Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 669
Filter
1.
Nature ; 615(7954): 934-938, 2023 03.
Article in English | MEDLINE | ID: mdl-36949187

ABSTRACT

Mitochondrial energy conversion requires an intricate architecture of the inner mitochondrial membrane1. Here we show that a supercomplex containing all four respiratory chain components contributes to membrane curvature induction in ciliates. We report cryo-electron microscopy and cryo-tomography structures of the supercomplex that comprises 150 different proteins and 311 bound lipids, forming a stable 5.8-MDa assembly. Owing to subunit acquisition and extension, complex I associates with a complex IV dimer, generating a wedge-shaped gap that serves as a binding site for complex II. Together with a tilted complex III dimer association, it results in a curved membrane region. Using molecular dynamics simulations, we demonstrate that the divergent supercomplex actively contributes to the membrane curvature induction and tubulation of cristae. Our findings highlight how the evolution of protein subunits of respiratory complexes has led to the I-II-III2-IV2 supercomplex that contributes to the shaping of the bioenergetic membrane, thereby enabling its functional specialization.


Subject(s)
Cryoelectron Microscopy , Electron Transport Complex III , Electron Transport Complex II , Electron Transport Complex IV , Electron Transport Complex I , Mitochondria , Mitochondrial Membranes , Electron Transport , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex III/ultrastructure , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Electron Transport Complex IV/ultrastructure , Mitochondria/chemistry , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Electron Transport Complex II/chemistry , Electron Transport Complex II/metabolism , Electron Transport Complex II/ultrastructure , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Electron Transport Complex I/ultrastructure , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Molecular Dynamics Simulation , Binding Sites , Evolution, Molecular
2.
J Exp Zool A Ecol Integr Physiol ; 339(2): 153-162, 2023 03.
Article in English | MEDLINE | ID: mdl-36285344

ABSTRACT

Management of fish populations for conservation in thermally variable systems requires an understanding of the fish's underlying physiology and responses to thermal stress. Physiological research at the organismal level provides information on the overall effects of stressors such as extreme temperature fluctuations. While experiments with whole organisms provide information as to the overall effects of temperature fluctuations, biochemical assays of thermal stress provide direct results of exposure that are both sensitive and specific. Electron transport system (ETS; Complex III) assays quantify a rate-limiting step of respiratory enzymes. Parameters that can be estimated via this approach include optimum thermal temperature (Topt ) and optimal breadth of thermal performance (Tbreadth ), which can both be related to organismal-level temperature thresholds. We exposed enzymes of seven fish species (native fish chosen to represent a typical community in Alabama streams) to temperatures in the range 11-44°C. The resultant enzymatic thermal performance curves showed that Topt , the lower temperature for enzyme optimal thermal performance (Tlow ), the upper temperature for enzyme optimal thermal performance (Tup ), and Tbreadth differed among species. Relationships between enzymatic activity and temperature for all fish followed a pattern of steadily increasing enzyme activity to Topt before gradually decreasing with increasing temperature. A comparison of our enzyme optimum and upper-temperature limit results versus published critical thermal maxima values supports that ETS Complex III assays may be useful for assessing organismal-level thermal tolerance.


Subject(s)
Electron Transport Complex III , Fishes , Animals , Alabama , Electron Transport , Electron Transport Complex III/chemistry , Electron Transport Complex III/physiology , Fishes/physiology , Temperature , Fish Proteins/chemistry , Fish Proteins/physiology
3.
Pest Manag Sci ; 78(6): 2657-2666, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35355395

ABSTRACT

BACKGROUND: Fenpicoxamid and florylpicoxamid are picolinamide fungicides targeting the Qi site of the cytochrome bc1 complex, via their primary metabolites UK-2A and CAS-649, respectively. We explore binding interactions and resistance mechanisms for picolinamides, antimycin A and ilicicolin H in yeast by testing effects of cytochrome b amino acid changes on fungicide sensitivity and interpreting results using molecular docking. RESULTS: Effects of amino acid changes on sensitivity to UK-2A and CAS-649 were similar, with highest resistance associated with exchanges involving G37 and substitutions N31K and L198F. These changes, as well as K228M, also affected antimycin A, while ilicicolin H was affected by changes at G37 and L198, as well as Q22E. N31 substitution patterns suggest that a lysine at position 31 introduces an electrostatic interaction with neighbouring D229, causing disruption of a key salt-bridge interaction with picolinamides. Changes involving G37 and L198 imply resistance primarily through steric interference. G37 changes also showed differences between CAS-649 and UK-2A or antimycin A with respect to branched versus unbranched amino acids. N31K and substitution of G37 by large amino acids reduced growth rate substantially while L198 substitutions showed little effect on growth. CONCLUSION: Binding of UK-2A and CAS-649 at the Qi site involves similar interactions such that general cross-resistance between fenpicoxamid and florylpicoxamid is anticipated in target pathogens. Some resistance mutations reduced growth rate and could carry a fitness penalty in pathogens. However, certain changes involving G37 and L198 carry little or no growth penalty and may pose the greatest risk for resistance development in the field. © 2022 Society of Chemical Industry.


Subject(s)
Electron Transport Complex III , Fungicides, Industrial , Picolinic Acids , Amino Acids , Antimycin A/pharmacology , Cytochromes , Electron Transport Complex III/chemistry , Electron Transport Complex III/genetics , Fungicides, Industrial/chemistry , Fungicides, Industrial/pharmacology , Lactones/chemistry , Lactones/metabolism , Molecular Docking Simulation , Mutation , Picolinic Acids/metabolism , Pyridines/chemistry , Pyridines/metabolism , Saccharomyces cerevisiae/genetics
4.
J Mol Model ; 28(2): 35, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35022913

ABSTRACT

The escalating burden of tuberculosis disease and drastic effects of current medicine has stimulated a search for alternative drugs. A medicinal plant Warburgia salutaris has been reported to possess inhibitory properties against M. tuberculosis. In this study, we apply computational methods to investigate the probability of W. salutaris compounds as potential inhibitors of M. tuberculosis QcrB protein. We performed molecular docking, molecular dynamics simulations, radius of gyration, principal component analysis (PCA), and molecular mechanics-generalized born surface area (MM-GBSA) binding-free energy calculations in explicit solvent to achieve our objective. The results suggested that ursolic acid (UA) and ursolic acid acetate (UAA) could serve as preferred potential inhibitors of mycobacterial QcrB compared to lansoprazole sulphide (LSPZ) and telacebec (Q203)-UA and UAA have a higher binding affinity to QcrB compared to LSPZ and Q203 drugs. UA binding affinity is attributed to hydrogen bond formation with Val120, Arg364 and Arg366, and largely resonated from van der Waals forces resulting from UA interactions with hydrophobic amino acids in its vicinity. UAA binds to the porphyrin ring binding site with higher binding affinity compared to LSPZ. The binding affinity results primarily from van der Waals forces between UAA and hydrophobic residues of QcrB in the porphyrin ring binding site where UAA binds competitively. UA and UAA formed stable complexes with the protein with reduced overall residue mobility, consequently supporting the magnitude of binding affinity of the respective ligands. UAA could potentially compete with the porphyrin ring for the binding site and deprive the mycobacterial cell from oxygen, consequently disturbing mycobacterial oxygen-dependent metabolic processes. Therefore, discovery of a compound that competes with porphyrin ring for the binding site may be useful in QcrB pharmocological studies. UA proved to be a superior compound, although its estimated toxicity profile revealed UA to be hepatotoxic within acceptable parameters. Although preliminary findings of this report still warrant experimental validation, they could serve as a baseline for the development of new anti-tubercular drugs from natural resources that target QcrB.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Electron Transport Complex III/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Triterpenes/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Electron Transport Complex III/antagonists & inhibitors , Ligands , Molecular Conformation , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Protein Binding , Structure-Activity Relationship , Triterpenes/pharmacology , Ursolic Acid
5.
Nat Commun ; 13(1): 545, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35087070

ABSTRACT

Proton-translocating respiratory complexes assemble into supercomplexes that are proposed to increase the efficiency of energy conversion and limit the production of harmful reactive oxygen species during aerobic cellular respiration. Cytochrome bc complexes and cytochrome aa3 oxidases are major drivers of the proton motive force that fuels ATP generation via respiration, but how wasteful electron- and proton transfer is controlled to enhance safety and efficiency in the context of supercomplexes is not known. Here, we address this question with the 2.8 Å resolution cryo-EM structure of the cytochrome bcc-aa3 (III2-IV2) supercomplex from the actinobacterium Corynebacterium glutamicum. Menaquinone, substrate mimics, lycopene, an unexpected Qc site, dioxygen, proton transfer routes, and conformational states of key protonable residues are resolved. Our results show how safe and efficient energy conversion is achieved in a respiratory supercomplex through controlled electron and proton transfer. The structure may guide the rational design of drugs against actinobacteria that cause diphtheria and tuberculosis.


Subject(s)
Actinobacteria/metabolism , Corynebacterium glutamicum/metabolism , Cytochromes/chemistry , Cytochromes/metabolism , Oxidoreductases/metabolism , Benzoquinones/chemistry , Binding Sites , Cryoelectron Microscopy , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Energy Metabolism , Models, Molecular , Oxygen/metabolism , Proton-Motive Force
6.
Structure ; 30(1): 129-138.e4, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34525326

ABSTRACT

Electron transfer between respiratory complexes drives transmembrane proton translocation, which powers ATP synthesis and membrane transport. The homodimeric respiratory complex III (CIII2) oxidizes ubiquinol to ubiquinone, transferring electrons to cytochrome c and translocating protons through a mechanism known as the Q cycle. The Q cycle involves ubiquinol oxidation and ubiquinone reduction at two different sites within each CIII monomer, as well as movement of the head domain of the Rieske subunit. We determined structures of Candida albicans CIII2 by cryoelectron microscopy (cryo-EM), revealing endogenous ubiquinone and visualizing the continuum of Rieske head domain conformations. Analysis of these conformations does not indicate cooperativity in the Rieske head domain position or ligand binding in the two CIIIs of the CIII2 dimer. Cryo-EM with the indazole derivative Inz-5, which inhibits fungal CIII2 and is fungicidal when administered with fungistatic azole drugs, showed that Inz-5 inhibition alters the equilibrium of Rieske head domain positions.


Subject(s)
Candida albicans/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Indazoles/pharmacology , Cryoelectron Microscopy , Electron Transport , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Indazoles/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Protein Multimerization , Ubiquinone/analogs & derivatives , Ubiquinone/chemistry
7.
Biochem J ; 478(17): 3253-3263, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34402504

ABSTRACT

The reaction centre light-harvesting 1 (RC-LH1) complex is the core functional component of bacterial photosynthesis. We determined the cryo-electron microscopy (cryo-EM) structure of the RC-LH1 complex from Rhodospirillum rubrum at 2.5 Šresolution, which reveals a unique monomeric bacteriochlorophyll with a phospholipid ligand in the gap between the RC and LH1 complexes. The LH1 complex comprises a circular array of 16 αß-polypeptide subunits that completely surrounds the RC, with a preferential binding site for a quinone, designated QP, on the inner face of the encircling LH1 complex. Quinols, initially generated at the RC QB site, are proposed to transiently occupy the QP site prior to traversing the LH1 barrier and diffusing to the cytochrome bc1 complex. Thus, the QP site, which is analogous to other such sites in recent cryo-EM structures of RC-LH1 complexes, likely reflects a general mechanism for exporting quinols from the RC-LH1 complex.


Subject(s)
Bacterial Proteins/chemistry , Cryoelectron Microscopy/methods , Light-Harvesting Protein Complexes/chemistry , Rhodospirillum rubrum/chemistry , Bacterial Proteins/isolation & purification , Bacteriochlorophylls/chemistry , Benzoquinones/chemistry , Binding Sites , Crystallization , Electron Transport Complex III/chemistry , Hydrogen Bonding , Hydroquinones/chemistry , Ligands , Light-Harvesting Protein Complexes/isolation & purification , Phospholipids/chemistry , Protein Conformation, alpha-Helical
8.
Biochemistry ; 60(46): 3497-3506, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34266238

ABSTRACT

Two major subclasses of mononuclear non-heme ferrous enzymes use two electron-donating organic cofactors (α-ketoglutarate or pterin) to activate O2 to form FeIV═O intermediates that further react with their substrates through hydrogen atom abstraction or electrophilic aromatic substitution. New spectroscopic methodologies have been developed, enabling the study of the active sites in these enzymes and their oxygen intermediates. Coupled to electronic structure calculations, the results of these spectroscopies provide fundamental insight into mechanism. This Perspective summarizes the results of these studies in elucidating the mechanism of dioxygen activation to form the FeIV═O intermediate and the geometric and electronic structure of this intermediate that enables its high reactivity and selectivity in product formation.


Subject(s)
Cysteine Dioxygenase/metabolism , Electron Transport Complex III/metabolism , Iron/metabolism , Ketoglutaric Acids/metabolism , Oxygen/metabolism , Catalytic Domain , Cysteine Dioxygenase/chemistry , Electron Transport Complex III/chemistry , Ketoglutaric Acids/chemistry , Pterins/metabolism , Superoxides/metabolism
9.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34259807

ABSTRACT

Long-lived proteins (LLPs) have recently emerged as vital components of intracellular structures whose function is coupled to long-term stability. Mitochondria are multifaceted organelles, and their function hinges on efficient proteome renewal and replacement. Here, using metabolic stable isotope labeling of mice combined with mass spectrometry (MS)-based proteomic analysis, we demonstrate remarkable longevity for a subset of the mitochondrial proteome. We discovered that mitochondrial LLPs (mt-LLPs) can persist for months in tissues harboring long-lived cells, such as brain and heart. Our analysis revealed enrichment of mt-LLPs within the inner mitochondrial membrane, specifically in the cristae subcompartment, and demonstrates that the mitochondrial proteome is not turned over in bulk. Pioneering cross-linking experiments revealed that mt-LLPs are spatially restricted and copreserved within protein OXPHOS complexes, with limited subunit exchange throughout their lifetimes. This study provides an explanation for the exceptional mitochondrial protein lifetimes and supports the concept that LLPs provide key structural stability to multiple large and dynamic intracellular structures.


Subject(s)
Electron Transport Complex III/metabolism , Electron Transport Complex II/metabolism , Electron Transport Complex IV/metabolism , Electron Transport Complex I/metabolism , Mitochondria/enzymology , Myocardium/enzymology , Proteome/metabolism , Animals , Binding Sites , Brain/enzymology , Citric Acid Cycle/genetics , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Electron Transport Complex II/chemistry , Electron Transport Complex II/genetics , Electron Transport Complex III/chemistry , Electron Transport Complex III/genetics , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/genetics , Gene Expression , Half-Life , Lipid Metabolism/genetics , Mice , Mitochondria/genetics , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/enzymology , Models, Molecular , Organ Specificity , Oxidative Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Stability , Proteome/chemistry , Proteome/genetics
10.
J Mol Cell Cardiol ; 161: 23-38, 2021 12.
Article in English | MEDLINE | ID: mdl-34331972

ABSTRACT

A serious consequence of myocardial ischemia-reperfusion injury (I/R) is oxidative damage, which causes mitochondrial dysfunction. The cascading ROS can propagate and potentially induce heme bleaching and protein cysteine sulfonation (PrSO3H) of the mitochondrial electron transport chain. Herein we studied the mechanism of I/R-mediated irreversible oxidative injury of complex III in mitochondria from rat hearts subjected to 30-min of ischemia and 24-h of reperfusion in vivo. In the I/R region, the catalytic activity of complex III was significantly impaired. Spectroscopic analysis indicated that I/R mediated the destruction of hemes b and c + c1 in the mitochondria, supporting I/R-mediated complex III impairment. However, no significant impairment of complex III activity and heme damage were observed in mitochondria from the risk region of rat hearts subjected only to 30-min ischemia, despite a decreased state 3 respiration. In the I/R mitochondria, carbamidomethylated C122/C125 of cytochrome c1 via alkylating complex III with a down regulation of HCCS was exclusively detected, supporting I/R-mediated thioether defect of heme c1. LC-MS/MS analysis showed that I/R mitochondria had intensely increased complex III PrSO3H levels at the C236 ligand of the [2Fe2S] cluster of the Rieske iron­sulfur protein (uqcrfs1), thus impairing the electron transport activity. MS analysis also indicated increased PrSO3H of the hinge protein at C65 and of cytochrome c1 at C140 and C220, which are confined in the intermembrane space. MS analysis also showed that I/R extensively enhanced the PrSO3H of the core 1 (uqcrc1) and core 2 (uqcrc2) subunits in the matrix compartment, thus supporting the conclusion that complex III releases ROS to both sides of the inner membrane during reperfusion. Analysis of ischemic mitochondria indicated a modest reduction from the basal level of complex III PrSO3H detected in the mitochondria of sham control hearts, suggesting that the physiologic hyperoxygenation and ROS overproduction during reperfusion mediated the enhancement of complex III PrSO3H. In conclusion, reperfusion-mediated heme damage with increased PrSO3H controls oxidative injury to complex III and aggravates mitochondrial dysfunction in the post-ischemic heart.


Subject(s)
Cysteine/metabolism , Electron Transport Complex III/metabolism , Heme/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/metabolism , Animals , Benzene Derivatives/chemistry , Cattle , Cysteine/chemistry , Cytochromes c1/chemistry , Cytochromes c1/metabolism , Electron Transport Complex III/chemistry , Heme/chemistry , Male , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Ischemia/metabolism , Peroxynitrous Acid/chemistry , Rats, Sprague-Dawley , Superoxide Dismutase/genetics
11.
Molecules ; 26(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299598

ABSTRACT

In this work we introduce a novel filtering and molecular modeling pipeline based on a fingerprint and descriptor similarity procedure, coupled with molecular docking and molecular dynamics (MD), to select potential novel quoinone outside inhibitors (QoI) of cytochrome bc1 with the aim of determining the same or different chromophores to usual. The study was carried out using the yeast cytochrome bc1 complex with its docked ligand (stigmatellin), using all the fungicides from FRAC code C3 mode of action, 8617 Drugbank compounds and 401,624 COCONUT compounds. The introduced drug repurposing pipeline consists of compound similarity with C3 fungicides and molecular docking (MD) simulations with final QM/MM binding energy determination, while aiming for potential novel chromophores and perserving at least an amide (R1HN(C=O)R2) or ester functional group of almost all up to date C3 fungicides. 3D descriptors used for a similarity test were based on the 280 most stable Padel descriptors. Hit compounds that passed fingerprint and 3D descriptor similarity condition and had either an amide or an ester group were submitted to docking where they further had to satisfy both Chemscore fitness and specific conformation constraints. This rigorous selection resulted in a very limited number of candidates that were forwarded to MD simulations and QM/MM binding affinity estimations by the ORCA DFT program. In this final step, stringent criteria based on (a) sufficiently high frequency of H-bonds; (b) high interaction energy between protein and ligand through the whole MD trajectory; and (c) high enough QM/MM binding energy scores were applied to further filter candidate inhibitors. This elaborate search pipeline led finaly to four Drugbank synthetic lead compounds (DrugBank) and seven natural (COCONUT database) lead compounds-tentative new inhibitors of cytochrome bc1. These eleven lead compounds were additionally validated through a comparison of MM/PBSA free binding energy for new leads against those obtatined for 19 QoIs.


Subject(s)
Electron Transport Complex III/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/enzymology , Drug Evaluation, Preclinical , Electron Transport Complex III/chemistry , Saccharomyces cerevisiae Proteins/chemistry
12.
Chem Rev ; 121(15): 9644-9673, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34184881

ABSTRACT

In the final steps of energy conservation in aerobic organisms, free energy from electron transfer through the respiratory chain is transduced into a proton electrochemical gradient across a membrane. In mitochondria and many bacteria, reduction of the dioxygen electron acceptor is catalyzed by cytochrome c oxidase (complex IV), which receives electrons from cytochrome bc1 (complex III), via membrane-bound or water-soluble cytochrome c. These complexes function independently, but in many organisms they associate to form supercomplexes. Here, we review the structural features and the functional significance of the nonobligate III2IV1/2 Saccharomyces cerevisiae mitochondrial supercomplex as well as the obligate III2IV2 supercomplex from actinobacteria. The analysis is centered around the Q-cycle of complex III, proton uptake by CytcO, as well as mechanistic and structural solutions to the electronic link between complexes III and IV.


Subject(s)
Cell Membrane/enzymology , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Saccharomyces cerevisiae , Electron Transport , Protons , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology
13.
Biochim Biophys Acta Bioenerg ; 1862(8): 148433, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33932366

ABSTRACT

Respiration is carried out by a series of membrane-bound complexes in the inner mitochondrial membrane or in the cytoplasmic membrane of bacteria. Increasing evidence shows that these complexes organize into larger supercomplexes. In this work, we identified a supercomplex composed of cytochrome (cyt.) bc1 and aa3-type cyt. c oxidase in Rhodobacter sphaeroides. We purified the supercomplex using a His-tag on either of these complexes. The results from activity assays, native and denaturing PAGE, size exclusion chromatography, electron microscopy, optical absorption spectroscopy and kinetic studies on the purified samples support the formation and coupled quinol oxidation:O2 reduction activity of the cyt. bc1-aa3 supercomplex. The potential role of the membrane-anchored cyt. cy as a component in supercomplexes was also investigated.


Subject(s)
Cell Membrane/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Hydroquinones/metabolism , Rhodobacter sphaeroides/enzymology , Electron Transport , Electron Transport Complex III/chemistry , Electron Transport Complex IV/chemistry , Kinetics , Oxidation-Reduction
14.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33876763

ABSTRACT

Complex II, also known as succinate dehydrogenase (SQR) or fumarate reductase (QFR), is an enzyme involved in both the Krebs cycle and oxidative phosphorylation. Mycobacterial Sdh1 has recently been identified as a new class of respiratory complex II (type F) but with an unknown electron transfer mechanism. Here, using cryoelectron microscopy, we have determined the structure of Mycobacterium smegmatis Sdh1 in the presence and absence of the substrate, ubiquinone-1, at 2.53-Å and 2.88-Å resolution, respectively. Sdh1 comprises three subunits, two that are water soluble, SdhA and SdhB, and one that is membrane spanning, SdhC. Within these subunits we identified a quinone-binding site and a rarely observed Rieske-type [2Fe-2S] cluster, the latter being embedded in the transmembrane region. A mutant, where two His ligands of the Rieske-type [2Fe-2S] were changed to alanine, abolished the quinone reduction activity of the Sdh1. Our structures allow the proposal of an electron transfer pathway that connects the substrate-binding and quinone-binding sites. Given the unique features of Sdh1 and its essential role in Mycobacteria, these structures will facilitate antituberculosis drug discovery efforts that specifically target this complex.


Subject(s)
Bacterial Proteins/chemistry , Electron Transport Complex III/chemistry , Flavoproteins/chemistry , Mycobacterium tuberculosis/enzymology , Bacterial Proteins/metabolism , Binding Sites , Cryoelectron Microscopy , Electron Transport Complex III/metabolism , Flavoproteins/metabolism , Molecular Dynamics Simulation , Protein Binding , Ubiquinone/chemistry , Ubiquinone/metabolism
15.
BMC Cancer ; 21(1): 427, 2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33865346

ABSTRACT

BACKGROUND: Associations between mitochondrial genetic abnormalities (variations and copy number, i.e. mtDNAcn, change) and elevated ROS have been reported in cancer compared to normal cells. Since excessive levels of ROS can trigger apoptosis, treating cancer cells with ROS-stimulating agents may enhance their death. This study aimed to investigate the link between baseline ROS levels and mitochondrial genetic abnormalities, and how mtDNA abnormalities might be used to predict cancer cells' response to ROS-stimulating therapy. METHODS: Intracellular and mitochondrial specific-ROS levels were measured using the DCFDA and MitoSOX probes, respectively, in four cancer and one non-cancerous cell lines. Cells were treated with ROS-stimulating agents (cisplatin and dequalinium) and the IC50s were determined using the MTS assay. Sanger sequencing and qPCR were conducted to screen the complete mitochondrial genome for variations and to relatively quantify mtDNAcn, respectively. Non-synonymous variations were subjected to 3-dimensional (3D) protein structural mapping and analysis. RESULTS: Our data revealed novel significant associations between the total number of variations in the mitochondrial respiratory chain (MRC) complex I and III genes, mtDNAcn, ROS levels, and ROS-associated drug response. Furthermore, functional variations in complexes I/III correlated significantly and positively with mtDNAcn, ROS levels and drug resistance, indicating they might mechanistically influence these parameters in cancer cells. CONCLUSIONS: Our findings suggest that mtDNAcn and complexes I/III functional variations have the potential to be efficient biomarkers to predict ROS-stimulating therapy efficacy in the future.


Subject(s)
Antineoplastic Agents/pharmacology , DNA, Mitochondrial , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemistry , Binding Sites , DNA Copy Number Variations , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , High-Throughput Nucleotide Sequencing , Humans , Models, Molecular , Molecular Conformation , Protein Binding , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836592

ABSTRACT

Energy conversion in aerobic organisms involves an electron current from low-potential donors, such as NADH and succinate, to dioxygen through the membrane-bound respiratory chain. Electron transfer is coupled to transmembrane proton transport, which maintains the electrochemical proton gradient used to produce ATP and drive other cellular processes. Electrons are transferred from respiratory complexes III to IV (CIII and CIV) by water-soluble cytochrome (cyt.) c In Saccharomyces cerevisiae and some other organisms, these complexes assemble into larger CIII2CIV1/2 supercomplexes, the functional significance of which has remained enigmatic. In this work, we measured the kinetics of the S. cerevisiae supercomplex cyt. c-mediated QH2:O2 oxidoreductase activity under various conditions. The data indicate that the electronic link between CIII and CIV is confined to the surface of the supercomplex. Single-particle electron cryomicroscopy (cryo-EM) structures of the supercomplex with cyt. c show the positively charged cyt. c bound to either CIII or CIV or along a continuum of intermediate positions. Collectively, the structural and kinetic data indicate that cyt. c travels along a negatively charged patch on the supercomplex surface. Thus, rather than enhancing electron transfer rates by decreasing the distance that cyt. c must diffuse in three dimensions, formation of the CIII2CIV1/2 supercomplex facilitates electron transfer by two-dimensional (2D) diffusion of cyt. c This mechanism enables the CIII2CIV1/2 supercomplex to increase QH2:O2 oxidoreductase activity and suggests a possible regulatory role for supercomplex formation in the respiratory chain.


Subject(s)
Cytochromes c/metabolism , Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Saccharomyces cerevisiae/metabolism , Cryoelectron Microscopy , Cytochromes c/chemistry , Electron Transport , Electron Transport Complex III/chemistry , Electron Transport Complex IV/chemistry , Kinetics , Mitochondria/metabolism , Models, Molecular , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
17.
Nat Commun ; 12(1): 929, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568648

ABSTRACT

Respiratory electron transport complexes are organized as individual entities or combined as large supercomplexes (SC). Gram-negative bacteria deploy a mitochondrial-like cytochrome (cyt) bc1 (Complex III, CIII2), and may have specific cbb3-type cyt c oxidases (Complex IV, CIV) instead of the canonical aa3-type CIV. Electron transfer between these complexes is mediated by soluble (c2) and membrane-anchored (cy) cyts. Here, we report the structure of an engineered bc1-cbb3 type SC (CIII2CIV, 5.2 Å resolution) and three conformers of native CIII2 (3.3 Å resolution). The SC is active in vivo and in vitro, contains all catalytic subunits and cofactors, and two extra transmembrane helices attributed to cyt cy and the assembly factor CcoH. The cyt cy is integral to SC, its cyt domain is mobile and it conveys electrons to CIV differently than cyt c2. The successful production of a native-like functional SC and determination of its structure illustrate the characteristics of membrane-confined and membrane-external respiratory electron transport pathways in Gram-negative bacteria.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Rhodobacter capsulatus/enzymology , Bacterial Proteins/genetics , Catalytic Domain , Coenzymes/chemistry , Coenzymes/metabolism , Cryoelectron Microscopy , Electron Transport , Electron Transport Complex III/genetics , Electron Transport Complex IV/genetics , Genetic Engineering , Rhodobacter capsulatus/chemistry , Rhodobacter capsulatus/genetics , Rhodobacter capsulatus/metabolism
18.
PLoS Pathog ; 17(2): e1009211, 2021 02.
Article in English | MEDLINE | ID: mdl-33524071

ABSTRACT

The mitochondrion is critical for the survival of apicomplexan parasites. Several major anti-parasitic drugs, such as atovaquone and endochin-like quinolones, act through inhibition of the mitochondrial electron transport chain at the coenzyme Q:cytochrome c oxidoreductase complex (Complex III). Despite being an important drug target, the protein composition of Complex III of apicomplexan parasites has not been elucidated. Here, we undertake a mass spectrometry-based proteomic analysis of Complex III in the apicomplexan Toxoplasma gondii. Along with canonical subunits that are conserved across eukaryotic evolution, we identify several novel or highly divergent Complex III components that are conserved within the apicomplexan lineage. We demonstrate that one such subunit, which we term TgQCR11, is critical for parasite proliferation, mitochondrial oxygen consumption and Complex III activity, and establish that loss of this protein leads to defects in Complex III integrity. We conclude that the protein composition of Complex III in apicomplexans differs from that of the mammalian hosts that these parasites infect.


Subject(s)
Electron Transport Complex III/metabolism , Toxoplasma/metabolism , Animals , Blotting, Western , Cells, Cultured , Electron Transport Complex III/chemistry , Fluorescent Antibody Technique , Humans , Mitochondria/metabolism , Oxygen/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Smegmamorpha , Toxoplasma/genetics
19.
Biochim Biophys Acta Bioenerg ; 1862(5): 148385, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33516769

ABSTRACT

Cytochrome bc1 complexes are energy-transducing enzymes and key components of respiratory electron chains. They contain Rieske 2Fe2S proteins that absorb very weakly in the visible absorption region compared to the heme cofactors of the cytochromes, but are known to yield photoproducts. Here, the photoreactions of isolated Rieske proteins from the hyperthermophilic bacterium Aquifex aeolicus are studied in two redox states using ultrafast transient fluorescence and absorption spectroscopy. We provide evidence, for the first time in iron­sulfur proteins, of very weak fluorescence of the excited state, in the oxidized as well as the reduced state. The excited states of the oxidized and reduced forms decay in 1.5 ps and 30 ps, respectively. In both cases they give rise to product states with lifetimes beyond 1 ns, reflecting photo-reduction of oxidized centers as well as photo-oxidation of reduced centers. Potential reaction partners are discussed and studied using site-directed mutagenesis. For the reduced state, a nearby disulfide bridge is suggested as an electron acceptor. The resulting photoproducts in either state may play a role in photoactivation processes.


Subject(s)
Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Fluorescence , Iron/metabolism , Photochemistry , Sulfur/metabolism , Aquifex/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Iron/chemistry , Oxidation-Reduction , Sulfur/chemistry
20.
Elife ; 102021 01 19.
Article in English | MEDLINE | ID: mdl-33463523

ABSTRACT

Mitochondrial complex III (CIII2) and complex IV (CIV), which can associate into a higher-order supercomplex (SC III2+IV), play key roles in respiration. However, structures of these plant complexes remain unknown. We present atomic models of CIII2, CIV, and SC III2+IV from Vigna radiata determined by single-particle cryoEM. The structures reveal plant-specific differences in the MPP domain of CIII2 and define the subunit composition of CIV. Conformational heterogeneity analysis of CIII2 revealed long-range, coordinated movements across the complex, as well as the motion of CIII2's iron-sulfur head domain. The CIV structure suggests that, in plants, proton translocation does not occur via the H channel. The supercomplex interface differs significantly from that in yeast and bacteria in its interacting subunits, angle of approach and limited interactions in the mitochondrial matrix. These structures challenge long-standing assumptions about the plant complexes and generate new mechanistic hypotheses.


Most living things including plants and animals use respiration to release energy from food. Respiration requires the activity of five large protein complexes typically called complex I, II, III, IV and V. Sometimes these complexes combine to form supercomplexes. The complexes are similar across plants, animals and other living things, but there are also many differences. Detailed structures of the respiratory complexes have been determined for many species of animals, fungi and bacteria, highlighting similarities and differences between organisms, and providing clues as to how respiration works. Yet, there is still a lot to learn about these complexes in plants. To bridge this gap, Maldonado et al. used a technique called cryo electron microscopy to study the structure of complexes III and IV and the supercomplex they form in the mung bean. This is the first study of the detailed structure of these two complexes in plants. The results showed many similarities to other species, as well as several features that are specific to plants. The way the two complexes interact to form a supercomplex is different than in other species, as are several other, smaller, structural features. Further examination of complex III revealed that it is flexible and that movements are coordinated across the length of the complex. Maldonado et al. speculate that this may allow it to coordinate its role in respiration with its other cellular roles. Understanding how plant respiratory complexes work could lead to improvements in crop yields or, since respiration is required for survival, result in the development of herbicides that block respiration in plants more effectively and specifically. Further researching the structure of the plant respiratory complexes and supercomplexes could also shed light on how plants adapt to different environments, including how they change to survive global warming.


Subject(s)
Electron Transport Complex III/chemistry , Electron Transport Complex IV/chemistry , Vigna/enzymology , Vigna/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...