Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791189

ABSTRACT

The membrane Fo factor of ATP synthase is highly sensitive to mutations in the proton half-channel leading to the functional blocking of the entire protein. To identify functionally important amino acids for the proton transport, we performed molecular dynamic simulations on the selected mutants of the membrane part of the bacterial FoF1-ATP synthase embedded in a native lipid bilayer: there were nine different mutations of a-subunit residues (aE219, aH245, aN214, aQ252) in the inlet half-channel. The structure proved to be stable to these mutations, although some of them (aH245Y and aQ252L) resulted in minor conformational changes. aH245 and aN214 were crucial for proton transport as they directly facilitated H+ transfer. The substitutions with nonpolar amino acids disrupted the transfer chain and water molecules or neighboring polar side chains could not replace them effectively. aE219 and aQ252 appeared not to be determinative for proton translocation, since an alternative pathway involving a chain of water molecules could compensate the ability of H+ transmembrane movement when they were substituted. Thus, mutations of conserved polar residues significantly affected hydration levels, leading to drastic changes in the occupancy and capacity of the structural water molecule clusters (W1-W3), up to their complete disappearance and consequently to the proton transfer chain disruption.


Subject(s)
Bacterial Proton-Translocating ATPases , Molecular Dynamics Simulation , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Mutation , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/genetics
2.
Elife ; 112022 02 02.
Article in English | MEDLINE | ID: mdl-35107420

ABSTRACT

In FoF1-ATP synthase, proton translocation through Fo drives rotation of the c-subunit oligomeric ring relative to the a-subunit. Recent studies suggest that in each step of the rotation, key glutamic acid residues in different c-subunits contribute to proton release to and proton uptake from the a-subunit. However, no studies have demonstrated cooperativity among c-subunits toward FoF1-ATP synthase activity. Here, we addressed this using Bacillus PS3 ATP synthase harboring a c-ring with various combinations of wild-type and cE56D, enabled by genetically fused single-chain c-ring. ATP synthesis and proton pump activities were decreased by a single cE56D mutation and further decreased by double cE56D mutations. Moreover, activity further decreased as the two mutation sites were separated, indicating cooperation among c-subunits. Similar results were obtained for proton transfer-coupled molecular simulations. The simulations revealed that prolonged proton uptake in mutated c-subunits is shared between two c-subunits, explaining the cooperation observed in biochemical assays.


Cells need to be able to store and transfer energy to fuel their various activities. To do this, they produce a small molecule called ATP to carry the energy, which is then released when the ATP is broken down. An enzyme found in plants, animals and bacteria, called FoF1 ATP synthase, can both create and use ATP. When it does this, protons, or positive hydrogen ions, are transported across cellular boundaries called membranes. The region of the enzyme that is responsible for pumping the protons contains different parts known as the c-ring and the a-subunit. The movement of protons drives the c-ring to rotate relative to the a-subunit, which leads to producing ATP. Previous research using simulations and the protein structures found there are two or three neighbouring amino acids in the c-ring that face the a-subunit, suggesting that these amino acids act together to drive the rotation. To test this hypothesis, Mitome et al. mutated these amino acids to examine the effect on the enzyme's ability to produce ATP. A single mutation reduced the production of ATP, which decreased even further with mutations in two of the amino acids. The extent of this decrease depended on the distance between the two mutations in the c-ring. Simulations of these changes also found similar results. This indicates there is coordination between different parts of the c-ring to increase the rate of ATP production. This study offers new insights into the molecular processes controlling ATP synthesis and confirms previous theoretical research. This will interest specialists in bioenergetics because it addresses a fundamental biological question with broad impact.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Protons , Bacillus , Bacterial Proton-Translocating ATPases/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Molecular Dynamics Simulation , Mutation , Protein Conformation
3.
Sci Rep ; 11(1): 21234, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707181

ABSTRACT

Membrane bound nicotinamide nucleotide transhydrogenase (TH) catalyses the hydride transfer from NADH to NADP+. Under physiological conditions, this reaction is endergonic and must be energized by the pmf, coupled to transmembrane proton transport. Recent structures of transhydrogenase holoenzymes suggest new mechanistic details, how the long-distance coupling between hydride transfer in the peripheral nucleotide binding sites and the membrane-localized proton transfer occurs that now must be tested experimentally. Here, we provide protocols for the efficient expression and purification of the Escherichia coli transhydrogenase and its reconstitution into liposomes, alone or together with the Escherichia coli F1F0 ATP synthase. We show that E. coli transhydrogenase is a reversible enzyme that can also work as a NADPH-driven proton pump. In liposomes containing both enzymes, NADPH driven H+-transport by TH is sufficient to instantly fuel ATP synthesis, which adds TH to the pool of pmf generating enzymes. If the same liposomes are energized with ATP, NADPH production by TH is stimulated > sixfold both by a pH gradient or a membrane potential. The presented protocols and results reinforce the tight coupling between hydride transfer in the peripheral nucleotide binding sites and transmembrane proton transport and provide powerful tools to investigate their coupling mechanism.


Subject(s)
Bacterial Proton-Translocating ATPases/metabolism , Energy Transfer , Escherichia coli Proteins/metabolism , NADP Transhydrogenases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proton-Translocating ATPases/chemistry , Escherichia coli Proteins/chemistry , Ion Transport , Liposomes/metabolism , NADP Transhydrogenases/chemistry
4.
Nat Commun ; 12(1): 4690, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344897

ABSTRACT

F1Fo ATP synthase interchanges phosphate transfer energy and proton motive force via a rotary catalysis mechanism. Isolated F1-ATPase catalytic cores can hydrolyze ATP, passing through six intermediate conformational states to generate rotation of their central γ-subunit. Although previous structural studies have contributed greatly to understanding rotary catalysis in the F1-ATPase, the structure of an important conformational state (the binding-dwell) has remained elusive. Here, we exploit temperature and time-resolved cryo-electron microscopy to determine the structure of the binding- and catalytic-dwell states of Bacillus PS3 F1-ATPase. Each state shows three catalytic ß-subunits in different conformations, establishing the complete set of six states taken up during the catalytic cycle and providing molecular details for both the ATP binding and hydrolysis strokes. We also identify a potential phosphate-release tunnel that indicates how ADP and phosphate binding are coordinated during synthesis. Overall these findings provide a structural basis for the entire F1-ATPase catalytic cycle.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Bacillus/enzymology , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Binding Sites , Catalysis , Cryoelectron Microscopy , Hydrolysis , Mutation , Phosphates/chemistry , Phosphates/metabolism , Protein Binding , Protein Conformation , Protein Subunits , Rotation , Temperature
5.
Acta Crystallogr D Struct Biol ; 76(Pt 4): 332-339, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32254057

ABSTRACT

Confidence maps provide complementary information for interpreting cryo-EM densities as they indicate statistical significance with respect to background noise. They can be thresholded by specifying the expected false-discovery rate (FDR), and the displayed volume shows the parts of the map that have the corresponding level of significance. Here, the basic statistical concepts of confidence maps are reviewed and practical guidance is provided for their interpretation and usage inside the CCP-EM suite. Limitations of the approach are discussed and extensions towards other error criteria such as the family-wise error rate are presented. The observed map features can be rendered at a common isosurface threshold, which is particularly beneficial for the interpretation of weak and noisy densities. In the current article, a practical guide is provided to the recommended usage of confidence maps.


Subject(s)
Cryoelectron Microscopy/methods , Bacterial Proton-Translocating ATPases/chemistry , Computer Graphics , Models, Molecular , Protein Conformation , Ribosomes/chemistry , Static Electricity , Statistics as Topic , Tobacco Mosaic Virus , User-Computer Interface
6.
Proc Natl Acad Sci U S A ; 114(43): 11291-11296, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073046

ABSTRACT

ATP synthase is a rotating membrane protein that synthesizes ATP through proton-pumping activity across the membrane. To unveil the mechanical impact of this molecular active pump on the bending properties of its lipid environment, we have functionally reconstituted the ATP synthase in giant unilamellar vesicles and tracked the membrane fluctuations by means of flickering spectroscopy. We find that ATP synthase rotates at a frequency of about 20 Hz, promoting large nonequilibrium deformations at discrete hot spots in lipid vesicles and thus inducing an overall membrane softening. The enhanced nonequilibrium fluctuations are compatible with an accumulation of active proteins at highly curved membrane sites through a curvature-protein coupling mechanism that supports the emergence of collective effects of rotating ATP synthases in lipid membranes.


Subject(s)
Bacterial Proton-Translocating ATPases/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Adenosine Triphosphate/biosynthesis , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/genetics , Cell Membrane/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Microscopy, Video , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rhodamine 123/chemistry , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism , Valinomycin/pharmacology
7.
J Bioenerg Biomembr ; 49(2): 171-181, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28078625

ABSTRACT

Subunit a is a membrane-bound stator subunit of the ATP synthase and is essential for proton translocation. The N-terminus of subunit a in E. coli is localized to the periplasm, and contains a sequence motif that is conserved among some bacteria. Previous work has identified mutations in this region that impair enzyme activity. Here, an internal deletion was constructed in subunit a in which residues 6-20 were replaced by a single lysine residue, and this mutant was unable to grow on succinate minimal medium. Membrane vesicles prepared from this mutant lacked ATP synthesis and ATP-driven proton translocation, even though immunoblots showed a significant level of subunit a. Similar results were obtained after purification and reconstitution of the mutant ATP synthase into liposomes. The location of subunit a with respect to its neighboring subunits b and c was probed by introducing cysteine substitutions that were known to promote cross-linking: a_L207C + c_I55C, a_L121C + b_N4C, and a_T107C + b_V18C. The last pair was unable to form cross-links in the background of the deletion mutant. The results indicate that loss of the N-terminal region of subunit a does not generally disrupt its structure, but does alter interactions with subunit b.


Subject(s)
Bacterial Proton-Translocating ATPases/genetics , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Sequence Deletion , Adenosine Triphosphate/biosynthesis , Amino Acid Sequence , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Cysteine/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ion Transport , Mutagenesis, Site-Directed , Protein Subunits/genetics , Protein Subunits/metabolism
8.
Biochem Biophys Res Commun ; 482(4): 922-927, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27890618

ABSTRACT

F1Fo-ATP synthase is a multisubunit enzyme responsible for the synthesis of ATP. Among its multiple subunits (8 in E. coli, 17 in yeast S. cerevisiae, 16 in vertebrates), two subunits a and c are known to play a central role controlling the H+ flow through the inner mitochondrial membrane which allows the subsequent synthesis of ATP, but the pathway followed by H+ within the two proteins is still a matter of debate. In fact, even though the structure of ATP synthase is now well defined, the molecular mechanisms determining the function of both F1 and FO domains are still largely unknown. In this study, we propose a pathway for proton migration along the ATP synthase by hydrogen-bonded chain mechanism, with a key role of serine and threonine residues, by X-ray diffraction data on the subunit a of E. coli Fo.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Protons , Serine/metabolism , Threonine/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Escherichia coli/chemistry , Humans , Hydrogen Bonding , Models, Molecular , Sequence Alignment , Serine/chemistry , Threonine/chemistry , X-Ray Diffraction
9.
Proteins ; 84(9): 1203-12, 2016 09.
Article in English | MEDLINE | ID: mdl-27177595

ABSTRACT

Rotary ATPases are involved in numerous physiological processes, with the three distinct types (F/A/V-ATPases) sharing functional properties and structural features. The basic mechanism involves the counter rotation of two motors, a soluble ATP hydrolyzing/synthesizing domain and a membrane-embedded ion pump connected through a central rotor axle and a stator complex. Within the A/V-ATPase family conformational flexibility of the EG stators has been shown to accommodate catalytic cycling and is considered to be important to function. For the A-ATPase three EG structures have been reported, thought to represent conformational states of the stator during different stages of rotary catalysis. Here we use long, detailed atomistic simulations to show that those structures are conformers explored through thermal fluctuations, but do not represent highly populated states of the EG stator in solution. We show that the coiled coil tail domain has a high persistence length (∼100 nm), but retains the ability to adapt to different conformational states through the presence of two hinge regions. Moreover, the stator network of the related V-ATPase has been suggested to adapt to subunit interactions in the collar region in addition to the nucleotide occupancy of the catalytic domain. The MD simulations reported here, reinforce this observation showing that the EG stators have enough flexibility to adapt to significantly different structural re-arrangements and accommodate structural changes in the catalytic domain whilst resisting the large torque generated by catalytic cycling. These results are important to understand the role the stators play in the rotary-ATPase mechanism. Proteins 2016; 84:1203-1212. © 2016 The Authors. Proteins: Structure, Function, and Bioinformatics Published by Wiley Periodicals, Inc.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/chemistry , Protein Subunits/chemistry , Thermus thermophilus/chemistry , Vacuolar Proton-Translocating ATPases/chemistry , Adenosine Triphosphate/chemistry , Biocatalysis , Catalytic Domain , Molecular Dynamics Simulation , Principal Component Analysis , Protein Structure, Secondary , Rotation , Thermus thermophilus/enzymology
10.
J Biol Chem ; 290(17): 10717-28, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25713065

ABSTRACT

Living organisms rely on the FoF1 ATP synthase to maintain the non-equilibrium chemical gradient of ATP to ADP and phosphate that provides the primary energy source for cellular processes. How the Fo motor uses a transmembrane electrochemical ion gradient to create clockwise torque that overcomes F1 ATPase-driven counterclockwise torque at high ATP is a major unresolved question. Using single FoF1 molecules embedded in lipid bilayer nanodiscs, we now report the observation of Fo-dependent rotation of the c10 ring in the ATP synthase (clockwise) direction against the counterclockwise force of ATPase-driven rotation that occurs upon formation of a leash with Fo stator subunit a. Mutational studies indicate that the leash is important for ATP synthase activity and support a mechanism in which residues aGlu-196 and cArg-50 participate in the cytoplasmic proton half-channel to promote leash formation.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Adenosine Triphosphate/biosynthesis , Amino Acid Sequence , Bacterial Proton-Translocating ATPases/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Protein Subunits , Rotation , Sequence Homology, Amino Acid , Static Electricity
11.
Int J Biol Macromol ; 70: 241-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25010476

ABSTRACT

Curcumin, a dietary phytopolyphenol isolated from a perennial herb (Curcuma longa), is a well-known compound effective for bacterial infections and tumors, and also as an antioxidant. In this study, we report the inhibitory effects of curcumin and its analogs on the Escherichia coli ATP synthase F1 sector. A structure-activity relationship study indicated the importance of 4'-hydroxy groups and a ß-diketone moiety for the inhibition. The 3'-demethoxy analog (DMC) inhibited F1 more strongly than curcumin did. Furthermore, these compounds inhibited E. coli growth through oxidative phosphorylation, consistent with their effects on ATPase activity. These results suggest that the two compounds affected bacterial growth through inhibition of ATP synthase. Derivatives including bis(arylmethylidene)acetones (C5 curcuminoids) exhibited only weak activity toward ATPase and bacterial growth.


Subject(s)
Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Curcumin/analogs & derivatives , Curcumin/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Curcumin/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Escherichia coli/growth & development , Oxidative Phosphorylation/drug effects , Structure-Activity Relationship
12.
J Mol Biol ; 426(14): 2547-53, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24838125

ABSTRACT

NMR structures of ζ-subunits, which are recently discovered α-proteobacterial F1F0-ATPase-regulatory proteins representing a Pfam protein family of 246 sequences from 219 species (PF07345), exhibit a four-helix bundle, which is different from all other known F1F0-ATPase inhibitors. Chemical shift mapping reveals a conserved ADP/ATP binding site in ζ-subunit, which mediates long-range conformational changes related to function, as revealed by the structure of the Paracoccus denitrificans ζ-subunit in complex with ADP. These structural data suggest a new mechanism of F1F0-ATPase regulation in α-proteobacteria.


Subject(s)
Alphaproteobacteria/chemistry , Bacterial Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proton-Translocating ATPases/metabolism , Binding Sites , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Paracoccus denitrificans/chemistry , Protein Conformation , Protein Subunits
13.
Mol Microbiol ; 92(5): 973-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24707994

ABSTRACT

In the c-ring rotor of ATP synthases ions are shuttled across the membrane during ATP synthesis by a unique rotary mechanism. We investigated characteristics of the c-ring from the alkaliphile Bacillus pseudofirmus OF4 with respect to evolutionary adaptations to operate with protons at high environmental pH. The X-ray structures of the wild-type c13 ring at pH 9.0 and a 'neutralophile-like' mutant (P51A) at pH 4.4, at 2.4 and 2.8 Šresolution, respectively, reveal a dependency of the conformation and protonation state of the proton-binding glutamate (E(54) ) on environmental hydrophobicity. Faster labelling kinetics with the inhibitor dicyclohexylcarbodiimide (DCCD) demonstrate a greater flexibility of E(54) in the mutant due to reduced water occupancy within the H(+) binding site. A second 'neutralophile-like' mutant (V21N) shows reduced growth at high pH, which is explained by restricted conformational freedom of the mutant's E(54) carboxylate. The study directly connects subtle structural adaptations of the c-ring ion binding site to in vivo effects of alkaliphile cell physiology.


Subject(s)
Bacillus/enzymology , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Dicyclohexylcarbodiimide/pharmacology , Hydrogen-Ion Concentration
14.
Biochem Biophys Res Commun ; 446(1): 358-63, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24607907

ABSTRACT

Motor enzymes such as F1-ATPase and kinesin utilize energy from ATP for their motion. Molecular motions of these enzymes are critical to their catalytic mechanisms and were analyzed thoroughly using a single molecule observation technique. As a tool to analyze and control the ATP-driven motor enzyme motion, we recently synthesized a photoresponsive ATP analog with a p-tert-butylazobenzene tethered to the 2' position of the ribose ring. Using cis/trans isomerization of the azobenzene moiety, we achieved a successful reversible photochromic control over a kinesin-microtubule system in an in vitro motility assay. Here we succeeded to control the hydrolytic activity and rotation of the rotary motor enzyme, F1-ATPase, using this photosensitive ATP analog. Subsequent single molecule observations indicated a unique pause occurring at the ATP binding angle position in the presence of cis form of the analog.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Azo Compounds/metabolism , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proton-Translocating ATPases/genetics , Cyanobacteria/enzymology , Cyanobacteria/genetics , Kinesins/metabolism , Kinetics , Light , Microtubules/metabolism , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Motion , Photochemical Processes , Rotation , Substrate Specificity
15.
Biochim Biophys Acta ; 1837(7): 1063-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24650630

ABSTRACT

H(+)-transporting F1Fo ATP synthase catalyzes the synthesis of ATP via coupled rotary motors within Fo and F1. H(+) transport at the subunit a-c interface in trans-membranous Fo drives rotation of the c-ring within the membrane, with subunit c being bound in a complex with the γ and ε subunits extending from the membrane. Finally, the rotation of subunit γ within the α3ß3 sector of F1 mechanically drives ATP synthesis within the catalytic sites. In this review, we propose and provide evidence supporting the route of proton transfer via half channels from one side of the membrane to the other, and the mechanism of gating H(+) binding to and release from Asp61 of subunit c, via conformational movements of Arg210 in subunit a. We propose that protons are gated from the inside of a four-helix bundle at the periplasmic side of subunit a to drive protonation of cAsp61, and that this gating movement is facilitated by the swiveling of trans-membrane helices (TMHs) 4 and 5 at the site of interaction with cAsp61 on the periphery of the c-ring. Proton release to the cytoplasmic half channel is facilitated by the movement of aArg210 as a consequence of this proposed helical swiveling. Finally, release from the cytoplasmic half channel is mediated by residues in a complex of interacting extra-membraneous loops formed between TMHs of both subunits a and c. This article is part of a Special Issue entitled: 18th European Bioenergetic Conference.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Escherichia coli/enzymology , Molecular Dynamics Simulation , Protons , Amino Acid Sequence , Bacterial Proton-Translocating ATPases/metabolism , Ion Transport , Molecular Sequence Data , Protein Structure, Tertiary
16.
Biochim Biophys Acta ; 1837(7): 1208-18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24513197

ABSTRACT

ATP synthase is a ubiquitous enzyme that is largely conserved across the kingdoms of life. This conservation is in accordance with its central role in chemiosmotic energy conversion, a pathway utilized by far by most living cells. On the other hand, in particular pathogenic bacteria whilst employing ATP synthase have to deal with energetically unfavorable conditions such as low oxygen tensions in the human host, e.g. Mycobacterium tuberculosis can survive in human macrophages for an extended time. It is well conceivable that such ATP synthases may carry idiosyncratic features that contribute to efficient ATP production. In this review genetic and biochemical data on mycobacterial ATP synthase are discussed in terms of rotary catalysis, stator composition, and regulation of activity. ATP synthase in mycobacteria is of particular interest as this enzyme has been validated as a target for promising new antibacterial drugs. A deeper understanding of the working of mycobacterial ATP synthase and its atypical features can provide insight in adaptations of bacterial energy metabolism. Moreover, pinpointing and understanding critical differences as compared with human ATP synthase may provide input for the design and development of selective ATP synthase inhibitors as antibacterials. This article is part of a Special Issue entitled: 18th European Bioenergetic Conference.


Subject(s)
Bacterial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mycobacterium tuberculosis/enzymology , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Bacterial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/chemistry , Molecular Sequence Data , Mycobacterium tuberculosis/drug effects , Protein Structure, Tertiary
17.
Biophys J ; 105(11): 2541-8, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24314084

ABSTRACT

Rotation of the γ subunit of the F1-ATPase plays an essential role in energy transduction by F1-ATPase. Hydrolysis of an ATP molecule induces a 120° step rotation that consists of an 80° substep and 40° substep. ATP binding together with ADP release causes the first 80° step rotation. Thus, nucleotide binding is very important for rotation and energy transduction by F1-ATPase. In this study, we introduced a ßY341W mutation as an optical probe for nucleotide binding to catalytic sites, and a ßE190Q mutation that suppresses the hydrolysis of nucleoside triphosphate (NTP). Using a mutant monomeric ßY341W subunit and a mutant α3ß3γ subcomplex containing the ßY341W mutation with or without an additional ßE190Q mutation, we examined the binding of various NTPs (i.e., ATP, GTP, and ITP) and nucleoside diphosphates (NDPs, i.e., ADP, GDP, and IDP). The affinity (1/Kd) of the nucleotides for the isolated ß subunit and third catalytic site in the subcomplex was in the order ATP/ADP > GTP/GDP > ITP/IDP. We performed van't Hoff analyses to obtain the thermodynamic parameters of nucleotide binding. For the isolated ß subunit, NDPs and NTPs with the same base moiety exhibited similar ΔH(0) and ΔG(0) values at 25°C. The binding of nucleotides with different bases to the isolated ß subunit resulted in different entropy changes. Interestingly, NDP binding to the α3ß(Y341W)3γ subcomplex had similar Kd and ΔG(0) values as binding to the isolated ß(Y341W) subunit, but the contributions of the enthalpy term and the entropy term were very different. We discuss these results in terms of the change in the tightness of the subunit packing, which reduces the excluded volume between subunits and increases water entropy.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Models, Biological , Nucleotides/metabolism , Thermodynamics , Bacillus/enzymology , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Kinetics , Mutation, Missense , Nucleotides/chemistry , Protein Binding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
18.
J Biol Chem ; 288(35): 25535-25541, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23864659

ABSTRACT

Subunit a plays a key role in promoting H(+) transport-coupled rotary motion of the subunit c ring in F1Fo ATP synthase. H(+) binding and release occur at Asp-61 in the middle of the second transmembrane helix (TMH) of Fo subunit c. H(+) are thought to reach cAsp61 via aqueous half-channels formed by TMHs 2-5 of subunit a. Movements of TMH4 and TMH5 have been proposed to facilitate protonation of cAsp61 from a half channel centered in a four helix bundle at the periplasmic side of subunit a. The possible necessity of these proposed TMH movements was investigated by assaying ATP driven H(+) pumping function before and after cross-linking paired Cys substitutions at the center of TMHs within subunit a. The cross-linking of the Cys pairs aG218C/I248C in TMH4 and TMH5, and aL120C/H245C in TMH2 and TMH5, inhibited H(+) pumping by 85-90%. H(+) pumping function was largely unaffected by modification of the same Cys residues in the absence of cross-link formation. The inhibition is consistent with the proposed requirement for TMH movements during the gating of periplasmic H(+) access to cAsp61. The cytoplasmic loops of subunit a have been implicated in gating H(+) release to the cytoplasm, and previous cross-linking experiments suggest that the chemically reactive regions of the loops may pack as a single domain. Here we show that Cys substitutions in these domains can be cross-linked with retention of function and conclude that these domains need not undergo large conformational changes during enzyme function.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Amino Acid Substitution , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mutation, Missense , Protein Structure, Secondary , Protein Structure, Tertiary
19.
FEBS Lett ; 587(7): 892-7, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23416299

ABSTRACT

The interaction of the membrane traversing stator subunits a and b of the rotary ATP synthase was probed by substitution of a single Cys into each subunit with subsequent Cu(2+) catalyzed cross-linking. Extensive interaction between the transmembrane (TM) region of one b subunit and TM2 of subunit a was indicated by cross-linking with 6 Cys pairs introduced into these regions. Additional disulfide cross-linking was observed between the N-terminus of subunit b and the periplasmic loop connecting TM4 and TM5 of subunit a. Finally, benzophenone-4-maleimide derivatized Cys in the 2-3 periplasmic loop of subunit a were shown to cross-link with the periplasmic N-terminal region of subunit b. These experiments help to define the juxtaposition of subunits b and a in the ATP synthase.


Subject(s)
ATP Synthetase Complexes/metabolism , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , ATP Synthetase Complexes/chemistry , ATP Synthetase Complexes/genetics , Bacterial Proton-Translocating ATPases/chemistry , Bacterial Proton-Translocating ATPases/genetics , Benzophenones/chemistry , Benzophenones/pharmacology , Binding Sites/genetics , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Disulfides/chemistry , Disulfides/pharmacology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Immunoblotting , Maleimides/chemistry , Maleimides/pharmacology , Models, Molecular , Mutation , Protein Binding/drug effects , Protein Multimerization , Protein Structure, Tertiary
20.
Philos Trans R Soc Lond B Biol Sci ; 368(1611): 20120023, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23267177

ABSTRACT

The rotary motor F(1)-ATPase from the thermophilic Bacillus PS3 (TF(1)) is one of the best-studied of all molecular machines. F(1)-ATPase is the part of the enzyme F(1)F(O)-ATP synthase that is responsible for generating most of the ATP in living cells. Single-molecule experiments have provided a detailed understanding of how ATP hydrolysis and synthesis are coupled to internal rotation within the motor. In this work, we present evidence that mesophilic F(1)-ATPase from Escherichia coli (EF(1)) is governed by the same mechanism as TF(1) under laboratory conditions. Using optical microscopy to measure rotation of a variety of marker particles attached to the γ-subunit of single surface-bound EF(1) molecules, we characterized the ATP-binding, catalytic and inhibited states of EF(1). We also show that the ATP-binding and catalytic states are separated by 35±3°. At room temperature, chemical processes occur faster in EF(1) than in TF(1), and we present a methodology to compensate for artefacts that occur when the enzymatic rates are comparable to the experimental temporal resolution. Furthermore, we show that the molecule-to-molecule variation observed at high ATP concentration in our single-molecule assays can be accounted for by variation in the orientation of the rotating markers.


Subject(s)
Bacterial Proton-Translocating ATPases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Actin Cytoskeleton/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/chemistry , Binding Sites , Enzyme Activation , Enzyme Assays , Hydrodynamics , Hydrolysis , Magnesium/chemistry , Optical Imaging/methods , Protein Binding , Protein Conformation , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...