Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
EMBO J ; 35(15): 1694-706, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27295975

ABSTRACT

Vacuolar ATPases (V-ATPases) are essential proton pumps that acidify the lumen of subcellular organelles in all eukaryotic cells and the extracellular space in some tissues. V-ATPase activity is regulated by a unique mechanism referred to as reversible disassembly, wherein the soluble catalytic sector, V1, is released from the membrane and its MgATPase activity silenced. The crystal structure of yeast V1 presented here shows that activity silencing involves a large conformational change of subunit H, with its C-terminal domain rotating ~150° from a position near the membrane in holo V-ATPase to a position at the bottom of V1 near an open catalytic site. Together with biochemical data, the structure supports a mechanistic model wherein subunit H inhibits ATPase activity by stabilizing an open catalytic site that results in tight binding of inhibitory ADP at another site.


Subject(s)
Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Vacuolar Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/metabolism , Catalytic Domain , Crystallography, X-Ray , Models, Biological , Models, Molecular , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
2.
Cell Mol Life Sci ; 76(20): 4023-4042, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31236625

ABSTRACT

Succinate dehydrogenase (SDH) also known as complex II or succinate:quinone oxidoreductase is an enzyme involved in both oxidative phosphorylation and tricarboxylic acid cycle; the processes that generate energy. SDH is a multi-subunit enzyme which requires a series of proteins for its proper assembly at several steps. This enzyme has medical significance as there is a broad range of human diseases from cancers to neurodegeneration related to SDH malfunction. Some of these disorders have recently been linked to defective assembly factors, reinvigorating further research in this area. Apart from that this enzyme has agricultural importance as many fungicides have been/will be designed targeting specifically this enzyme in plant fungal pathogens. In addition, we speculate it might be possible to design novel fungicides specifically targeting fungal assembly factors. Considering the medical and agricultural implications of SDH, the aim of this review is an overview of the SDH assembly factors and critical analysis of controversial issues around them.


Subject(s)
Mitochondria/enzymology , Neoplasms/enzymology , Neurodegenerative Diseases/enzymology , Protein Subunits/chemistry , Proteins/genetics , Succinate Dehydrogenase/chemistry , Animals , Citric Acid Cycle/genetics , Coenzymes/chemistry , Coenzymes/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungi , Fungicides, Industrial/chemistry , Fungicides, Industrial/pharmacology , Gene Expression , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondria/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Oxidative Phosphorylation , Plants , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
3.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198276

ABSTRACT

Nitric oxide (NO) is a well-known active site ligand and inhibitor of respiratory terminal oxidases. Here, we investigated the interaction of NO with a purified chimeric bcc-aa3 supercomplex composed of Mycobacterium tuberculosis cytochrome bcc and Mycobacterium smegmatisaa3-type terminal oxidase. Strikingly, we found that the enzyme in turnover with O2 and reductants is resistant to inhibition by the ligand, being able to metabolize NO at 25 °C with an apparent turnover number as high as ≈303 mol NO (mol enzyme)-1 min-1 at 30 µM NO. The rate of NO consumption proved to be proportional to that of O2 consumption, with 2.65 ± 0.19 molecules of NO being consumed per O2 molecule by the mycobacterial bcc-aa3. The enzyme was found to metabolize the ligand even under anaerobic reducing conditions with a turnover number of 2.8 ± 0.5 mol NO (mol enzyme)-1 min-1 at 25 °C and 8.4 µM NO. These results suggest a protective role of mycobacterial bcc-aa3 supercomplexes against NO stress.


Subject(s)
Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Nitric Oxide/pharmacology , Bacterial Proteins/metabolism , Catalysis , Catalytic Domain , Electron Transport , Free Radicals , Ligands , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Nitric Oxide/chemistry , Oxidoreductases/metabolism , Oxygen , Protein Binding
4.
J Biol Chem ; 290(23): 14350-60, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25861988

ABSTRACT

Recently, energy production pathways have been shown to be viable antitubercular drug targets to combat multidrug-resistant tuberculosis and eliminate pathogen in the dormant state. One family of drugs currently under development, the imidazo[1,2-a]pyridine derivatives, is believed to target the pathogen's homolog of the mitochondrial bc1 complex. This complex, denoted cytochrome bcc, is highly divergent from mitochondrial Complex III both in subunit structure and inhibitor sensitivity, making it a good target for drug development. There is no soluble cytochrome c in mycobacteria to transport electrons from the bcc complex to cytochrome oxidase. Instead, the bcc complex exists in a "supercomplex" with a cytochrome aa3-type cytochrome oxidase, presumably allowing direct electron transfer. We describe here purification and initial characterization of the mycobacterial cytochrome bcc-aa3 supercomplex using a strain of M. smegmatis that has been engineered to express the M. tuberculosis cytochrome bcc. The resulting hybrid supercomplex is stable during extraction and purification in the presence of dodecyl maltoside detergent. It is hoped that this purification procedure will potentiate functional studies of the complex as well as crystallographic studies of drug binding and provide structural insight into a third class of the bc complex superfamily.


Subject(s)
Bacterial Proteins/chemistry , Electron Transport Complex III/chemistry , Electron Transport Complex IV/chemistry , Mycobacterium Infections/microbiology , Mycobacterium smegmatis/chemistry , Mycobacterium tuberculosis/chemistry , Bacterial Proteins/isolation & purification , Electron Transport , Electron Transport Complex III/isolation & purification , Electron Transport Complex IV/isolation & purification , Humans
5.
Biochim Biophys Acta ; 1827(11-12): 1258-77, 2013.
Article in English | MEDLINE | ID: mdl-23624176

ABSTRACT

X-ray crystal structures of bc1 complexes obtained over the last 15 years have provided a firm structural basis for our understanding of the complex. For the most part there is good agreement between structures from different species, different crystal forms, and with different inhibitors bound. In this review we focus on some of the remaining unexplained differences, either between the structures themselves or the interpretations of the structural observations. These include the structural basis for the motion of the Rieske iron-sulfur protein in response to inhibitors, a possible conformational change involving tyrosine132 of cytochrome (cyt) b, the presence of cis-peptides at the beginnings of transmembrane helices C, E, and H, the structural insight into the function of the so-called "Core proteins", different modelings of the retained signal peptide, orientation of the low-potential heme b, and chirality of the Met ligand to heme c1. This article is part of a Special Issue entitled: Respiratory complex III and related bc complexes.


Subject(s)
Electron Transport Complex III/chemistry , Protein Conformation , Amino Acid Sequence , Animals , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Heme/chemistry , Heme/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Homology, Amino Acid
6.
J Membr Biol ; 247(9-10): 981-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24942818

ABSTRACT

Detergents classically are used to keep membrane proteins soluble in aqueous solutions, but they tend to destabilize them. This problem can be largely alleviated thanks to the use of amphipols (APols), small amphipathic polymers designed to substitute for detergents. APols adsorb at the surface of the transmembrane region of membrane proteins, keeping them water-soluble while stabilizing them bio-chemically. Membrane protein/APol complexes have proven, however, difficult to crystallize. In this study, the composition and solution properties of complexes formed between mitochondrial cytochrome bc1 and A8-35, the most extensively used APol to date, have been studied by means of size exclusion chromatography, sucrose gradient sedimentation, and small-angle neutron scattering. Stable, monodisperse preparations of bc1/A8-35 complexes can be obtained, which, depending on the medium, undergo either repulsive or attractive interactions. Under crystallization conditions, diffracting three-dimensional crystals of A8-35-stabilized cytochrome bc1 formed, but only in the concomitant presence of APol and detergent.


Subject(s)
Crystallization/methods , Detergents/chemistry , Electron Transport Complex III/chemistry , Electron Transport Complex III/ultrastructure , Polymers/chemistry , Propylamines/chemistry , Surface-Active Agents/chemistry , Hydrophobic and Hydrophilic Interactions , Protein Conformation , Protein Folding , Solubility , Solutions , Water/chemistry
7.
Article in English | MEDLINE | ID: mdl-23519794

ABSTRACT

The X-ray crystal structure of ribosome hibernation promoting factor (HPF) from Vibrio cholerae is presented at 2.0 Šresolution. The crystal was phased by two-wavelength MAD using cocrystallized cobalt. The asymmetric unit contained two molecules of HPF linked by four Co atoms. The metal-binding sites observed in the crystal are probably not related to biological function. The structure of HPF has a typical ß-α-ß-ß-ß-α fold consistent with previous structures of YfiA and HPF from Escherichia coli. Comparison of the new structure with that of HPF from E. coli bound to the Thermus thermophilus ribosome [Polikanov et al. (2012), Science, 336, 915-918] shows that no significant structural changes are induced in HPF by binding.


Subject(s)
Cobalt/chemistry , Escherichia coli Proteins/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Vibrio cholerae/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/isolation & purification , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Ribosomal Proteins/isolation & purification , Ribosomes/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermus thermophilus/chemistry , Thermus thermophilus/metabolism , Vibrio cholerae/metabolism
8.
Biochim Biophys Acta ; 1807(10): 1349-63, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21575592

ABSTRACT

The modified Q cycle mechanism accounts for the proton and charge translocation stoichiometry of the bc(1) complex, and is now widely accepted. However the mechanism by which the requisite bifurcation of electron flow at the Q(o) site reaction is enforced is not clear. One of several proposals involves conformational gating of the docking of the Rieske ISP at the Q(o) site, controlled by the stage of the reaction cycle. Effects of different Q(o)-site inhibitors on the position of the ISP seen in crystals may reflect the same conformational mechanism, in which case understanding how different inhibitors control the position of the ISP may be a key to understanding the enforcement of bifurcation at the Q(o) site (Table 1). Here we examine the available structures of cytochrome bc(1) with different Q(o)-site inhibitors and different ISP positions to look for clues to this mechanism. The effect of ISP removal on binding affinity of the inhibitors stigmatellin and famoxadone suggest a "mutual stabilization" of inhibitor binding and ISP docking, however this thermodynamic observation sheds little light on the mechanism. The cd(1) helix of cytochrome b moves in such a way as to accommodate docking when inhibitors favoring docking are bound, but it is impossible with the current structures to say whether this movement of α-cd(1) is a cause or result of ISP docking. One component of the movement of the linker between E and F helices also correlates with the type of inhibitor and ISP position, and seems to be related to the H-bonding pattern of Y279 of cytochrome b. An H-bond from Y279 to the ISP, and its possible modulation by movement of F275 in the presence of famoxadone and related inhibitors, or its competition with an alternate H-bond to I269 of cytochrome b that may be destabilized by bound famoxadone, suggest other possible mechanisms. This article is part of a Special Issue entitled: Allosteric cooperativity in respiratory proteins.


Subject(s)
Electron Transport Complex III/chemistry , Enzyme Inhibitors/chemistry , Protein Conformation , Ubiquinone/chemistry , Binding Sites/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , Cytochromes/chemistry , Cytochromes/metabolism , Cytochromes c/chemistry , Cytochromes c/metabolism , Databases, Protein , Electron Spin Resonance Spectroscopy , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Enzyme Inhibitors/metabolism , Hydrogen Bonding , Methacrylates/chemistry , Methacrylates/metabolism , Methacrylates/pharmacology , Models, Molecular , Molecular Structure , Mutation , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Polyenes/chemistry , Polyenes/metabolism , Polyenes/pharmacology , Principal Component Analysis , Protein Binding , Protein Structure, Secondary , Strobilurins , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism , Ubiquinone/metabolism
9.
J Am Chem Soc ; 134(27): 11168-76, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22690928

ABSTRACT

A critical challenge to the fragment-based drug discovery (FBDD) is its low-throughput nature due to the necessity of biophysical method-based fragment screening. Herein, a method of pharmacophore-linked fragment virtual screening (PFVS) was successfully developed. Its application yielded the first picomolar-range Q(o) site inhibitors of the cytochrome bc(1) complex, an important membrane protein for drug and fungicide discovery. Compared with the original hit compound 4 (K(i) = 881.80 nM, porcine bc(1)), the most potent compound 4f displayed 20 507-fold improved binding affinity (K(i) = 43.00 pM). Compound 4f was proved to be a noncompetitive inhibitor with respect to the substrate cytochrome c, but a competitive inhibitor with respect to the substrate ubiquinol. Additionally, we determined the crystal structure of compound 4e (K(i) = 83.00 pM) bound to the chicken bc(1) at 2.70 Å resolution, providing a molecular basis for understanding its ultrapotency. To our knowledge, this study is the first application of the FBDD method in the discovery of picomolar inhibitors of a membrane protein. This work demonstrates that the novel PFVS approach is a high-throughput drug discovery method, independent of biophysical screening techniques.


Subject(s)
Drug Design , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Chickens , Crystallography, X-Ray , Electron Transport Complex III/metabolism , Models, Molecular , Protein Binding , Swine , Thermodynamics
10.
Biochim Biophys Acta ; 1797(3): 360-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20025846

ABSTRACT

Ascochlorin is an isoprenoid antibiotic that is produced by the phytopathogenic fungus Ascochyta viciae. Similar to ascofuranone, which specifically inhibits trypanosome alternative oxidase by acting at the ubiquinol binding domain, ascochlorin is also structurally related to ubiquinol. When added to the mitochondrial preparations isolated from rat liver, or the yeast Pichia (Hansenula) anomala, ascochlorin inhibited the electron transport via CoQ in a fashion comparable to antimycin A and stigmatellin, indicating that this antibiotic acted on the cytochrome bc(1) complex. In contrast to ascochlorin, ascofuranone had much less inhibition on the same activities. On the one hand, like the Q(i) site inhibitors antimycin A and funiculosin, ascochlorin induced in H. anomala the expression of nuclear-encoded alternative oxidase gene much more strongly than the Q(o) site inhibitors tested. On the other hand, it suppressed the reduction of cytochrome b and the generation of superoxide anion in the presence of antimycin A(3) in a fashion similar to the Q(o) site inhibitor myxothiazol. These results suggested that ascochlorin might act at both the Q(i) and the Q(o) sites of the fungal cytochrome bc(1) complex. Indeed, the altered electron paramagnetic resonance (EPR) lineshape of the Rieske iron-sulfur protein, and the light-induced, time-resolved cytochrome b and c reduction kinetics of Rhodobacter capsulatus cytochrome bc(1) complex in the presence of ascochlorin demonstrated that this inhibitor can bind to both the Q(o) and Q(i) sites of the bacterial enzyme. Additional experiments using purified bovine cytochrome bc(1) complex showed that ascochlorin inhibits reduction of cytochrome b by ubiquinone through both Q(i) and Q(o) sites. Moreover, crystal structure of chicken cytochrome bc(1) complex treated with excess ascochlorin revealed clear electron densities that could be attributed to ascochlorin bound at both the Q(i) and Q(o) sites. Overall findings clearly show that ascochlorin is an unusual cytochrome bc(1) inhibitor that acts at both of the active sites of this enzyme.


Subject(s)
Alkenes/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Mitochondria, Liver/enzymology , Phenols/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antimycin A/pharmacology , Catalytic Domain , Cattle , Chickens , Crystallography, X-Ray , Cytochromes b/metabolism , Cytochromes c/metabolism , Electron Spin Resonance Spectroscopy , Electron Transport , Electron Transport Complex III/metabolism , Male , Mitochondrial Proteins , Oxidoreductases/metabolism , Pichia/metabolism , Plant Proteins , Polyenes/pharmacology , Rats , Rats, Wistar , Respiration , Rhodobacter capsulatus/metabolism , Superoxides/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
11.
Biochim Biophys Acta Proteins Proteom ; 1869(9): 140679, 2021 09.
Article in English | MEDLINE | ID: mdl-34089891

ABSTRACT

The quinone binding site (Q-site) of Mitochondrial Complex II (succinate-ubiquinone oxidoreductase) is the target for a number of inhibitors useful for elucidating the mechanism of the enzyme. Some of these have been developed as fungicides or pesticides, and species-specific Q-site inhibitors may be useful against human pathogens. We report structures of chicken Complex II with six different Q-site inhibitors bound, at resolutions 2.0-2.4 Å. These structures show the common interactions between the inhibitors and their binding site. In every case a carbonyl or hydroxyl oxygen of the inhibitor is H-bonded to Tyr58 in subunit SdhD and Trp173 in subunit SdhB. Two of the inhibitors H-bond Ser39 in subunit SdhC directly, while two others do so via a water molecule. There is a distinct cavity that accepts the 2-substituent of the carboxylate ring in flutolanil and related inhibitors. A hydrophobic "tail pocket" opens to receive a side-chain of intermediate-length inhibitors. Shorter inhibitors fit entirely within the main binding cleft, while the long hydrophobic side chains of ferulenol and atpenin A5 protrude out of the cleft into the bulk lipid region, as presumably does that of ubiquinone. Comparison of mitochondrial and Escherichia coli Complex II shows a rotation of the membrane-anchor subunits by 7° relative to the iron­sulfur protein. This rotation alters the geometry of the Q-site and the H-bonding pattern of SdhB:His216 and SdhD:Asp57. This conformational difference, rather than any active-site mutation, may be responsible for the different inhibitor sensitivity of the bacterial enzyme.


Subject(s)
Electron Transport Complex II/antagonists & inhibitors , Electron Transport Complex II/ultrastructure , Ubiquinone/ultrastructure , Amino Acid Sequence/genetics , Animals , Benzoquinones , Binding Sites , Chickens/genetics , Electron Transport Complex II/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutagenesis, Site-Directed , Quinones/chemistry , Sequence Alignment , Sus scrofa/genetics , Ubiquinone/chemistry
12.
Bioorg Med Chem ; 16(24): 10345-55, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18996700

ABSTRACT

Extensive molecular modeling based on crystallographic data was used to aid the design of synthetic analogues of the fungicidal naturally occurring respiration inhibitors crocacins A and D, and an inhibitor binding model to the mammalian cytochrome bc(1) complex was constructed. Simplified analogues were made which showed high activity in a mitochondrial beef heart respiration assay, and which were also active against certain plant pathogens in glasshouse tests. A crystal structure was obtained of an analogue of crocacin D bound to the chicken heart cytochrome bc(1) complex, which validated the binding model and which confirmed that the crocacins are a new class of inhibitor of the cytochrome bc(1) complex.


Subject(s)
Alkenes/chemistry , Amides/chemistry , Antifungal Agents/chemistry , Electron Transport Complex III/chemistry , Models, Molecular , Antifungal Agents/isolation & purification , Antifungal Agents/pharmacology , Computer Simulation , Crystallography, X-Ray , Drug Design , Electron Spin Resonance Spectroscopy , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Inhibitory Concentration 50 , Mitochondria/drug effects , Models, Chemical , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
13.
Biochim Biophys Acta ; 1757(9-10): 1073-83, 2006.
Article in English | MEDLINE | ID: mdl-16935256

ABSTRACT

Mitochondrial Complex II (succinate:ubiquinone oxidoreductase) is purified in a partially inactivated state, which can be activated by removal of tightly bound oxaloacetate (E.B. Kearney, et al., Biochem. Biophys. Res. Commun. 49 1115-1121). We crystallized Complex II in the presence of oxaloacetate or with the endogenous inhibitor bound. The structure showed a ligand essentially identical to the "malate-like intermediate" found in Shewanella Flavocytochrome c crystallized with fumarate (P. Taylor, et al., Nat. Struct. Biol. 6 1108-1112) Crystallization of Complex II in the presence of excess fumarate also gave the malate-like intermediate or a mixture of that and fumarate at the active site. In order to more conveniently monitor the occupation state of the dicarboxylate site, we are developing a library of UV/Vis spectral effects induced by binding different ligands to the site. Treatment with fumarate results in rapid development of the fumarate difference spectrum and then a very slow conversion into a species spectrally similar to the OAA-liganded complex. Complex II is known to be capable of oxidizing malate to the enol form of oxaloacetate (Y.O. Belikova, et al., Biochim. Biophys. Acta 936 1-9). The observations above suggest it may also be capable of interconverting fumarate and malate. It may be useful for understanding the mechanism and regulation of the enzyme to identify the malate-like intermediate and its pathway of formation from oxaloacetate or fumarate.


Subject(s)
Electron Transport Complex II/chemistry , Electron Transport Complex II/metabolism , Oxaloacetic Acid/pharmacology , Animals , Binding Sites , Chickens , Crystallography, X-Ray , Electron Transport Complex II/antagonists & inhibitors , Fumarates/metabolism , Iron-Sulfur Proteins/metabolism , Ligands , Malates/metabolism , Malonates/metabolism , Oxaloacetic Acid/metabolism , Protein Structure, Secondary , Spectrophotometry, Ultraviolet , Time Factors
14.
J Mol Biol ; 351(3): 573-97, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-16024040

ABSTRACT

Antimycin A (antimycin), one of the first known and most potent inhibitors of the mitochondrial respiratory chain, binds to the quinone reduction site of the cytochrome bc1 complex. Structure-activity relationship studies have shown that the N-formylamino-salicyl-amide group is responsible for most of the binding specificity, and suggested that a low pKa for the phenolic OH group and an intramolecular H-bond between that OH and the carbonyl O of the salicylamide linkage are important. Two previous X-ray structures of antimycin bound to vertebrate bc1 complex gave conflicting results. A new structure reported here of the bovine mitochondrial bc1 complex at 2.28 A resolution with antimycin bound, allows us for the first time to reliably describe the binding of antimycin and shows that the intramolecular hydrogen bond described in solution and in the small-molecule structure is replaced by one involving the NH rather than carbonyl O of the amide linkage, with rotation of the amide group relative to the aromatic ring. The phenolic OH and formylamino N form H-bonds with conserved Asp228 of cytochrome b, and the formylamino O H-bonds via a water molecule to Lys227. A strong density, the right size and shape for a diatomic molecule is found between the other side of the dilactone ring and the alphaA helix.


Subject(s)
Antimycin A/analogs & derivatives , Electron Transport Complex III/metabolism , Mitochondria/enzymology , Animals , Antimycin A/metabolism , Antimycin A/pharmacology , Cattle , Electron Transport/drug effects , Heme/metabolism , Ubiquinone/metabolism , X-Ray Diffraction
15.
Mol Cells ; 39(6): 495-500, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27215198

ABSTRACT

We have solved the crystal structure of a predicted fructose-specific enzyme IIB(fruc) from Escherichia coli (EcEIIB(fruc)) involved in the phosphoenolpyruvate-carbohydrate phosphotransferase system transferring carbohydrates across the cytoplasmic membrane. EcEIIB(fruc) belongs to a sequence family with more than 5,000 sequence homologues with 25-99% amino-acid sequence identity. It reveals a conventional Rossmann-like α-ß-α sandwich fold with a unique ß-sheet topology. Its C-terminus is longer than its closest relatives and forms an additional ß-strand whereas the shorter C-terminus is random coil in the relatives. Interestingly, its core structure is similar to that of enzyme IIB(cellobiose) from E. coli (EcIIB(cel)) transferring a phosphate moiety. In the active site of the closest EcEIIB(fruc) homologues, a unique motif CXXGXAHT comprising a P-loop like architecture including a histidine residue is found. The conserved cysteine on this loop may be deprotonated to act as a nucleophile similar to that of EcIIB(cel). The conserved histidine residue is presumed to bind the negatively charged phosphate. Therefore, we propose that the catalytic mechanism of EcEIIB(fruc) is similar to that of EcIIB(cel) transferring phosphoryl moiety to a specific carbohydrate.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Fructose/metabolism , Phosphoenolpyruvate Sugar Phosphotransferase System/chemistry , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Cysteine/metabolism , Escherichia coli/chemistry , Models, Molecular , Protein Folding , Protein Structure, Secondary , Substrate Specificity
16.
FEBS Lett ; 555(1): 13-20, 2003 Nov 27.
Article in English | MEDLINE | ID: mdl-14630312

ABSTRACT

A direct hydrogen bond between ubiquinone/quinol bound at the QO site and a cluster-ligand histidine of the iron-sulfur protein (ISP) is described as a major determining factor explaining much experimental data on position of the ISP ectodomain, electron paramagnetic resonance (EPR) lineshape and midpoint potential of the iron-sulfur cluster, and the mechanism of the bifurcated electron transfer from ubiquinol to the high and low potential chains of the bc1 complex.


Subject(s)
Electron Transport Complex III/chemistry , Animals , Binding Sites , Cattle , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Electron Transport , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Histidine/chemistry , Hydrogen Bonding , Hydroquinones/chemistry , In Vitro Techniques , Models, Molecular , Oxidation-Reduction , Protein Conformation , Static Electricity
17.
Photosynth Res ; 81(3): 251-75, 2004.
Article in English | MEDLINE | ID: mdl-16034531

ABSTRACT

Ubihydroquinone: cytochrome (cyt)c oxidoreductase, or cyt bc (1), is a widespread, membrane integral enzyme that plays a crucial role during photosynthesis and respiration. It is one of the major contributors of the electrochemical proton gradient, which is subsequently used for ATP synthesis. The simplest form of the cyt bc (1) is found in bacteria, and it contains only the three ubiquitously conserved catalytic subunits: the Fe-S protein, cyt b and cyt c (1). Here we present a preliminary X-ray structure of Rhodobacter capsulatus cyt bc (1) at 3.8 A and compare it to the available structures of its homologues from mitochondria and chloroplast. Using the bacterial enzyme structure, we highlight the structural similarities and differences that are found among the three catalytic subunits between the members of this family of enzymes. In addition, we discuss the locations of currently known critical mutations, and their implications in terms of the cyt bc (1) catalysis.

18.
Structure ; 20(11): 1881-92, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23000382

ABSTRACT

Vacuolar ATPases (V-ATPases) are multisubunit rotary motor proton pumps that function to acidify subcellular organelles in all eukaryotic organisms. V-ATPase is regulated by a unique mechanism that involves reversible dissociation into V1-ATPase and V0 proton channel, a process that involves breaking of protein interactions mediated by subunit C, the cytoplasmic domain of subunit "a" and three "peripheral stalks," each made of a heterodimer of E and G subunits. Here, we present crystal structures of a yeast V-ATPase heterotrimeric complex composed of EG heterodimer and the head domain of subunit C (C(head)). The structures show EG heterodimer folded in a noncanonical coiled coil that is stabilized at its N-terminal ends by binding to C(head). The coiled coil is disrupted by a bulge of partially unfolded secondary structure in subunit G and we speculate that this unique feature in the eukaryotic V-ATPase peripheral stalk may play an important role in enzyme structure and regulation by reversible dissociation.


Subject(s)
Saccharomyces cerevisiae/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Conformation , Vacuolar Proton-Translocating ATPases/chemistry
19.
J Mol Biol ; 416(4): 495-502, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22245575

ABSTRACT

Domain swapping is a mechanism for forming protein dimers and oligomers with high specificity. It is distinct from other forms of oligomerization in that the binding interface is formed by reciprocal exchange of polypeptide segments. Swapping plays a physiological role in protein-protein recognition, and it can also potentially be exploited as a mechanism for controlled self-assembly. Here, we demonstrate that domain-swapped interfaces can be engineered by inserting one protein into a surface loop of another protein. The key to facilitating a domain swap is to destabilize the protein when it is monomeric but not when it is oligomeric. We achieve this condition by employing the "mutually exclusive folding" design to apply conformational stress to the monomeric state. Ubiquitin (Ub) is inserted into one of six surface loops of barnase (Bn). The 38-Å amino-to-carboxy-terminal distance of Ub stresses the Bn monomer, causing it to split at the point of insertion. The 2.2-Å X-ray structure of one insertion variant reveals that strain is relieved by intermolecular folding with an identically unfolded Bn domain, resulting in a domain-swapped polymer. All six constructs oligomerize, suggesting that inserting Ub into each surface loop of Bn results in a similar domain-swapping event. Binding affinity can be tuned by varying the length of the peptide linkers used to join the two proteins, which modulates the extent of stress. Engineered, swapped proteins have the potential to be used to fabricate "smart" biomaterials, or as binding modules from which to assemble heterologous, multi-subunit protein complexes.


Subject(s)
Protein Binding , Protein Engineering/methods , Protein Structure, Tertiary , Amino Acid Sequence , Bacterial Proteins , Computer Simulation , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Ribonucleases/chemistry , Ubiquitin/chemistry
20.
J Biol Inorg Chem ; 13(4): 481-98, 2008 May.
Article in English | MEDLINE | ID: mdl-18418633

ABSTRACT

Early investigation of the electron paramagnetic resonance spectra of bis-histidine-coordinated membrane-bound ferriheme proteins led to the description of a spectral signal that had only one resolved feature. These became known as "highly anisotropic low-spin" or "large g(max)" ferriheme centers. Extensive work with small-molecule model heme complexes showed that this spectroscopic signature occurs in bis-imidazole ferrihemes in which the planes of the imidazole ligands are nearly perpendicular, deltaphi = 57-90 degrees. In the last decade protein crystallographic studies have revealed the atomic structures of a number of examples of bis-histidine heme proteins. A frequent characteristic of these large g(max) ferrihemes in membrane-bound proteins is the occurrence of the heme within a four-helix bundle with a left-handed twist. The histidine ligands occur at the same level on two diametrically opposed helices of the bundle. These ligands have the same side-chain conformation and ligate heme iron on the bundle axis, resulting in a quasi-twofold symmetric structure. The two non-ligand-bearing helices also obey this symmetry, and have a conserved small residue, usually glycine, where the edge of the heme ring makes contact with the helix backbones. In many cases this small residue is preceded by a threonine or serine residue whose side-chain hydroxyl oxygen acts as a hydrogen-bond acceptor from the N(delta1) atom of the heme-ligating histidine. The deltaphi angle is thus determined by the common histidine side-chain conformation and the crossing angle of the ligand-bearing helices, in some cases constrained by hydrogen bonds to the serine/threonine residues on the non-ligand-bearing helices.


Subject(s)
Cell Membrane/enzymology , Cytochromes/chemistry , Cytochromes/metabolism , Heme/chemistry , Heme/metabolism , Imidazoles/chemistry , Mitochondria/enzymology , Cell Membrane/chemistry , Histidine/chemistry , Histidine/metabolism , Mitochondria/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL