Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
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
2.
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
3.
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
4.
Biochimie ; 165: 156-160, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31377193

ABSTRACT

Mycobacterium tuberculosis (Mtb) protein tyrosine phosphatase (PtpA) has so far been known to control intracellular survival of mycobacteria; whereas the ATP synthase which is essential for mycobacterial growth has recently been contemplated in developing a breakthrough anti-TB drug, diarylquinoline. Since both of these enzymes have been established as validated drug targets; we report a robust and functional relationship between these two enzymes through a series of experiments using Mtb H37Ra. In the present study we report that the mycobacterial ATP synthase alpha subunit is regulated by PtpA. We generated gene knock-out for the enzyme PtpA and subjected to determine the mycobacterial replication and the proteome profile of wild type, mutant (ΔptpA) and complemented (ΔptpA:ptpA) strains of Mtb H37Ra. A substantial amount of decrease in the protein level of ATP synthase alpha subunit (AtpA) in case of mutant H37Ra was observed, while the levels of the enzyme were either increased or remained unchanged, in wild type and in the complemented strains.


Subject(s)
Bacterial Proteins/physiology , Bacterial Proton-Translocating ATPases/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Protein Tyrosine Phosphatases/physiology , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Diarylquinolines/pharmacology , Gene Knockout Techniques , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Protein Tyrosine Phosphatases/genetics
5.
J Food Sci ; 84(1): 138-146, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30569590

ABSTRACT

Many novel bacterial targets and natural inhibitors of enzymes are currently being considered to overcome antibiotic resistance of Escherichia coli. Hence, in this study, 20 essential oil constituents were screened for their potential inhibitory effect on E. coli ATP synthase. This enzyme is involved in the hydrolysis of ATP into ADP and inorganic phosphate (Pi). First, E. coli membrane ATP synthase was isolated via cell lysis. A spectrophotometric method was optimized to quantify the released phosphate from ATP hydrolysis in order to follow the enzymatic activity. The method was validated by determining the kinetic parameters of this reaction (Km = 144.66 µM and Vmax = 270.27 µM/min), and through the inhibition assays of ATP synthase using three reference inhibitors, thymoquinone (half maximal inhibitory concentration [IC50 ] = 50.93 µM), resveratrol (maximum inhibition of 40%), and quercetin (IC50 = 29.01 µM). Among the studied essential oil components, α-terpinene was the most potent inhibitor (IC50 = 19.74 µM) followed by ß-pinene, isoeugenol, eugenol, and estragole.


Subject(s)
Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Escherichia coli/drug effects , Oils, Volatile/analysis , Adenosine Triphosphate/metabolism , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli/enzymology , Hydrogen-Ion Concentration , Hydrolysis , Inhibitory Concentration 50 , Phosphates/analysis
6.
Cell ; 175(2): 571-582.e11, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30146159

ABSTRACT

Elucidating the benefits of individual microbiota-derived molecules in host animals is important for understanding the symbiosis between humans and their microbiota. The bacteria-secreted enterobactin (Ent) is an iron scavenging siderophore with presumed negative effects on hosts. However, the high prevalence of Ent-producing commensal bacteria in the human gut raises the intriguing question regarding a potential host mechanism to beneficially use Ent. We discovered an unexpected and striking role of Ent in supporting growth and the labile iron pool in C. elegans. We show that Ent promotes mitochondrial iron uptake and does so, surprisingly, by binding to the ATP synthase α subunit, which acts inside of mitochondria and independently of ATP synthase. We also demonstrated the conservation of this mechanism in mammalian cells. This study reveals a distinct paradigm for the "iron tug of war" between commensal bacteria and their hosts and an important mechanism for mitochondrial iron uptake and homeostasis.


Subject(s)
Enterobactin/physiology , Iron/metabolism , Siderophores/physiology , Adenosine Triphosphate/metabolism , Animals , Bacterial Proton-Translocating ATPases/metabolism , Bacterial Proton-Translocating ATPases/physiology , Biological Transport , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Enterobactin/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , HEK293 Cells , Humans , Iron/physiology , Mitochondria/metabolism
7.
Tuberculosis (Edinb) ; 108: 56-63, 2018 01.
Article in English | MEDLINE | ID: mdl-29523328

ABSTRACT

A limited number of anti-tuberculosis drug candidates with novel mode of action have entered clinical trials in recent years. ATP synthase is one such validated drug target which has yielded a drug recently. The aim of this study was to identify the novel chemical scaffolds targeting the Mycobacterium tuberculosis (M. tuberculosis) ATP synthase. In this study, inverted membrane vesicles of Mycobacterium smegmatis were prepared to establish luciferin based ATP estimation assay. This assay was used to screen 700 compounds which were earlier found to be active on the whole cell of M. tuberculosis. Antibacterial activity of hits against various susceptible and drug-resistant strains of M. tuberculosis was evaluated using the microplate alamar blue assay and their cytotoxicity was also determined to select the safe compounds for further study. Screening of 700 compounds resulted in the identification of two compounds (5228485 and 5220632) exhibiting an IC50 of 0.32 and 4.0 µg/ml respectively. Both compounds showed excellent anti-TB activity (MIC of 0.5-2.0 µg/ml against Mtb H37Rv) and low cytotoxicity in human cell line and sub-mitochondrial particles. The three-dimensional structure of M. tuberculosis ATPase was predicted using in-silico approach and docking studies were performed with the active compounds. The interaction between compounds and bacterial ATP synthase was confirmed by molecular docking analysis. In conclusion screening of compound library has resulted in the identification of two novel chemical scaffolds targeting mycobacterial ATP synthase.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/drug effects , Small Molecule Libraries , Adenosine Triphosphate/biosynthesis , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/metabolism , Bacterial Proton-Translocating ATPases/metabolism , Binding Sites , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Hep G2 Cells , Humans , Mice , Microbial Sensitivity Tests , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Conformation , Time Factors
8.
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
9.
Article in English | MEDLINE | ID: mdl-28807917

ABSTRACT

Pulmonary infections caused by Mycobacterium abscessus are emerging as a global threat, especially in cystic fibrosis patients. Further intensifying the concern of M. abscessus infection is the recent evidence of human-to-human transmission of the infection. M. abscessus is a naturally multidrug-resistant fast-growing pathogen for which pharmacological options are limited. Repurposing antitubercular drugs represents an attractive option for the development of chemotherapeutic alternatives against M. abscessus infections. Bedaquiline (BDQ), an ATP synthase inhibitor, has recently been approved for the treatment of multidrug-resistant tuberculosis. Herein, we show that BDQ has a very low MIC against a vast panel of clinical isolates. Despite being bacteriostatic in vitro, BDQ was highly efficacious in a zebrafish model of M. abscessus infection. Remarkably, a very short period of treatment was sufficient to protect the infected larvae from M. abscessus-induced killing. This was corroborated with reduced numbers of abscesses and cords, considered to be major pathophysiological signs in infected zebrafish. Mode-of-action studies revealed that BDQ triggered a rapid depletion of ATP in M. abscessusin vitro, consistent with the drug targeting the FoF1 ATP synthase. Importantly, despite a failure to select in vitro for spontaneous mutants that are highly resistant to BDQ, the transfer of single nucleotide polymorphisms leading to D29V or A64P substitutions in atpE conferred high resistance, thus resolving the target of BDQ in M. abscessus Overall, this study indicates that BDQ is active against M. abscessusin vitro and in vivo and should be considered for clinical use against the difficult-to-manage M. abscessus pulmonary infections.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proton-Translocating ATPases/antagonists & inhibitors , Diarylquinolines/pharmacology , Mycobacterium abscessus/drug effects , Adenosine Triphosphate/metabolism , Animals , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/metabolism , Polymorphism, Single Nucleotide , Zebrafish/microbiology
10.
Biochim Biophys Acta Biomembr ; 1859(6): 1124-1132, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28284722

ABSTRACT

Mitochondria, chloroplasts and photosynthetic bacteria are characterized by the presence of complex and intricate membrane systems. In contrast, non-photosynthetic bacteria lack membrane structures within their cytoplasm. However, large scale over-production of some membrane proteins, such as the fumarate reductase, the mannitol permease MtlA, the glycerol acyl transferase PlsB, the chemotaxis receptor Tsr or the ATP synthase subunit b, can induce the proliferation of intra cellular membranes (ICMs) in the cytoplasm of Escherichia coli. These ICMs are particularly rich in cardiolipin (CL). Here, we have studied the effect of CL in the generation of these membranous structures. We have deleted the three genes (clsA, clsB and clsC) responsible of CL biosynthesis in E. coli and analysed the effect of these mutations by fluorescent and electron microscopy and by lipid mass spectrometry. We have found that CL is essential in the formation of non-lamellar structures in the cytoplasm of E. coli cells. These results could help to understand the structuration of membranes in E. coli and other membrane organelles, such as mitochondria and ER.


Subject(s)
Bacterial Proteins/metabolism , Cardiolipins/metabolism , Endoplasmic Reticulum/metabolism , Escherichia coli/metabolism , Membrane Proteins/deficiency , Mitochondria/metabolism , Transferases (Other Substituted Phosphate Groups)/deficiency , Bacterial Proteins/genetics , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Endoplasmic Reticulum/ultrastructure , Escherichia coli/ultrastructure , Fluorescent Dyes/chemistry , Gene Deletion , Gene Expression , Isoenzymes/deficiency , Isoenzymes/genetics , Membrane Proteins/genetics , Mitochondria/ultrastructure , Time-Lapse Imaging , Transferases (Other Substituted Phosphate Groups)/genetics
11.
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
12.
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
13.
Genet Mol Res ; 15(3)2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27706660

ABSTRACT

Enterococcus faecalis is the major pathogen of post-endodontic disease and refractory periapical periodontitis, and recent research on this species has focused on its pathogenicity. E. faecalis most often causes disease in the form of a biofilm, and total protein expression shows a strong association with its virulence. Therefore, the purpose of our study was to explore different methods of extracting the total proteins of the E. faecalis (ATCC 33186 standard strain) biofilm. The total proteins in the biofilm were extracted using an ultrasonication method with varied parameters, including duration, amplitude setting, period, and duty cycle. After the optimal conditions of ultrasonication were determined based on the protein profile from sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis, the total protein content in the biofilm was detected using the bicinchoninic acid assay, Bradford Coomassie brilliant blue assay, and Lowry assay, and the results were compared and analyzed. The parameters for the optimal conditions of ultrasonication were as follows: a processing duration of 2 min, amplitude setting of 20%, and ultrasonication period of 4 s at a 50% duty cycle. The total protein content was 2299.1 mg/dish when measured by the bicinchoninic assay, 3793.8 mg/dish when measured by the Bradford Coomassie brilliant blue assay, and 1858.0 mg/dish when measured by the Lowry assay. These results demonstrate that the Bradford Coomassie brilliant blue assay is a simple and feasible method for use in detecting the total protein content in a bacterial biofilm.


Subject(s)
Bacterial Proton-Translocating ATPases/isolation & purification , Biofilms , Enterococcus faecalis/physiology , Bacterial Proton-Translocating ATPases/metabolism , Electrophoresis, Polyacrylamide Gel , Sonication
14.
J Biomol Struct Dyn ; 34(8): 1705-16, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26513379

ABSTRACT

Succinic acid is an important platform chemical with a variety of applications. Model-guided metabolic engineering strategies in Escherichia coli for strain improvement to increase succinic acid production using glucose and glycerol remain largely unexplored. Herein, we report what are, to our knowledge, the first metabolic knockout of the atpE gene to have increased succinic acid production using both glucose and alternative glycerol carbon sources in E. coli. Guided by a genome-scale metabolic model, we engineered the E. coli host to enhance anaerobic production of succinic acid by deleting the atpE gene, thereby generating additional reducing equivalents by blocking H(+) conduction across the mutant cell membrane. This strategy produced 1.58 and .49 g l(-1) of succinic acid from glycerol and glucose substrate, respectively. This work further elucidates a model-guided and/or system-based metabolic engineering, involving only a single-gene deletion strategy for enhanced succinic acid production in E. coli.


Subject(s)
Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Knockout Techniques , Models, Biological , Succinic Acid/metabolism , Fermentation , Glucose/metabolism , Glycerol/metabolism , Metabolic Networks and Pathways , Mutation
15.
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
16.
Sci Rep ; 4: 6951, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25377721

ABSTRACT

Propionic acid (PA) is an important platform chemical in the food, agriculture, and pharmaceutical industries and is mainly biosynthesized by propionibacteria. Acid tolerance in PA-producing strains is crucial. In previous work, we investigated the acid tolerance mechanism of Propionibacterium acidipropionici at microenvironmental levels by analyzing physiological changes in the parental strain and three PA-tolerant mutants obtained by genome shuffling. However, the molecular mechanism of PA tolerance in P. acidipropionici remained unclear. Here, we performed a comparative proteomics study of P. acidipropionici CGMCC 1.2230 and the acid-tolerant mutant P. acidipropionici WSH1105; MALDI-TOF/MS identified 24 proteins that significantly differed between the parental and shuffled strains. The differentially expressed proteins were mainly categorized as key components of crucial biological processes and the acid stress response. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) was used to confirm differential expression of nine key proteins. Overexpression of the secretory protein glyceraldehyde-3-phosphate dehydrogenase and ATP synthase subunit α in Escherichia coli BL21 improved PA and acetic acid tolerance; overexpression of NADH dehydrogenase and methylmalonyl-CoA epimerase improved PA tolerance. These results provide new insights into the acid tolerance of P. acidipropionici and will facilitate the development of PA production through fermentation by propionibacteria.


Subject(s)
Bacterial Proteins/genetics , DNA Shuffling/methods , Gene Expression Regulation, Bacterial , Propionates/metabolism , Propionibacterium/genetics , Proteomics , Acetic Acid/metabolism , Adaptation, Physiological , Bacterial Proteins/metabolism , Bacterial Proton-Translocating ATPases/genetics , Bacterial Proton-Translocating ATPases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fermentation , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Propionibacterium/metabolism , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Stress, Physiological , Transgenes
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...