemical tags for the two-step conjugation reaction. We found that instead of strain-promoted click chemistry, Diels-Alder cycloaddition of a pair of cyclopropene incorporated into 49 kDa Asp160 (via ligand-directed tosyl chemistry) and externally added tetrazine is more efficient for the pinpoint modification. An excess of quinone-site inhibitors did not interfere with Diels-Alder cycloaddition between the cyclopropene and tetrazine. These results along with the previous findings (cited above) strongly suggest that in contrast to the predicted quinone-access channel modeled by X-ray crystallographic and single-particle cryo-electron microscopic studies, the channel is open or undergoes large structural rearrangements to allow bulky ligands into the proximity of 49 kDa Asp160.


Subject(s)
Electron Transport Complex I/chemistry , Mitochondria, Heart/enzymology , Models, Molecular , Molecular Probes/chemistry , Animals , Cattle , Click Chemistry/methods , Cyclopropanes/chemistry
12.
Biochemistry ; 55(23): 3189-97, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27211056

ABSTRACT

Asp160 in the 49 kDa subunit of bovine mitochondrial complex I, which is located in the inner part of the quinone binding cavity, is considered to be an essential residue for energy conversion of the enzyme. To elucidate the catalytic function of this residue, we attempted to specifically methylate 49 kDa Asp160 [Asp(COO)-CH3] through a ligand-directed tosyl (LDT) chemistry technique with an acetogenin derivative (ALM) as a high-affinity ligand. We confirmed the specific methylation of 49 kDa Asp160 through liquid chromatography-tandem mass spectrometry analysis of the tryptic digests of the 49 kDa subunit. The binding affinity of a quinazoline-type inhibitor ([(125)I]AzQ) occupying the quinone binding cavity was not affected by methylation, indicating that this chemical modification does not induce significant structural changes inside the quinone binding cavity. The methylation of 49 kDa Asp160 did not lead to the complete loss of catalytic activity; the modified enzyme retained partial electron transfer and proton translocation activities. These results along with the fact that 49 kDa Asp160 elicits a very strong nucleophilicity against various LDT reagents in the local protein environment strongly suggest that this residue is free from strict interactions (such as electrostatic interaction) arising from nearby residue(s) and is functionally important but not essential for the energy conversion of complex I.


Subject(s)
Aspartic Acid/chemistry , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Mitochondria, Heart/metabolism , Quinazolines/chemistry , Quinones/metabolism , Animals , Aspartic Acid/metabolism , Binding Sites , Catalytic Domain , Cattle , Electron Transport , Enzyme Inhibitors/chemistry , Ligands , Methylation , Mitochondria, Heart/drug effects , Protein Conformation , Protein Subunits , Quinones/chemistry , Tosyl Compounds/chemistry , Tosyl Compounds/pharmacology
13.
Biosci Biotechnol Biochem ; 80(8): 1464-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27140857

ABSTRACT

We previously produced the unique ubiquinone QT ("decoupling" quinone), the catalytic reduction of which in NADH-quinone oxidoreduction with bovine heart mitochondrial NADH-ubiquinone oxidoreductase (complex I) is completely decoupled from proton translocation across the membrane domain. This feature is markedly distinct from those of typical short-chain quinones such as ubiquinone-1. To further characterize the features of the QT reaction with complex I, we herein synthesized three QT analogs, QT2-QT4, and characterized their electron transfer reactions. We found that all aspects of electron transfer (e.g. electron-accepting activity and membrane potential formation) vary significantly among these analogs. The features of QT2 as decoupling quinone were slightly superior to those of original QT. Based on these results, we conclude that the bound positions of QTs within the quinone binding cavity susceptibly change depending on their side-chain structures, and the positions, in turn, govern the behavior of QTs as electron acceptors.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria, Heart/metabolism , NAD/metabolism , Protons , Quinones/metabolism , Ubiquinone/metabolism , Animals , Binding Sites , Cattle , Cell Fractionation , Electron Transport , Electron Transport Complex I/chemistry , Kinetics , Membrane Potential, Mitochondrial , Mitochondria, Heart/chemistry , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NAD/chemistry , Oxidation-Reduction , Quinones/chemistry , Structure-Activity Relationship , Ubiquinone/analogs & derivatives , Ubiquinone/chemical synthesis
14.
Biochemistry ; 53(49): 7816-23, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25419630

ABSTRACT

Through a ligand-directed tosyl (LDT) chemistry strategy using the synthetic acetogenin ligand AL1, we succeeded in the pinpoint alkynylation (-C≡CH) of Asp160 in the 49 kDa subunit of bovine complex I, which may be located in the inner part of the putative quinone binding cavity of the enzyme [Masuya, T., et al. (2014) Biochemistry, 53, 2307-2317]. This study provided a promising technique for diverse chemical modifications of complex I. To further improve this technique for its adaptation to intact complex I, we here synthesized the new acetogenin ligand AL2, possessing an azido (-N3) group in place of the terminal alkyne in AL1, and attempted the pinpoint azidation of complex I in bovine heart submitochondrial particles. Careful proteomic analyses revealed that, just as in the case of AL1, azidation occurred at 49 kDa Asp160 with a reaction yield of ∼50%, verifying the high site specificity of our LDT chemistry using acetogenin ligands. This finding prompted us to speculate that a reactivity of the azido group incorporated into Asp160 (Asp160-N3) against externally added chemicals can be employed to characterize the structural features of the quinone/inhibitor binding cavity. Consequently, we used a ring-strained cycloalkyne possessing a rhodamine fluorophore (TAMRA-DIBO), which can covalently attach to an azido group via so-called click chemistry without Cu¹âº catalysis, as the reaction partner of Asp160-N3. We found that bulky TAMRA-DIBO is capable of reacting directly with Asp160-N3 in intact complex I. Unexpectedly, the presence of an excess amount of short-chain ubiquinones as well as some strong inhibitors (e.g., quinazoline and fenpyroximate) did not interfere with the reaction between TAMRA-DIBO and Asp160-N3; nevertheless, bullatacin, a member of the natural acetogenins, markedly interfered with this reaction. Taking the marked bulkiness of TAMRA-DIBO into consideration, it appears to be difficult to reconcile these results with the proposal that only a narrow entry point accessing to the quinone/inhibitor binding cavity exists in complex I [Baradaran, R., et al. (2013) Nature, 494, 443-448]; rather, they suggest that there may be another access path for TAMRA-DIBO to the cavity.


Subject(s)
Aspartic Acid/chemistry , Electron Transport Complex I/chemistry , Models, Molecular , NADH Dehydrogenase/chemistry , Acetogenins/chemistry , Acetogenins/metabolism , Animals , Benzoates/chemistry , Benzoates/pharmacology , Catalytic Domain/drug effects , Cattle , Click Chemistry , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Furans/chemistry , Furans/pharmacology , Indicators and Reagents/chemistry , Indicators and Reagents/pharmacology , Ligands , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/metabolism , Molecular Weight , NADH Dehydrogenase/antagonists & inhibitors , NADH Dehydrogenase/metabolism , Protein Conformation , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Tosyl Compounds/antagonists & inhibitors , Tosyl Compounds/chemistry , Tosyl Compounds/pharmacology
15.
Biochemistry ; 53(14): 2307-17, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24660830

ABSTRACT

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.


Subject(s)
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
16.
FEBS Lett ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38924556

ABSTRACT

Mitochondrial NADH-ubiquinone oxidoreductase (complex I) couples electron transfer from NADH to ubiquinone with proton translocation in its membrane part. Structural studies have identified a long (~ 30 Å), narrow, tunnel-like cavity within the enzyme, through which ubiquinone may access a deep reaction site. Although various inhibitors are considered to block the ubiquinone reduction by occupying the tunnel's interior, this view is still debatable. We synthesized a phosphatidylcholine-quinazoline hybrid compound (PC-Qz1), in which a quinazoline-type toxophore was attached to the sn-2 acyl chain to prevent it from entering the tunnel. However, PC-Qz1 inhibited complex I and suppressed photoaffinity labeling by another quinazoline derivative, [125I]AzQ. This study provides further experimental evidence that is difficult to reconcile with the canonical ubiquinone-accessing tunnel model.

17.
Biochim Biophys Acta Bioenerg ; 1865(4): 149147, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38906315

ABSTRACT

Ubiquinone (UQ) is an essential player in the respiratory electron transfer system. In Saccharomyces cerevisiae strains lacking the ability to synthesize UQ6, exogenously supplied UQs can be taken up and delivered to mitochondria through an unknown mechanism, restoring the growth of UQ6-deficient yeast in non-fermentable medium. Since elucidating the mechanism responsible may markedly contribute to therapeutic strategies for patients with UQ deficiency, many attempts have been made to identify the machinery involved in UQ trafficking in the yeast model. However, definite experimental evidence of the direct interaction of UQ with a specific protein(s) has not yet been demonstrated. To gain insight into intracellular UQ trafficking via a chemistry-based strategy, we synthesized a hydrophobic UQ probe (pUQ5), which has a photoreactive diazirine group attached to a five-unit isoprenyl chain and a terminal alkyne to visualize and/or capture the labeled proteins via click chemistry. pUQ5 successfully restored the growth of UQ6-deficient S. cerevisiae (Δcoq2) on a non-fermentable carbon source, indicating that this UQ was taken up and delivered to mitochondria, and served as a UQ substrate of respiratory enzymes. Through photoaffinity labeling of the mitochondria isolated from Δcoq2 yeast cells cultured in the presence of pUQ5, we identified many labeled proteins, including voltage-dependent anion channel 1 (VDAC1) and cytochrome c oxidase subunit 3 (Cox3). The physiological relevance of UQ binding to these proteins is discussed.

18.
ACS Med Chem Lett ; 14(2): 211-216, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36793437

ABSTRACT

Mitochondrial oxidative phosphorylation (OXPHOS) is an essential cellular metabolic process that generates ATP. The enzymes involved in OXPHOS are considered to be promising druggable targets. Through screening of an in-house synthetic library with bovine heart submitochondrial particles, we identified a unique symmetric bis-sulfonamide, KPYC01112 (1) as an inhibitor targeting NADH-quinone oxidoreductase (complex I). Structural modifications of KPYC01112 (1) led to the discovery of the more potent inhibitors 32 and 35 possessing long alkyl chains (IC50 = 0.017 and 0.014 µM, respectively). A photoaffinity labeling experiment using a newly synthesized photoreactive bis-sulfonamide ([125I]-43) revealed that it binds to the 49-kDa, PSST, and ND1 subunits which make up the quinone-accessing cavity of complex I.

19.
Biochim Biophys Acta Bioenerg ; 1863(2): 148520, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34896079

ABSTRACT

Tamoxifen has been widely used in the treatment of estrogen receptor (ER)-positive breast cancer, whereas it also exhibits ER-independent anticancer effects in various cancer cell types. As one of the convincing mechanisms underlying the ER-independent effects, induction of apoptosis through mitochondrial dysfunction has been advocated. However, the mechanism of action of tamoxifen even at the isolated mitochondrial level is not fully understood and remains controversial. Here, we attempted to comprehensively understand tamoxifen's multiple actions in isolated rat liver mitochondria through not only revisiting the actions hitherto reported but also conducting originally designed experiments. Using submitochondrial particles, we found that tamoxifen has potential as an inhibitor of both respiratory complex I and ATP synthase. However, these inhibitory effects were not elicited in intact mitochondria, likely because penetration of tamoxifen across the inner mitochondrial membrane is highly restricted owing to its localized positive charge (-N+H(CH3)2). This restricted penetration may also explain why tamoxifen is unable to function as a protonophore-type uncoupler in mitochondria. Moreover, tamoxifen suppressed opening of the mitochondrial permeability transition pore induced by Ca2+ overload through enhancing phosphate uptake into the matrix. The photoaffinity labeling experiments using a photolabile tamoxifen derivative (pTAM1) indicated that pTAM1 specifically binds to voltage-dependent anion channels (VDACs) 1 and 3, which regulate transport of various substances into mitochondria. The binding of tamoxifen to VDAC1 and/or VDAC3 could be responsible for the enhancement of phosphate uptake. Taking all the results together, we consider the principal impairment of mitochondrial functions caused by tamoxifen.


Subject(s)
Tamoxifen
20.
Nat Commun ; 13(1): 4082, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35882843

ABSTRACT

The Na+-pumping NADH-ubiquinone oxidoreductase (Na+-NQR) couples electron transfer from NADH to ubiquinone with Na+-pumping, generating an electrochemical Na+ gradient that is essential for energy-consuming reactions in bacteria. Since Na+-NQR is exclusively found in prokaryotes, it is a promising target for highly selective antibiotics. However, the molecular mechanism of inhibition is not well-understood for lack of the atomic structural information about an inhibitor-bound state. Here we present cryo-electron microscopy structures of Na+-NQR from Vibrio cholerae with or without a bound inhibitor at 2.5- to 3.1-Å resolution. The structures reveal the arrangement of all six redox cofactors including a herein identified 2Fe-2S cluster located between the NqrD and NqrE subunits. A large part of the hydrophilic NqrF is barely visible in the density map, suggesting a high degree of flexibility. This flexibility may be responsible to reducing the long distance between the 2Fe-2S centers in NqrF and NqrD/E. Two different types of specific inhibitors bind to the N-terminal region of NqrB, which is disordered in the absence of inhibitors. The present study provides a foundation for understanding the function of Na+-NQR and the binding manner of specific inhibitors.


Subject(s)
Quinone Reductases , Vibrio cholerae , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Oxidation-Reduction , Quinone Reductases/metabolism , Sodium/metabolism , Vibrio cholerae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL