Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Mol Sci ; 23(14)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35887384

ABSTRACT

As an effective and versatile strategy to compartmentalize cellular components without the need for lipid membranes, phase separation has been found to underpin a wide range of intranuclear processes, particularly those involving chromatin. Many of the unique physico-chemical properties of chromatin-based phase condensates are harnessed by the cell to accomplish complex regulatory functions in a spatially and temporally controlled manner. Here, we survey key recent findings on the mechanistic roles of phase separation in regulating the organization and dynamics of chromatin-based molecular processes across length scales, packing states and intranuclear functions, with a particular emphasis on quantitative characterizations of these condensates enabled by advanced imaging-based approaches. By illuminating the complex interplay between chromatin and various chromatin-interacting molecular species mediated by phase separation, this review sheds light on an emerging multi-scale, multi-modal and multi-faceted landscape that hierarchically regulates the genome within the highly crowded and dynamic nuclear space. Moreover, deficiencies in existing studies also highlight the need for mechanism-specific criteria and multi-parametric approaches for the characterization of chromatin-based phase separation using complementary techniques and call for greater efforts to correlate the quantitative features of these condensates with their functional consequences in close-to-native cellular contexts.


Subject(s)
Cell Nucleus , Chromatin , Cell Nucleus/genetics , Chromatin/genetics , Genome
2.
Molecules ; 24(3)2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30704145

ABSTRACT

F-ATP synthases use proton flow through the FO domain to synthesize ATP in the F1 domain. In Escherichia coli, the enzyme consists of rotor subunits γεc10 and stator subunits (αß)3δab2. Subunits c10 or (αß)3 alone are rotationally symmetric. However, symmetry is broken by the b2 homodimer, which together with subunit δa, forms a single eccentric stalk connecting the membrane embedded FO domain with the soluble F1 domain, and the central rotating and curved stalk composed of subunit γε. Although each of the three catalytic binding sites in (αß)3 catalyzes the same set of partial reactions in the time average, they might not be fully equivalent at any moment, because the structural symmetry is broken by contact with b2δ in F1 and with b2a in FO. We monitored the enzyme's rotary progression during ATP hydrolysis by three single-molecule techniques: fluorescence video-microscopy with attached actin filaments, Förster resonance energy transfer between pairs of fluorescence probes, and a polarization assay using gold nanorods. We found that one dwell in the three-stepped rotary progression lasting longer than the other two by a factor of up to 1.6. This effect of the structural asymmetry is small due to the internal elastic coupling.


Subject(s)
Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Actins/chemistry , Actins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Gold , Kinetics , Models, Molecular , Molecular Conformation , Molecular Structure , Nanotubes/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship
3.
Biochim Biophys Acta Bioenerg ; 1859(9): 775-788, 2018 09.
Article in English | MEDLINE | ID: mdl-29932911

ABSTRACT

F-type ATP synthases are extraordinary multisubunit proteins that operate as nanomotors. The Escherichia coli (E. coli) enzyme uses the proton motive force (pmf) across the bacterial plasma membrane to drive rotation of the central rotor subunits within a stator subunit complex. Through this mechanical rotation, the rotor coordinates three nucleotide binding sites that sequentially catalyze the synthesis of ATP. Moreover, the enzyme can hydrolyze ATP to turn the rotor in the opposite direction and generate pmf. The direction of net catalysis, i.e. synthesis or hydrolysis of ATP, depends on the cell's bioenergetic conditions. Different control mechanisms have been found for ATP synthases in mitochondria, chloroplasts and bacteria. This review discusses the auto-inhibitory behavior of subunit ε found in FOF1-ATP synthases of many bacteria. We focus on E. coli FOF1-ATP synthase, with insights into the regulatory mechanism of subunit ε arising from structural and biochemical studies complemented by single-molecule microscopy experiments.


Subject(s)
Adenosine Triphosphate/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Proton-Translocating ATPases/metabolism , Energy Metabolism , Protein Subunits/metabolism
4.
J Biol Chem ; 292(27): 11262-11279, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28495884

ABSTRACT

The F1F0 -ATP (F-ATP) synthase is essential for growth of Mycobacterium tuberculosis, the causative agent of tuberculosis (TB). In addition to their synthase function most F-ATP synthases possess an ATP-hydrolase activity, which is coupled to proton-pumping activity. However, the mycobacterial enzyme lacks this reverse activity, but the reason for this deficiency is unclear. Here, we report that a Mycobacterium-specific, 36-amino acid long C-terminal domain in the nucleotide-binding subunit α (Mtα) of F-ATP synthase suppresses its ATPase activity and determined the mechanism of suppression. First, we employed vesicles to show that in intact membrane-embedded mycobacterial F-ATP synthases deletion of the C-terminal domain enabled ATPase and proton-pumping activity. We then generated a heterologous F-ATP synthase model system, which demonstrated that transfer of the mycobacterial C-terminal domain to a standard F-ATP synthase α subunit suppresses ATPase activity. Single-molecule rotation assays indicated that the introduction of this Mycobacterium-specific domain decreased the angular velocity of the power-stroke after ATP binding. Solution X-ray scattering data and NMR results revealed the solution shape of Mtα and the 3D structure of the subunit α C-terminal peptide 521PDEHVEALDEDKLAKEAVKV540 of M. tubercolosis (Mtα(521-540)), respectively. Together with cross-linking studies, the solution structural data lead to a model, in which Mtα(521-540) comes in close proximity with subunit γ residues 104-109, whose interaction may influence the rotation of the camshaft-like subunit γ. Finally, we propose that the unique segment Mtα(514-549), which is accessible at the C terminus of mycobacterial subunit α, is a promising drug epitope.


Subject(s)
Adaptation, Physiological , Bacterial Proteins/chemistry , Evolution, Molecular , Models, Molecular , Mycobacterium tuberculosis/enzymology , Peptides/chemistry , Proton-Translocating ATPases/chemistry , Bacterial Proteins/genetics , Mycobacterium tuberculosis/genetics , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics , Proton-Translocating ATPases/genetics , X-Ray Diffraction
5.
J Biol Chem ; 291(49): 25351-25363, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27729450

ABSTRACT

The angular velocities of ATPase-dependent power strokes as a function of the rotational position for the A-type molecular motor A3B3DF, from the Methanosarcina mazei Gö1 A-ATP synthase, and the thermophilic motor α3ß3γ, from Geobacillus stearothermophilus (formerly known as Bacillus PS3) F-ATP synthase, are resolved at 5 µs resolution for the first time. Unexpectedly, the angular velocity profile of the A-type was closely similar in the angular positions of accelerations and decelerations to the profiles of the evolutionarily distant F-type motors of thermophilic and mesophilic origins, and they differ only in the magnitude of their velocities. M. mazei A3B3DF power strokes occurred in 120° steps at saturating ATP concentrations like the F-type motors. However, because ATP-binding dwells did not interrupt the 120° steps at limiting ATP, ATP binding to A3B3DF must occur during the catalytic dwell. Elevated concentrations of ADP did not increase dwells occurring 40° after the catalytic dwell. In F-type motors, elevated ADP induces dwells 40° after the catalytic dwell and slows the overall velocity. The similarities in these power stroke profiles are consistent with a common rotational mechanism for A-type and F-type rotary motors, in which the angular velocity is limited by the rotary position at which ATP binding occurs and by the drag imposed on the axle as it rotates within the ring of stator subunits.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Archaeal Proteins/chemistry , Methanosarcina/enzymology , Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Archaeal Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Geobacillus stearothermophilus/enzymology , Proton-Translocating ATPases/metabolism
6.
Biochim Biophys Acta ; 1857(2): 177-187, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26682760

ABSTRACT

A1AO ATP synthases couple ion-transport of the AO sector and ATP synthesis/hydrolysis of the A3B3-headpiece via their stalk subunits D and F. Here, we produced and purified stable A3B3D- and A3B3DF-complexes of the Methanosarcina mazei Gö1 A-ATP synthase as confirmed by electron microscopy. Enzymatic studies with these complexes showed that the M. mazei Gö1 A-ATP synthase subunit F is an ATPase activating subunit. The maximum ATP hydrolysis rates (Vmax) of A3B3D and A3B3DF were determined by substrate-dependent ATP hydrolysis experiments resulting in a Vmax of 7.9 s(-1) and 30.4 s(-1), respectively, while the KM is the same for both. Deletions of the N- or C-termini of subunit F abolished the effect of ATP hydrolysis activation. We generated subunit F mutant proteins with single amino acid substitutions and demonstrated that the subunit F residues S84 and R88 are important in stimulating ATP hydrolysis. Hybrid formation of the A3B3D-complex with subunit F of the related eukaryotic V-ATPase of Saccharomyces cerevisiae or subunit ε of the F-ATP synthase from Mycobacterium tuberculosis showed that subunit F of the archaea and eukaryotic enzymes are important in ATP hydrolysis.


Subject(s)
ATP Synthetase Complexes/chemistry , Adenosine Triphosphate/chemistry , Archaeal Proteins/chemistry , Methanosarcina/chemistry , Protein Subunits/chemistry , ATP Synthetase Complexes/genetics , ATP Synthetase Complexes/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acid Substitution , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrolysis , Kinetics , Methanosarcina/enzymology , Models, Molecular , Molecular Sequence Data , Mutation , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Sequence Deletion , Species Specificity
7.
Nature ; 459(7245): 364-70, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19458712

ABSTRACT

Adenosine triphosphate (ATP), the universal fuel of the cell, is synthesized from adenosine diphosphate (ADP) and inorganic phosphate (P(i)) by 'ATP synthase' (F(O)F(1)-ATPase). During respiration or photosynthesis, an electrochemical potential difference of protons is set up across the respective membranes. This powers the enzyme's electrical rotary nanomotor (F(O)), which drives the chemical nanomotor (F(1)) by elastic mechanical-power transmission, producing ATP with high kinetic efficiency. Attempts to understand in detail the mechanisms of torque generation in this simple and robust system have been both aided and complicated by a wealth of sometimes conflicting data.


Subject(s)
Elasticity , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Torque , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/metabolism , Catalytic Domain , Thermodynamics
8.
Proc Natl Acad Sci U S A ; 108(10): 3924-9, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21368147

ABSTRACT

ATP is synthesized by ATP synthase (F(O)F(1)-ATPase). Its rotary electromotor (F(O)) translocates protons (in some organisms sodium cations) and generates torque to drive the rotary chemical generator (F(1)). Elastic power transmission between F(O) and F(1) is essential for smoothing the cooperation of these stepping motors, thereby increasing their kinetic efficiency. A particularly compliant elastic domain is located on the central rotor (c(10-15)/ε/γ), right between the two sites of torque generation and consumption. The hinge on the active lever on subunit ß adds further compliance. It is under contention whether or not the peripheral stalk (and the "stator" as a whole) also serves as elastic buffer. In the enzyme from Escherichia coli, the most extended component of the stalk is the homodimer b(2), a right-handed α-helical coiled coil. By fluctuation analysis we determined the spring constant of the stator in response to twisting and bending, and compared wild-type with b-mutant enzymes. In both deformation modes, the stator was very stiff in the wild type. It was more compliant if b was elongated by 11 amino acid residues. Substitution of three consecutive residues in b by glycine, expected to destabilize its α-helical structure, further reduced the stiffness against bending deformation. In any case, the stator was at least 10-fold stiffer than the rotor, and the enzyme retained its proton-coupled activity.


Subject(s)
Molecular Motor Proteins/metabolism , Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Elasticity , Escherichia coli/enzymology , Magnetics , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/genetics , Molecular Sequence Data , Mutation , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/genetics , Sequence Homology, Amino Acid
9.
Int J Biochem Cell Biol ; 176: 106667, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39362301

ABSTRACT

Chromatin remodeling, carried out by four major subfamilies of ATP-dependent remodeler complexes across eukaryotes, alleviates the topological challenge posed by nucleosomes to regulate genome access. Recently, single-molecule and single-cell imaging techniques have been widely employed to probe this crucial process, both in vitro and in cellulo. Herein, we provide an integrated account of key recent efforts that leverage these approaches to visualize, quantify and map chromatin remodelers at work, elucidating diverse aspects of the remodeling process in both space and time, including molecular mechanisms of DNA wrapping/unwrapping, nucleosome translocation and histone exchange, dynamics of chromatin binding/target search and their intranuclear organization into hotspots or phase condensates, as well as functional coupling with transcription. The mechanistic insights and quantitative parameters revealed shed light on a multi-modal yet shared landscape for regulating remodeling across molecular and cellular scales, and pave the way for further interrogating the implications of its misregulation in disease contexts.

10.
Nat Commun ; 15(1): 7646, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223123

ABSTRACT

Despite their prevalent cancer implications, the in vivo dynamics of SWI/SNF chromatin remodelers and how misregulation of such dynamics underpins cancer remain poorly understood. Using live-cell single-molecule tracking, we quantify the intranuclear diffusion and chromatin-binding of three key subunits common to all major human SWI/SNF remodeler complexes (BAF57, BAF155 and BRG1), and resolve two temporally distinct stable binding modes for the fully assembled complex. Super-resolved density mapping reveals heterogeneous, nanoscale remodeler binding "hotspots" across the nucleoplasm where multiple binding events (especially longer-lived ones) preferentially cluster. Importantly, we uncover distinct roles of the bromodomain in modulating chromatin binding/targeting in a DNA-accessibility-dependent manner, pointing to a model where successive longer-lived binding within "hotspots" leads to sustained productive remodeling. Finally, systematic comparison of six common BRG1 mutants implicated in various cancers unveils alterations in chromatin-binding dynamics unique to each mutant, shedding insight into a multi-modal landscape regulating the spatio-temporal organizational dynamics of SWI/SNF remodelers.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Chromosomal Proteins, Non-Histone , DNA Helicases , Neoplasms , Nuclear Proteins , Single Molecule Imaging , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Single Molecule Imaging/methods , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Protein Binding , Mutation , Cell Line, Tumor , Protein Domains , Adenosine Triphosphatases
11.
Nanoscale ; 14(6): 2327-2336, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35084006

ABSTRACT

Single-molecule Förster resonance energy transfer (smFRET) is a key technique to observe conformational changes in molecular motors and to access the details of single-molecule static and dynamic disorder during catalytic processes. However, studying freely diffusing molecules in solution is limited to a few tens of milliseconds, while surface attachment often bears the risk to restrict their natural motion. In this paper we combine smFRET and electrokinetic trapping (ABEL trap) to non-invasively hold single FOF1-ATP synthases for up to 3 s within the detection volume, thereby extending the observation time by a factor of 10 as compared to Brownian diffusion without surface attachment. In addition, we are able to monitor complete reaction cycles and to selectively trap active molecules based on their smFRET signal, thus speeding up the data acquisition process. We demonstrate the capability of our method to study the dynamics of single molecules by recording the ATP-hydrolysis driven rotation of individual FOF1-ATP synthase molecules over numerous reaction cycles and extract their kinetic rates. We argue that our method is not limited to motor proteins. Instead, it can be applied to monitor conformational changes with millisecond time resolution for a wide range of enzymes, thereby making it a versatile tool for studying protein dynamics.


Subject(s)
Adenosine Triphosphate , Fluorescence Resonance Energy Transfer , Diffusion , Kinetics
12.
Chemphyschem ; 12(3): 627-33, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21344597

ABSTRACT

Single-molecule Förster resonance energy transfer (FRET) experiments were performed on the enzyme RNase H specifically labeled with a FRET dye pair and diffusing freely in solutions containing between 0 and 6 M of the chemical denaturant GdmCl. We measured FRET efficiency histograms with high statistical accuracy to identify the well-known folding intermediate of RNase H, which escaped observation in our previous smFRET studies on immobilized preparations. Even with excellent data statistics, a folding intermediate is not obvious from the raw data. However, it can be uncovered by a global fitting procedure applied to the FRET histograms at all 22 GdmCl concentrations, in which a number of parameters were constrained. Most importantly, the fractional populations of the folded, unfolded and intermediate states were coupled by assuming the Boltzmann relation and a linear dependence of the free energies on the GdmCl concentration. The analysis not only resolves the apparent discrepancy with other data on RNase H, but yields free energy differences between the three populations in agreement with literature data. In addition, it removes the strong and unexplained broadening of the unfolded-state distribution in the transition region that was seen earlier in the two-state analysis.


Subject(s)
Ribonuclease H/chemistry , Fluorescence Resonance Energy Transfer , Guanidine/chemistry , Models, Molecular , Protein Denaturation , Protein Folding , Protein Structure, Tertiary
13.
Proc Natl Acad Sci U S A ; 105(46): 17760-5, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-19001275

ABSTRACT

The 2 nanomotors of rotary ATP synthase, ionmotive F(O) and chemically active F(1), are mechanically coupled by a central rotor and an eccentric bearing. Both motors rotate, with 3 steps in F(1) and 10-15 in F(O). Simulation by statistical mechanics has revealed that an elastic power transmission is required for a high rate of coupled turnover. Here, we investigate the distribution in the F(O)F(1) structure of compliant and stiff domains. The compliance of certain domains was restricted by engineered disulfide bridges between rotor and stator, and the torsional stiffness (kappa) of unrestricted domains was determined by analyzing their thermal rotary fluctuations. A fluorescent magnetic bead was attached to single molecules of F(1) and a fluorescent actin filament to F(O)F(1), respectively. They served to probe first the functional rotation and, after formation of the given disulfide bridge, the stochastic rotational motion. Most parts of the enzyme, in particular the central shaft in F(1), and the long eccentric bearing were rather stiff (torsional stiffness kappa > 750 pNnm). One domain of the rotor, namely where the globular portions of subunits gamma and epsilon of F(1) contact the c-ring of F(O), was more compliant (kappa congruent with 68 pNnm). This elastic buffer smoothes the cooperation of the 2 stepping motors. It is located were needed, between the 2 sites where the power strokes in F(O) and F(1) are generated and consumed.


Subject(s)
Escherichia coli/enzymology , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Rotation , Actin Cytoskeleton/metabolism , Compliance , Enzyme Activation , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Hydrolysis , Magnetics , Microspheres , Models, Molecular , Oxidation-Reduction , Protein Structure, Tertiary , Quantum Dots
14.
FEBS J ; 286(10): 1894-1907, 2019 05.
Article in English | MEDLINE | ID: mdl-30791207

ABSTRACT

The Na+ translocating F1 FO ATP synthase from Acetobacterium woodii shows a subunit stoichiometry of α3 :ß3 :γ:δ:ε:a:b2 :(c2/3 )9 :c1 and reveals an evolutionary path between synthases and pumps involving adaptations in the rotor c-ring, which is composed of F- and vacuolar-type c subunits in a stoichiometry of 9 : 1. This hybrid turbine couples rotation with Na+ translocation in the FO part and rotation of the central stalk subunits γ-ε to drive ATP synthesis in the catalytic α3 :ß3 headpiece. Here, we isolated a highly pure recombinant A. woodii F-ATP synthase and present the first projected structure of this hybrid engine as determined by negative-stain electron microscopy and single-particle analysis. The uniqueness of the A. woodii F-ATP synthase is also reflected by an extra 17 amino acid residues loop (195 TSGKVKITEETKEEKSK211 ) in subunit γ. Deleting the loop-encoding DNA sequence (γΔ195-211 ) and purifying the recombinant F-ATP synthase γΔ195-211 mutant provided a platform to study its effect in enzyme stability and activity. The recombinant F-ATP synthase γΔ195-211 mutant revealed the same subunit composition as the wild-type enzyme and a minor reduction in ATP hydrolysis. When reconstituted into proteoliposomes ATP synthesis and Na+ transport were diminished, demonstrating the importance of the γ195-211 loop in both enzymatic processes. Based on a structural model, a coupling mechanism for this enzyme is proposed, highlighting the role of the γ-loop. Finally, the γ195-211 loop of A. woodii is discussed in comparison with the extra γ-loops of mycobacterial and chloroplasts F-ATP synthases described to be involved in species-specific regulatory mechanisms.


Subject(s)
Acetobacterium/enzymology , Adenosine Triphosphate/biosynthesis , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Sodium/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microscopy, Electron , Models, Molecular , Mutation , Protein Conformation , Proteolipids/metabolism , Proton-Translocating ATPases/genetics
15.
Biophys J ; 95(10): 4979-87, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18723591

ABSTRACT

The F(O)F(1)-ATPase is a rotary molecular motor. Driven by ATP-hydrolysis, its central shaft rotates in 80 degrees and 40 degrees steps, interrupted by catalytic and ATP-waiting dwells. We recorded rotations and halts by means of microvideography in laboratory coordinates. A correlation with molecular coordinates was established by using an engineered pair of cysteines that, under oxidizing conditions, formed zero-length cross-links between the rotor and the stator in an orientation as found in crystals. The fixed orientation coincided with that of the catalytic dwell, whereas the ATP waiting dwell was displaced from it by +40 degrees . In crystals, the convex side of the cranked central shaft faces an empty nucleotide binding site, as if holding it open for arriving ATP. Functional studies suggest that three sites are occupied during a catalytic dwell. Our data imply that the convex side faces a nucleotide-occupied rather than an empty site. The enzyme conformation in crystals seems to differ from the conformation during either dwell of the active enzyme. A revision of current schemes of the mechanism is proposed.


Subject(s)
Adenosine Triphosphate/chemistry , Models, Chemical , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/ultrastructure , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/ultrastructure , Computer Simulation , Crystallography , Protein Conformation , Rotation , Statistics as Topic
16.
BMC Biotechnol ; 7: 48, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17697378

ABSTRACT

BACKGROUND: As a new class of therapeutic and diagnostic reagents, more than fifteen years ago RNA and DNA aptamers were identified as binding molecules to numerous small compounds, proteins and rarely even to complete pathogen particles. Most aptamers were isolated from complex libraries of synthetic nucleic acids by a process termed SELEX based on several selection and amplification steps. Here we report the application of a new one-step selection method (MonoLEX) to acquire high-affinity DNA aptamers binding Vaccinia virus used as a model organism for complex target structures. RESULTS: The selection against complete Vaccinia virus particles resulted in a 64-base DNA aptamer specifically binding to orthopoxviruses as validated by dot blot analysis, Surface Plasmon Resonance, Fluorescence Correlation Spectroscopy and real-time PCR, following an aptamer blotting assay. The same oligonucleotide showed the ability to inhibit in vitro infection of Vaccinia virus and other orthopoxviruses in a concentration-dependent manner. CONCLUSION: The MonoLEX method is a straightforward procedure as demonstrated here for the identification of a high-affinity DNA aptamer binding Vaccinia virus. MonoLEX comprises a single affinity chromatography step, followed by subsequent physical segmentation of the affinity resin and a single final PCR amplification step of bound aptamers. Therefore, this procedure improves the selection of high affinity aptamers by reducing the competition between aptamers of different affinities during the PCR step, indicating an advantage for the single-round MonoLEX method.


Subject(s)
Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/isolation & purification , Chromatography, Affinity/methods , DNA, Viral/genetics , DNA, Viral/isolation & purification , Gene Targeting/methods , Vaccinia virus/genetics , Polymerase Chain Reaction/methods
18.
FEBS Lett ; 591(6): 854-862, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28231387

ABSTRACT

In archaea the A1 AO ATP synthase uses a transmembrane electrochemical potential to generate ATP, while the soluble A1 domain (subunits A3 B3 DF) alone can hydrolyse ATP. The three nucleotide-binding AB pairs form a barrel-like structure with a central orifice that hosts the rotating central stalk subunits DF. ATP binding, hydrolysis and product release cause a conformational change inside the A:B-interface, which enforces the rotation of subunits DF. Recently, we reported that subunit F is a stimulator of ATPase activity. Here, we investigated the nucleotide-dependent conformational changes of subunit F relative to subunit D during ATP hydrolysis in the A3 B3 DF complex of the Methanosarcina mazei Gö1 A-ATP synthase using single-molecule Förster resonance energy transfer. We found two conformations for subunit F during ATP hydrolysis.


Subject(s)
ATP Synthetase Complexes/metabolism , Archaeal Proteins/metabolism , Fluorescence Resonance Energy Transfer/methods , Methanosarcina/enzymology , ATP Synthetase Complexes/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Archaeal Proteins/chemistry , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Kinetics , Microscopy, Confocal , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism
19.
J Mol Biol ; 427(10): 1866-74, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25681694

ABSTRACT

HAMLET (human alpha-lactalbumin made lethal to tumor cells) interacts with multiple tumor cell compartments, affecting cell morphology, metabolism, proteasome function, chromatin structure and viability. This study investigated if these diverse effects of HAMLET might be caused, in part, by a direct effect on the ATP synthase and a resulting reduction in cellular ATP levels. A dose-dependent reduction in cellular ATP levels was detected in A549 lung carcinoma cells, and by confocal microscopy, co-localization of HAMLET with the nucleotide-binding subunits α (non-catalytic) and ß (catalytic) of the energy converting F1F0 ATP synthase was detected. As shown by fluorescence correlation spectroscopy, HAMLET binds to the F1 domain of the F1F0 ATP synthase with a dissociation constant (KD) of 20.5µM. Increasing concentrations of the tumoricidal protein HAMLET added to the enzymatically active α3ß3γ complex of the F-ATP synthase lowered its ATPase activity, demonstrating that HAMLET binding to the F-ATP synthase effects the catalysis of this molecular motor. Single-molecule analysis was applied to study HAMLET-α3ß3γ complex interaction. Whereas the α3ß3γ complex of the F-ATP synthase rotated in a counterclockwise direction with a mean rotational rate of 3.8±0.7s(-1), no rotation could be observed in the presence of bound HAMLET. Our findings suggest that direct effects of HAMLET on the F-ATP synthase may inhibit ATP-dependent cellular processes.


Subject(s)
Apoptosis/drug effects , Lactalbumin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Molecular Targeted Therapy , Oleic Acids/pharmacology , Adenosine Triphosphate/metabolism , Humans , Lung Neoplasms/pathology , Mitochondria/enzymology , Protein Binding/drug effects , Tumor Cells, Cultured
20.
Philos Trans R Soc Lond B Biol Sci ; 368(1611): 20120024, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23267178

ABSTRACT

F(O)F(1)-ATP synthases are ubiquitous proton- or ion-powered membrane enzymes providing ATP for all kinds of cellular processes. The mechanochemistry of catalysis is driven by two rotary nanomotors coupled within the enzyme. Their different step sizes have been observed by single-molecule microscopy including videomicroscopy of fluctuating nanobeads attached to single enzymes and single-molecule Förster resonance energy transfer. Here we review recent developments of approaches to monitor the step size of subunit rotation and the transient elastic energy storage mechanism in single F(O)F(1)-ATP synthases.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Membrane Transport Proteins/chemistry , Proton-Translocating ATPases/chemistry , Adenosine Triphosphate/chemistry , Algorithms , Catalysis , Cell Membrane/chemistry , Computational Biology , Computer Simulation , Luminescent Proteins/chemistry , Membrane Proteins/chemistry , Microscopy, Fluorescence , Microscopy, Video , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Rotation
SELECTION OF CITATIONS
SEARCH DETAIL