Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Biotechnol Bioeng ; 120(3): 793-802, 2023 03.
Article in English | MEDLINE | ID: mdl-36510694

ABSTRACT

Nonribosomal peptides (NRPs) are a large family of secondary metabolites with notable bioactivities, which distribute widely in natural resources across microbes and plants. To obtain these molecules, heterologous production of NRPs in robust surrogate hosts like Escherichia coli represent a feasible approach. However, reconstitution of the full biosynthetic pathway in a host often leads to low productivity, which is at least in part due to the low efficiency of enzyme interaction in vivo except for the well-known reasons of metabolic burden (e.g., expression of large NRP synthetases-NRPSs with molecular weights of >100 kDa) and cellular toxicity on host cells. To enhance the catalytic efficiency of large NRPSs in vivo, here we propose to staple NRPS enzymes by using short peptide/protein pairs (e.g., SpyTag/SpyCatcher) for enhanced NRP production. We achieve this goal by introducing a stapled NRPS system for the biosynthesis of the antibiotic NRP valinomycin in E. coli. The results indicate that stapled valinomycin synthetase (Vlm1 and Vlm2) enables higher product accumulation than those two free enzymes (e.g., the maximum improvement is nearly fourfold). After further optimization by strain and bioprocess engineering, the final valinomycin titer maximally reaches about 2800 µg/L, which is 73 times higher than the initial titer of 38 µg/L. We expect that stapling NRPS enzymes will be a promising catalytic strategy for high-level biosynthesis of NRP natural products.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Valinomycin/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Biosynthetic Pathways , Peptide Synthases/genetics , Peptide Synthases/metabolism , Peptides/metabolism
2.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664570

ABSTRACT

A dodecadepsipeptide valinomycin (VLM) has been most recently reported to be a potential anti-coronavirus drug that could be efficiently produced on a large scale. It is thus of importance to study solid-phase forms of VLM in order to be able to ensure its polymorphic purity in drug formulations. The previously available solid-state NMR (SSNMR) data are combined with the plane-wave DFT computations in the NMR crystallography framework. Structural/spectroscopical predictions (the PBE functional/GIPAW method) are obtained to characterize four polymorphs of VLM. Interactions which confer a conformational stability to VLM molecules in these crystalline forms are described in detail. The way how various structural factors affect the values of SSNMR parameters is thoroughly analyzed, and several SSNMR markers of the respective VLM polymorphs are identified. The markers are connected to hydrogen bonding effects upon the corresponding (13C/15N/1H) isotropic chemical shifts of (CO, Namid, Hamid, Hα) VLM backbone nuclei. These results are expected to be crucial for polymorph control of VLM and in probing its interactions in dosage forms.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Valinomycin/chemistry , Betacoronavirus/chemistry , Betacoronavirus/isolation & purification , Betacoronavirus/metabolism , COVID-19 , Carbon Isotopes/chemistry , Coronavirus Infections/pathology , Coronavirus Infections/virology , Crystallography , Hydrogen Bonding , Nitrogen Isotopes/chemistry , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2 , Valinomycin/metabolism
3.
World J Microbiol Biotechnol ; 35(8): 128, 2019 Aug 02.
Article in English | MEDLINE | ID: mdl-31375920

ABSTRACT

Large patch disease, caused by Rhizoctonia solani AG2-2, is the most devastating disease in Zoysiagrass (Zoysia japonica). Current large patch disease control strategies rely primarily upon the use of chemical pesticides. Streptomyces sp. S8 is known to possess exceptional antagonistic properties that could potentially suppress the large patch pathogen found at turfgrass plantations. This study aims to demonstrate the feasibility of using the strain as a biological control mechanism. Sequencing of the S8 strain genome revealed a valinomycin biosynthesis gene cluster. This cluster is composed of the vlm1 and vlm2 genes, which are known to produce antifungal compounds. In order to verify this finding for the large patch pathogen, a valinomycin biosynthesis knockout mutant was created via the CRISPR/Cas9 system. The mutant lost antifungal activity against the large patch pathogen. Consequently, it is anticipated that eco-friendly microbial preparations derived from the S8 strain can be utilized to biologically control large patch disease.


Subject(s)
Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Rhizoctonia/drug effects , Streptomyces/metabolism , Valinomycin/metabolism , Valinomycin/pharmacology , Biosynthetic Pathways/genetics , Gene Knockout Techniques , Genome, Bacterial , Multigene Family , Pest Control, Biological/methods , Plant Diseases/microbiology , Plant Diseases/prevention & control , Poaceae/microbiology , Rhizoctonia/growth & development , Sequence Analysis, DNA , Streptomyces/genetics
4.
Chemphyschem ; 19(4): 359-366, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29124837

ABSTRACT

A molecular robot is a next-generation biochemical machine that imitates the actions of microorganisms. It is made of biomaterials such as DNA, proteins, and lipids. Three prerequisites have been proposed for the construction of such a robot: sensors, intelligence, and actuators. This Minireview focuses on recent research on synthetic ion channels and DNA computing technologies, which are viewed as potential candidate components of molecular robots. Synthetic ion channels, which are embedded in artificial cell membranes (lipid bilayers), sense ambient ions or chemicals and import them. These artificial sensors are useful components for molecular robots with bodies consisting of a lipid bilayer because they enable the interface between the inside and outside of the molecular robot to function as gates. After the signal molecules arrive inside the molecular robot, they can operate DNA logic gates, which perform computations. These functions will be integrated into the intelligence and sensor sections of molecular robots. Soon, these molecular machines will be able to be assembled to operate as a mass microrobot and play an active role in environmental monitoring and in vivo diagnosis or therapy.


Subject(s)
DNA/chemistry , Ion Channels/chemistry , Lipid Bilayers/chemistry , Robotics , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , DNA/metabolism , Humans , Ion Channels/chemical synthesis , Ion Channels/metabolism , MicroRNAs/analysis , Nanopores , Neoplasms/genetics , Neoplasms/pathology , Valinomycin/chemistry , Valinomycin/metabolism
5.
Indoor Air ; 27(1): 13-23, 2017 01.
Article in English | MEDLINE | ID: mdl-26806918

ABSTRACT

Indoor exposure to microbes and their structural and metabolic compounds is notoriously complex. To study proinflammatory interactions between the multiple microbial agents, macrophages derived from human THP-1 monocytic cells were exposed to several concentrations of microbial toxins alone (emodin, enniatin B, physcion, sterigmatocystin, valinomycin) and in combination with microbial structural components (bacterial lipopolysaccharide [LPS] or fungal ß-glucan). While the expression of proinflammatory cytokines TNFα and IL-1ß to single toxins alone was modest, low-dose co-exposure with structural components increased the responses of emodin, enniatin B, and valinomycin synergistically, both at the mRNA and protein level, as measured by RT-qPCR and ELISA, respectively. Co-exposure of toxins and ß-glucan resulted in consistent synergistically increased expression of several inflammation-related genes, while some of the responses with LPS were also inhibitory. Co-exposure of toxins with either ß-glucan or LPS induced also mitochondrial damage and autophagocytosis. The results demonstrate that microbial toxins together with bacterial and fungal structural components characteristic to moisture-damaged buildings can have drastic synergistic proinflammatory interactions at low exposure levels.


Subject(s)
Air Pollution, Indoor/analysis , Bacteria/metabolism , Fungi/metabolism , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Depsipeptides/metabolism , Emodin/analogs & derivatives , Emodin/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Lipopolysaccharides/metabolism , Macrophages/metabolism , Macrophages/microbiology , Real-Time Polymerase Chain Reaction , Sterigmatocystin/metabolism , THP-1 Cells , Valinomycin/metabolism , beta-Glucans/metabolism
6.
Chembiochem ; 16(2): 223-7, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25487354

ABSTRACT

Nonribosomal depsipeptides are a class of potent microbial natural products, which include several clinically approved pharmaceutical agents. Genome sequencing has revealed a large number of uninvestigated natural-product biosynthetic gene clusters. However, while novel informatic search methods to access these gene clusters have been developed to identify peptide natural products, depsipeptide detection has proven challenging. Herein, we present an improved version of our informatic search algorithm for natural products (iSNAP), which facilitates the detection of known and genetically predicted depsipeptides in complex microbial culture extracts. We validated this technology by identifying several depsipeptides from novel producers, and located a large number of novel depsipeptide gene clusters for future study. This approach highlights the value of chemoinformatic search methods for the discovery of genetically encoded metabolites by targeting specific areas of chemical space.


Subject(s)
Algorithms , Computational Biology/methods , Depsipeptides , Streptomyces/genetics , Streptomyces/metabolism , Biological Products , Computer Simulation , Depsipeptides/genetics , Genome, Bacterial , Markov Chains , Multigene Family , Tandem Mass Spectrometry , Valinomycin/metabolism
7.
Acc Chem Res ; 46(12): 2898-909, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-23514113

ABSTRACT

The natural product Valinomycin is a well-known transmembrane cation carrier. Despite being uncharged, this molecule can extract potassium ions from water without counterions and ferry them through a membrane interior. Because it only transports positive ions, it is electrogenic, mediating a flow of charge across the membrane. Equivalent agents for anions would be valuable research tools and may have therapeutic applications, especially in the treatment of "channelopathies" such as cystic fibrosis. However, no such molecules have been found in nature. In this Account, we describe our research toward synthetic and rationally designed "anti-Valinomycins". As our core approach to this problem, we used the steroid nucleus, provided by cholic acid, as a scaffold for the assembly of anion receptors. By positioning H-bond donors on this framework, especially urea and thiourea groups in conformationally constrained axial positions, we created binding sites capable of exceptionally high affinities (up to 10(11) M(-1) for R4N(+)Cl(-) in chloroform). The extended hydrocarbon surface of the steroid helped to maintain compatibility with nonpolar media. When we tested these "cholapods" for chloride transport in vesicles, they provided the first evidence for electrogenic anion transport mediated by electroneutral organic carriers: in other words, they are the first authenticated anti-Valinomycins. They also proved active in live cells that we grew and assayed in an Ussing chamber. In subsequent work, we have shown that the cholapods can exhibit very high activities, with transport observed down to carrier/lipid ratios of 1:250,000. We also understand some of the effects of structure on the activity of these molecules. For example, in most cases, powerful transporters also act as powerful receptors. On the other hand, some modifications which favor binding do not promote transport. We gained functional advantages by cyclizing the cholapod architecture, which encloses the anion binding site. We could also simplify the structure without compromising function. A steroid-inspired trans-decalin framework has proved highly effective and may lead to agents with practical advantages. Changing an ester side-chain in this system revealed a surprising effect, whereby increased length and/or lipophilicity resulted in substantially raised activity. Although much remains to be discovered about these anionophores, their high activities and intrinsic tuneabilities bode well for applications. In future work, we plan to develop and exploit these molecules as tools for biophysical research and to explore the possibility of useful biological activity.


Subject(s)
Drug Design , Models, Biological , Steroids/chemistry , Valinomycin/chemistry , Anions/chemistry , Drug Carriers/chemistry , Molecular Structure , Steroids/metabolism , Valinomycin/metabolism
8.
Toxicol In Vitro ; 83: 105407, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35659575

ABSTRACT

INTRODUCTION: Commercially-available resazurin-based reagents used for cell viability assessment contain varying amounts of resorufin; these may contribute to differences in autofluorescence, signal-to-background (S/B) ratio and the dynamic range of the assay. OBJECTIVES: This in vitro study compares the sensitivity of a new, high-sensitivity PrestoBlue (hs-PB) assay with standard PrestoBlue (PB) in assessing the efficacy of valinomycin and antimycin A in human vascular endothelial EA.hy926 cells, as well as cell viability. METHODS: The metabolic activity of EA.hy926 was evaluated based on resorufin fluorescence (PB assays) or formazan absorbance (MTT assay). RESULTS: The hs-PB assay demonstrated lower resorufin autofluorescence than the PB, resulting in a ≥ 1.4-fold increase in S/B ratio in hs-PB compared to PB. Valinomycin was more potent cytotoxic agent than antimycin A. The hs-PB, PB and MTT produced similar IC50 values for valinomycin. Antimycin A showed significantly higher potency in the MTT than in the resazurin-based assays. The EA.hy926 cells demonstrated higher metabolic activity in the presence of the antimycin A solvent - DMSO. CONCLUSION: All the examined methods may be used interchangeably to analyze drug cytotoxicity. Any differences in drug cytotoxicity observed between the assays may be due to relatively low drug potency and/or the influence of solvent on metabolism of assay reagent. The hs-PB assay appears to more effectively detect cell viability and produce a stronger signal than its conventional counterpart.


Subject(s)
Endothelial Cells , Antimycin A/metabolism , Antimycin A/toxicity , Cell Survival , Humans , Indicators and Reagents/pharmacology , Solvents/pharmacology , Valinomycin/metabolism , Valinomycin/pharmacology
9.
J Biomol Struct Dyn ; 40(17): 8018-8029, 2022 10.
Article in English | MEDLINE | ID: mdl-33826484

ABSTRACT

Interaction between the SARS-COV-2 (2019 novel coronavirus) spike protein and ACE2 receptors expressed on cellular surfaces initialises viral attachment and consequent infection. Blocking this interaction shows promise for blocking or ameliorating the virus' pathological effects on the body. By contrast to work focusing on the coronavirus, which has significant potential diversity through possible accumulation of mutations during transmission, targeting the conserved ACE2 protein expressed on human cells offers an attractive alternative route to developing pharmacological prophylactics against viral invasion. In this study, we screened a virtual database of natural peptides in silico, with ACE2 as the target, and performed structural analyses of the interface region in the SARS-COV-2 RBD/ACE2 complex. These analyses have identified 15 potentially effective compounds. Analyses of ACE2/polypeptide interactions suggest that these peptides can block viral invasion of cells by stably binding in the ACE2 active site pocket. Molecular simulation results for Complestatin and Valinomycin indicate that they may share this mechanism. The discovery of this probable binding mechanism provides a frame of reference for further optimization, and design of high affinity ACE2 inhibitors that could serve as leads for production of drugs with preventive and therapeutic effects against SARS-COV-2. Communicated by Ramaswamy H. Sarma.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Humans , Peptides/metabolism , Peptides/pharmacology , Peptidyl-Dipeptidase A/chemistry , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Valinomycin/metabolism
10.
Biochim Biophys Acta Bioenerg ; 1863(8): 148908, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35961396

ABSTRACT

Mitochondria play an important role not only in producing energy for the cell but also for regulating mitochondrial and cell function depending on the cell's needs and environment. Uptake of cations, anions, and substrates requires a stable, polarized transmembrane charge potential (ΔΨm). Chemiosmosis requires ion exchangers to remove Na+, K+, Ca2+, PO43-, and other charged species that enter mitochondria. Knowledge of the kinetics of mitochondrial (m) cation channels and exchangers is important in understanding their roles in regulating mitochondrial chemiosmosis and bioenergetics. The influx/efflux of K+, the most abundant mitochondrial cation, alters mitochondrial volume and shape by bringing in anions and H2O by osmosis. The effects of K+ uptake through ligand-specific mK+ channels stimulated/inhibited by agonists/antagonists on mitochondrial volume (swelling/contraction) are well known. However, a more important role for K+ influx is likely its effects on H+ cycling and bioenergetics facilitated by mitochondrial (m) K+/H+ exchange (mKHE), though the kinetics and consequences of K+ efflux by KHE are not well described. We hypothesized that a major role of K+ influx/efflux is stimulation of respiration via the influx of H+ by KHE. We proposed to modulate KHE activity by energizing guinea pig heart isolated mitochondria and by altering the mK+ cycle to capture changes in mitochondrial volume, pHm, ΔΨm, and respiration that would reflect a role for H+ influx via KHE to regulate bioenergetics. To test this, mitochondria were suspended in a 150 mM K+ buffer at pH 6.9, or in a 140 mM Cs+ buffer at pH 7.6 or 6.9 with added 10 mM K+, minimal Ca2+ and free of Na+. O2 content was measured by a Clark electrode, and pHm, ΔΨm, and volume, were measured by fluorescence spectrophotometry and light-scattering. Adding pyruvic acid (PA) alone caused increases in volume and respiration and a rapid decrease in the transmembrane pH gradient (ΔpHm = pHin-pHext) at pHext 6.9> > 7.6, so that ΔΨm was charged and maintained. BKCa agonist NS1619 and antagonist paxilline modified these effects, and KHE inhibitor quinine and K+ ionophore valinomycin depolarized ΔΨm. We postulate that K+ efflux-induced H+ influx via KHE causes an inward H+ leak that stimulates respiration, but at buffer pH 6.9 also utilizes the energy of ΔpHm, the smaller component of the overall proton motive force, ΔµH+. Thus ΔpHm establishes and maintains the ΔΨm required for utilization of substrates, entry of all cations, and for oxidative phosphorylation. Thus, K+ influx/efflux appears to play a pivotal role in regulating energetics while maintaining mitochondrial ionic balance and volume homeostasis.


Subject(s)
Pyruvic Acid , Quinine , Animals , Anions/metabolism , Energy Metabolism , Guinea Pigs , Hydrogen-Ion Concentration , Ionophores/metabolism , Ionophores/pharmacology , Ligands , Mitochondria, Heart/metabolism , Potassium/metabolism , Pyruvic Acid/metabolism , Pyruvic Acid/pharmacology , Quinine/metabolism , Quinine/pharmacology , Valinomycin/metabolism , Valinomycin/pharmacology
11.
Biophys J ; 100(6): 1542-9, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21402037

ABSTRACT

In studying ion-selectivity in biomaterials, it is common to study ion-protein interactions within a local neighborhood around the ion. This local system analysis for the S(2) site of KcsA, its semisynthetic analog, and valinomycin yields the free energy change in exchanging K(+) with Na(+) in quantitative agreement with the value obtained by considering ion-interactions with the entire system. But the energetics of ion binding in the local system and in the entire system differ significantly and lead to different conclusions regarding the physical basis of ion selectivity. For configurations sampled from an all-atom simulation, we show that the selectivity free energy can be decomposed into a contribution arising from interactions of the ion with its local neighborhood, ΔW(local), and a term arising from the field imposed on the ion and the binding site by the rest of the medium, ΔW(ϕ). The local contribution ΔW(local) is numerically close to the actual free energy difference because the field contribution is small. The field contribution is small because of cancellation of inversely related ion-medium and site-medium interactions. Our analysis presents a rigorous foundation for the numerical success of the local system analysis and shows that its implications do not always hold for the entire protein.


Subject(s)
Molecular Dynamics Simulation , Potassium Channels/chemistry , Potassium Channels/metabolism , Valinomycin/metabolism , Binding Sites , Mutation , Potassium Channels/genetics , Protein Binding , Protein Conformation , Thermodynamics , Valinomycin/chemistry
12.
Biophys J ; 98(12): 2904-13, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20550903

ABSTRACT

It was recently demonstrated that significant local deformations of biological membranes take place due to the fields of charged peptides and ions, challenging the standard model of membrane electrostatics. The ability of ions to retain their immediate hydration environment, combined with the lack of sensitivity of permeability to ion type or even ion pairs, led us to question the extent to which hydration energetics and electrostatics control membrane ion permeation. Using the arginine analog methyl-guanidinium as a test case, we find that although hydrocarbon electronic polarizability causes dramatic changes in ion solvation free energy, as well as a significant change (approximately 0.4 V) in the membrane dipole potential, little change in membrane permeation energetics occurs. We attribute this to compensation of solvation terms from polar and polarizable nonpolar components within the membrane, and explain why the dipole potential is not fully sensed in terms of the locally deformed bilayer interface. Our descriptions provide a deeper understanding of the translocation process and allow predictions for poly-ions, ion pairs, charged lipids, and lipid flip-flop. We also report simulations of large hydrophobic-ion-like membrane defects and the ionophore valinomycin, which exhibit little membrane deformation, as well as hydrophilic defects and the ion channel gramicidin A, to provide parallels to membranes deformed by unassisted ion permeation.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Static Electricity , Cell Membrane/metabolism , Gramicidin/chemistry , Gramicidin/metabolism , Hydrophobic and Hydrophilic Interactions , Ion Channels/chemistry , Ion Channels/metabolism , Lipid Bilayers/metabolism , Molecular Dynamics Simulation , Porosity , Protein Conformation , Valinomycin/metabolism
13.
J Biol Chem ; 284(50): 35073-8, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19843525

ABSTRACT

The vesicular inhibitory amino acid transporter (VIAAT) is a synaptic vesicle protein responsible for the vesicular storage of gamma-aminobutyrate (GABA) and glycine which plays an essential role in GABAergic and glycinergic neurotransmission. The transport mechanism of VIAAT remains largely unknown. Here, we show that proteoliposomes containing purified VIAAT actively took up GABA upon formation of membrane potential (Deltapsi) (positive inside) but not DeltapH. VIAAT-mediated GABA uptake had an absolute requirement for Cl(-) and actually accompanied Cl(-) movement. Kinetic analysis indicated that one GABA molecule and two Cl(-) equivalents were transported during one transport cycle. VIAAT in which Glu(213) was specifically mutated to alanine completely lost the ability to take up both GABA and Cl(-). Essentially the same results were obtained with glycine, another substrate of VIAAT. These results demonstrated that VIAAT is a vesicular Cl(-) transporter that co-transports Cl(-) with GABA or glycine in a Deltapsi dependent manner. It is concluded that Cl(-) plays an essential role in vesicular storage of GABA and glycine.


Subject(s)
Chlorides/metabolism , Symporters/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Glycine/metabolism , Ionophores/metabolism , Liposomes/metabolism , Membrane Potentials/physiology , Rats , Symporters/genetics , Valinomycin/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics
14.
Appl Environ Microbiol ; 76(23): 7683-90, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20889782

ABSTRACT

An oxalate-fermenting brown rot fungus, Fomitopsis palustris, secretes large amounts of oxalic acid during wood decay. Secretion of oxalic acid is indispensable for the degradation of wood cell walls, but almost nothing is known about the transport mechanism by which oxalic acid is secreted from F. palustris hyphal cells. We characterized the mechanism for oxalate transport using membrane vesicles of F. palustris. Oxalate transport in F. palustris was ATP dependent and was strongly inhibited by several inhibitors, such as valinomycin and NH(4)(+), suggesting the presence of a secondary oxalate transporter in this fungus. We then isolated a cDNA, FpOAR (Fomitopsis palustris oxalic acid resistance), from F. palustris by functional screening of yeast transformants with cDNAs grown on oxalic acid-containing plates. FpOAR is predicted to be a membrane protein that possesses six transmembrane domains but shows no similarity with known oxalate transporters. The yeast transformant possessing FpOAR (FpOAR-transformant) acquired resistance to oxalic acid and contained less oxalate than the control transformant. Biochemical analyses using membrane vesicles of the FpOAR-transformant showed that the oxalate transport property of FpOAR was consistent with that observed in membrane vesicles of F. palustris. The quantity of FpOAR transcripts was correlated with increasing oxalic acid accumulation in the culture medium and was induced when exogenous oxalate was added to the medium. These results strongly suggest that FpOAR plays an important role in wood decay by acting as a secondary transporter responsible for secretion of oxalate by F. palustris.


Subject(s)
Coriolaceae/enzymology , Coriolaceae/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Oxalates/metabolism , Adenosine Triphosphate/metabolism , Cluster Analysis , Coriolaceae/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , DNA, Fungal/chemistry , DNA, Fungal/genetics , Enzyme Inhibitors/metabolism , Gene Expression Profiling , Molecular Sequence Data , Phylogeny , Quaternary Ammonium Compounds/metabolism , Secretory Vesicles/enzymology , Sequence Analysis, DNA , Sequence Homology , Valinomycin/metabolism , Wood/metabolism , Wood/microbiology
15.
Biochim Biophys Acta ; 1777(10): 1301-10, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18619941

ABSTRACT

The membrane-embedded F(0) part of ATP synthases is responsible for ion translocation during ATP synthesis and hydrolysis. Here, we describe an in vitro system for measuring proton fluxes through F(0) complexes by fluorescence changes of the entrapped fluorophore pyranine. Starting from purified enzyme, the F(0) part was incorporated unidirectionally into phospholipid vesicles. This allowed analysis of proton transport in either synthesis or hydrolysis direction with Deltapsi or DeltapH as driving forces. The system displayed a high signal-to-noise ratio and can be accurately quantified. In contrast to ATP synthesis in the Escherichia coli F(1)F(0) holoenzyme, no significant difference was observed in the efficiency of DeltapH or Deltapsi as driving forces for H(+)-transport through F(0). Transport rates showed linear dependency on the driving force. Proton transport in hydrolysis direction was about 2400 H(+)/(s x F(0)) at Deltapsi of 120 mV, which is approximately twice as fast as in synthesis direction. The chloroplast enzyme was faster and catalyzed H(+)-transport at initial rates of 6300 H(+)/(s x F(0)) under similar conditions. The new method is an ideal tool for detailed kinetic investigations of the ion transport mechanism of ATP synthases from various organisms.


Subject(s)
Ion Transport/physiology , Membrane Potential, Mitochondrial/physiology , Mitochondrial Proton-Translocating ATPases , Proton-Motive Force , Protons , Chloroplasts/metabolism , Escherichia coli/cytology , Escherichia coli/enzymology , Ionophores/metabolism , Liposomes/chemistry , Liposomes/metabolism , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Spinacia oleracea/cytology , Trialkyltin Compounds/metabolism , Valinomycin/metabolism
16.
Biochim Biophys Acta ; 1709(3): 195-202, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16139788

ABSTRACT

New and old data pertinent to the electrochemical potentials across the inner mitochondrial membrane are reviewed with the intent of reconciling the various findings in the light of new perspectives provided by more recent knowledge. A careful scrutiny of old data permits ruling out the presence of a significant metabolically dependent electrical membrane potential. Recent technological advances make it possible to test the proposed alternatives. These proposals recast the original idea, and the possible mechanisms that are emerging also invoke a protonmotive force. Our conclusions that DeltaPsi is not involved in oxidative-phosphorylation finds parallel observations in Halobacterium halobium [H. Michel, D. Oesterhelt, Electrochemical proton gradient across the cell membrane of Halobacterium halobium: comparison of the light-induced increase with the increase of intracellular adenosine triphosphate under steady-state illumination, Biochemistry 19 (1980) 4615-4619] and thylakoid vesicles [D.R. Ort, R.A. Dilley, N.E. Good, Photophosphorylation as a function of illumination time II. Effects of permeant buffers, Biochim. Biophys. Acta 449 (1976) 108-129] in which light-induced ATP synthesis occurs in the absence of an apparent DeltaPsi or DeltapH, suggesting the presence of mechanisms similar to the one proposed for mitochondria.


Subject(s)
Energy Metabolism/physiology , Mitochondrial Membranes/physiology , Models, Chemical , Proton-Motive Force/physiology , Membrane Potentials/physiology , Mitochondrial Membranes/metabolism , Phosphorylation , Potassium/metabolism , Valinomycin/metabolism
17.
In Vitro Cell Dev Biol Anim ; 42(1-2): 45-9, 2006.
Article in English | MEDLINE | ID: mdl-16618212

ABSTRACT

The mammalian intestinal epithelium has been found, based on in vivo experiments, to be resistant to insecticidal Cry toxins, which are derived from Bacillus thuringiensis and fatally damage insect midgut cells. Thus, the toxins are commonly used as a genetic resource in insect-resistant transgenic plants for feed. However, Cry toxins bind to the cellular brush border membrane vesicle (BBMV) of mammalian intestinal cells. In this study, we investigated the affinity of Cry1Ab toxin, a lepidopteran-specific Cry1-type toxin, to the cellular BBMV of two mammalian intestinal cells as well as the effect of the toxin on the membrane potential of three mammalian intestinal cells compared to its effects on the silkworm midgut cell. We found that Cry1Ab toxin did bind to the bovine and porcine BBMV, but far more weakly than it did to the silkworm midgut BBMV. Furthermore, although the silkworm midgut cells developed severe membrane potential changes within 1 h following the toxin treatment at a final concentration of 2 mug/ml, no such membraneous changes were observed on the bovine, porcine, and human intestinal cells. The present in vitro results suggest that, although Cry1Ab toxin may bind weakly or nonspecifically to certain BBMV components in the mammalian intestinal cell, it does not damage the cell's membrane integrity, thus exerting no subsequent adverse effects on the cell.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Endotoxins/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/cytology , Animals , Bacillus thuringiensis Toxins , Bombyx , Cattle , Cells, Cultured , Epithelial Cells/cytology , Hemolysin Proteins , Humans , In Vitro Techniques , Ionophores/metabolism , Membrane Potentials/physiology , Swine , Valinomycin/metabolism
18.
Biochim Biophys Acta ; 1283(1): 1-3, 1996 Aug 14.
Article in English | MEDLINE | ID: mdl-8765086

ABSTRACT

When the thickness of monolayer membranes formed by bolaform archaeal lipids is reduced to the approximate length of two valinomycin molecules, the zero-current conductance does not show any more a linear dependence on valinomycin concentration; instead, a quadratic behaviour is observed. This suggests that a dimer permeation pore is formed and therefore the conduction mechanism changes from carrier to channel.


Subject(s)
Ion Channels , Membranes, Artificial , Valinomycin/metabolism , Alkanes , Chloroform , Diglycerides/chemistry , Diglycerides/metabolism , Electric Conductivity , Lipid Bilayers/metabolism
19.
Biochim Biophys Acta ; 1237(2): 121-6, 1995 Jul 26.
Article in English | MEDLINE | ID: mdl-7632704

ABSTRACT

The present study demonstrates that the permanently positively charged, lipid-conjugated rhodamine, R18, can be transported from the outer to the inner leaflet of lipid bilayers in response of a transmembrane potential (negative inside). This conclusion was based on the following observations. (i) A fast decrease of the R18 fluorescence, when present at self-quenching concentrations in DOPC large unilamellar vesicles, was revealed upon induction of a valinomycin-induced K(+)-diffusion potential. (ii) Iodide quenching experiments demonstrated that R18 was no longer accessible to externally added aqueous quencher after application of a transmembrane potential. (iii) 2H-NMR measurements, using DOPC, specifically deuterated at the alpha-position of the phosphocholine head group, revealed a massive transbilayer movement of R18 upon induction of a membrane potential. The extent of the fluorescence changes were found to be dependent on the magnitude of the applied transmembrane potential, which opens possibilities for novel applications of R18 as an internal lipid-conjugated membrane potential probe.


Subject(s)
Lipids/chemistry , Membrane Potentials , Rhodamines/metabolism , Biological Transport , Magnetic Resonance Spectroscopy , Potassium/metabolism , Rhodamines/chemistry , Sodium/metabolism , Spectrometry, Fluorescence , Valinomycin/metabolism
20.
Biochim Biophys Acta ; 1323(1): 137-44, 1997 Jan 14.
Article in English | MEDLINE | ID: mdl-9030220

ABSTRACT

The studies on the decay of the pH difference, delta pH, across soyabean phospholipid vesicular membrane have shown that the rates of net proton transport and the associated Li+ and Na+ ion transport across the membrane can be enhanced by the combined action of gramicidin, valinomycin and carbonyl cyanide m-chlorophenylhydrazone (CCCP) in K(+)-free vesicle solutions. The data obtained under different experimental conditions suggest that this enhancement is a consequence of facilitation of CCCP- transport (1) by complexing CCCP- with the highly membrane permeant valinomycin without the metal ion bound to it and (2) by the associated Li+ or Na+ transport through the gramicidin channel such that no net charge is transported across the membrane. The dissociation constant of the weak valinomycin-CCCP- complex has been estimated to be > 200 mM in the membrane. The delta pH in these experiments were created by temperature jump.


Subject(s)
Carbonyl Cyanide m-Chlorophenyl Hydrazone/metabolism , Gramicidin/metabolism , Membrane Lipids/metabolism , Phospholipids/metabolism , Valinomycin/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Ion Transport , Lithium/metabolism , Protons , Sodium/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL