Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 11(4): 420-8, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26662886

ABSTRACT

Vacuolar-type H(+) -ATPases (V-ATPases) have gained recent attention as highly promising anticancer drug targets, and therefore detailed structural analyses and studies of inhibitor interactions are very important research objectives. Spin labeling of the V-ATPase holoenzyme from the tobacco hornworm Manduca sexta and V-ATPase in isolated yeast (Saccharomyces cerevisiae) vacuoles was accomplished by two novel methods involving the covalent binding of a (2,2,6,6-tetramethylpiperidin-1-yl)oxyl (TEMPO) derivative of N,N'-dicyclohexylcarbodiimide (DCC) to the essential glutamate residue in the active site and the noncovalent interaction of a radical analogue of the highly potent inhibitor archazolid, a natural product from myxobacteria. Both complexes were evaluated in detail by electron paramagnetic resonance (EPR) spectroscopic studies and double electron-electron resonance (DEER) measurements, revealing insight into the inhibitor binding mode, dynamics, and stoichiometry as well as into the structure of the central functional subunit c of these medicinally important hetero-multimeric proton-translocating proteins. This study also demonstrates the usefulness of natural product derived spin labels as tools in medicinal chemistry.


Subject(s)
Dicyclohexylcarbodiimide/pharmacology , Electron Spin Resonance Spectroscopy , Enzyme Inhibitors/pharmacology , Macrolides/pharmacology , Manduca/enzymology , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Binding Sites , Cyclic N-Oxides/chemistry , Dicyclohexylcarbodiimide/chemistry , Enzyme Inhibitors/chemistry , Macrolides/chemistry , Manduca/drug effects , Models, Molecular , Spin Labels , Thiazoles/chemistry , Vacuolar Proton-Translocating ATPases/chemistry
2.
Structure ; 23(3): 461-471, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25661654

ABSTRACT

Vacuolar H(+)-ATPases are multisubunit complexes that operate with rotary mechanics and are essential for membrane proton transport throughout eukaryotes. Here we report a ∼ 1 nm resolution reconstruction of a V-ATPase in a different conformational state from that previously reported for a lower-resolution yeast model. The stator network of the V-ATPase (and by implication that of other rotary ATPases) does not change conformation in different catalytic states, and hence must be relatively rigid. We also demonstrate that a conserved bearing in the catalytic domain is electrostatic, contributing to the extraordinarily high efficiency of rotary ATPases. Analysis of the rotor axle/membrane pump interface suggests how rotary ATPases accommodate different c ring stoichiometries while maintaining high efficiency. The model provides evidence for a half channel in the proton pump, supporting theoretical models of ion translocation. Our refined model therefore provides new insights into the structure and mechanics of the V-ATPases.


Subject(s)
Insect Proteins/chemistry , Manduca/enzymology , Vacuolar Proton-Translocating ATPases/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Conserved Sequence , Cryoelectron Microscopy , Fourier Analysis , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Secondary
3.
J Biol Chem ; 290(6): 3680-92, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25505184

ABSTRACT

The vacuolar (H(+))-ATPases (V-ATPases) are a family of ATP-driven proton pumps that couple ATP hydrolysis with translocation of protons across membranes. Previous studies have implicated V-ATPases in cancer cell invasion. It has been proposed that V-ATPases participate in invasion by localizing to the plasma membrane and causing acidification of the extracellular space. To test this hypothesis, we utilized two separate approaches to specifically inhibit plasma membrane V-ATPases. First, we stably transfected highly invasive MDA-MB231 cells with a V5-tagged construct of the membrane-embedded c subunit of the V-ATPase, allowing for extracellular expression of the V5 epitope. We evaluated the effect of addition of a monoclonal antibody directed against the V5 epitope on both V-ATPase-mediated proton translocation across the plasma membrane and invasion using an in vitro Matrigel assay. The addition of anti-V5 antibody resulted in acidification of the cytosol and a decrease in V-ATPase-dependent proton flux across the plasma membrane in transfected but not control (untransfected) cells. These results demonstrate that the anti-V5 antibody inhibits activity of plasma membrane V-ATPases in transfected cells. Addition of the anti-V5 antibody also inhibited in vitro invasion of transfected (but not untransfected) cells. Second, we utilized a biotin-conjugated form of the specific V-ATPase inhibitor bafilomycin. When bound to streptavidin, this compound cannot cross the plasma membrane. Addition of this compound to MDA-MB231 cells also inhibited in vitro invasion. These studies suggest that plasma membrane V-ATPases play an important role in invasion of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Protons , Vacuolar Proton-Translocating ATPases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Ion Transport , Macrolides/pharmacology , Neoplasm Invasiveness , Protein Transport , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
4.
Biochem J ; 462(1): 185-97, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24805887

ABSTRACT

Primary active proton transport by eukaryotic V-ATPases (vacuolar ATPases) is regulated via the reversible disassembly of the V1Vo holoenzyme into its peripheral catalytic V1 complex and its membrane-bound proton-translocating Vo complex. This nutrient-dependent phenomenon had been first detected in the midgut epithelium of non-feeding moulting tobacco hornworms (Manduca sexta) and in glucose-deprived yeast cells (Saccharomyces cerevisiae). Since reversible disassembly to date had been investigated mostly in vitro, we wanted to test this phenomenon under in vivo conditions. We used living yeast cells with V-ATPase subunits fused to green, yellow or cyan fluorescent protein and found that only the V1 subunit C (Vma5) was released into the cytosol after substitution of extracellular glucose with galactose, whereas the other V1 subunits remained at or near the membrane. FRET analysis demonstrated close proximity between V1 and Vo even under glucose-starvation conditions. Disassembly, but not reassembly, depended on functional microtubules. Results from overlay blots, pull-down assays and bimolecular fluorescence complementation support the assumption that subunit C interacts directly with microtubules without involvement of linker proteins.


Subject(s)
Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Cytosol/metabolism , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae/enzymology
5.
J Biol Chem ; 289(23): 16399-408, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24795045

ABSTRACT

The vacuolar ATPase (V-ATPase) is a 1MDa transmembrane proton pump that operates via a rotary mechanism fuelled by ATP. Essential for eukaryotic cell homeostasis, it plays central roles in bone remodeling and tumor invasiveness, making it a key therapeutic target. Its importance in arthropod physiology also makes it a promising pesticide target. The major challenge in designing lead compounds against the V-ATPase is its ubiquitous nature, such that any therapeutic must be capable of targeting particular isoforms. Here, we have characterized the binding site on the V-ATPase of pea albumin 1b (PA1b), a small cystine knot protein that shows exquisitely selective inhibition of insect V-ATPases. Electron microscopy shows that PA1b binding occurs across a range of equivalent sites on the c ring of the membrane domain. In the presence of Mg·ATP, PA1b localizes to a single site, distant from subunit a, which is predicted to be the interface for other inhibitors. Photoaffinity labeling studies show radiolabeling of subunits c and e. In addition, weevil resistance to PA1b is correlated with bafilomycin resistance, caused by mutation of subunit c. The data indicate a binding site to which both subunits c and e contribute and inhibition that involves locking the c ring rotor to a static subunit e and not subunit a. This has implications for understanding the V-ATPase mechanism and that of inhibitors with therapeutic or pesticidal potential. It also provides the first evidence for the position of subunit e within the complex.


Subject(s)
Albumins/metabolism , Insecticides/metabolism , Pisum sativum/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Albumins/antagonists & inhibitors , Base Sequence , Benzophenones/metabolism , Binding Sites , Biotin/metabolism , DNA Primers , Insecticides/chemistry , Microscopy, Electron , Photoaffinity Labels , Protein Binding , Vacuolar Proton-Translocating ATPases/chemistry
6.
J Mol Biol ; 426(2): 286-300, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24075871

ABSTRACT

The vacuolar H(+)-ATPase (V-ATPase) is an ATP-driven proton pump essential to the function of eukaryotic cells. Its cytoplasmic V1 domain is an ATPase, normally coupled to membrane-bound proton pump Vo via a rotary mechanism. How these asymmetric motors are coupled remains poorly understood. Low energy status can trigger release of V1 from the membrane and curtail ATP hydrolysis. To investigate the molecular basis for these processes, we have carried out cryo-electron microscopy three-dimensional reconstruction of deactivated V1 from Manduca sexta. In the resulting model, three peripheral stalks that are parts of the mechanical stator of the V-ATPase are clearly resolved as unsupported filaments in the same conformations as in the holoenzyme. They are likely therefore to have inherent stiffness consistent with a role as flexible rods in buffering elastic power transmission between the domains of the V-ATPase. Inactivated V1 adopted a homogeneous resting state with one open active site adjacent to the stator filament normally linked to the H subunit. Although present at 1:1 stoichiometry with V1, both recombinant subunit C reconstituted with V1 and its endogenous subunit H were poorly resolved in three-dimensional reconstructions, suggesting structural heterogeneity in the region at the base of V1 that could indicate positional variability. If the position of H can vary, existing mechanistic models of deactivation in which it binds to and locks the axle of the V-ATPase rotary motor would need to be re-evaluated.


Subject(s)
Insect Proteins/metabolism , Insect Proteins/ultrastructure , Manduca/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/ultrastructure , Animals , Cryoelectron Microscopy , Imaging, Three-Dimensional , Models, Molecular , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism
7.
PLoS One ; 8(12): e82207, 2013.
Article in English | MEDLINE | ID: mdl-24312643

ABSTRACT

The V-ATPase is a membrane-bound protein complex which pumps protons across the membrane to generate a large proton motive force through the coupling of an ATP-driven 3-stroke rotary motor (V1) to a multistroke proton pump (Vo). This is done with near 100% efficiency, which is achieved in part by flexibility within the central rotor axle and stator connections, allowing the system to flex to minimise the free energy loss of conformational changes during catalysis. We have used electron microscopy to reveal distinctive bending along the V-ATPase complex, leading to angular displacement of the V1 domain relative to the Vo domain to a maximum of ~30°. This has been complemented by elastic network normal mode analysis that shows both flexing and twisting with the compliance being located in the rotor axle, stator filaments, or both. This study provides direct evidence of flexibility within the V-ATPase and by implication in related rotary ATPases, a feature predicted to be important for regulation and their high energetic efficiencies.


Subject(s)
Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cryoelectron Microscopy , Manduca/enzymology , Saccharomyces/enzymology , Vacuolar Proton-Translocating ATPases/ultrastructure
8.
Bioorg Med Chem Lett ; 22(24): 7735-8, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23122818

ABSTRACT

The water-solubility of the highly potent V-ATPase inhibitors archazolid A and the glucosylated derivative archazolid C was studied in the presence of a wide range of cosolvents, revealing very low solubilites. The first water-soluble analogue was then designed, synthesized, and evaluated for V-ATPase inhibitory activity in vitro.


Subject(s)
Macrolides/pharmacology , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Design , Macrolides/chemical synthesis , Macrolides/chemistry , Molecular Structure , Solubility , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Vacuolar Proton-Translocating ATPases/metabolism , Water/chemistry
9.
J Chem Inf Model ; 52(8): 2265-72, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22747331

ABSTRACT

Vacuolar ATPases are a potential therapeutic target because of their involvement in a variety of severe diseases such as osteoporosis or cancer. Archazolide A (1) and related analogs have been previously identified as selective inhibitors of V-ATPases with potency down to the subnanomolar range. Herein we report on the determination of the ligand binding mode by a combination of molecular docking, molecular dynamics simulations, and biochemical experiments, resulting in a sound model for the inhibitory mechanism of this class of putative anticancer agents. The binding site of archazolides was confirmed to be located in the equatorial region of the membrane-embedded V(O)-rotor, as recently proposed on the basis of site-directed mutagenesis. Quantification of the bioactivity of a series of archazolide derivatives, together with the docking-derived binding mode of archazolides to the V-ATPase, revealed favorable ligand profiles, which can guide the development of a simplified archazolide analog with potential therapeutic relevance.


Subject(s)
Macrolides/metabolism , Macrolides/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Thiazoles/metabolism , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Macrolides/chemistry , Mice , Protein Binding , Protein Structure, Secondary , Reproducibility of Results , Saccharomyces cerevisiae/enzymology , Substrate Specificity , Thiazoles/chemistry , Vacuolar Proton-Translocating ATPases/chemistry
10.
J Biol Chem ; 287(38): 31866-76, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22815478

ABSTRACT

The investigation of V-ATPases as potential therapeutic drug targets and hence of their specific inhibitors is a promising approach in osteoporosis and cancer treatment because the occurrence of these diseases is interrelated to the function of the V-ATPase. Apicularen belongs to the novel inhibitor family of the benzolactone enamides, which are highly potent but feature the unique characteristic of not inhibiting V-ATPases from fungal sources. In this study we specify, for the first time, the binding site of apicularen within the membrane spanning V(O) complex. By photoaffinity labeling using derivatives of apicularen and of the plecomacrolides bafilomycin and concanamycin, each coupled to (14)C-labeled 4-(3-trifluoromethyldiazirin-3-yl)benzoic acid, we verified that apicularen binds at the interface of the V(O) subunits a and c. The binding site is in the vicinity to those of the plecomacrolides and of the archazolids, a third family of V-ATPase inhibitors. Expression of subunit c homologues from Homo sapiens and Manduca sexta, both species sensitive to benzolactone enamides, in a Saccharomyces cerevisiae strain lacking the corresponding intrinsic gene did not transfer this sensitivity to yeast. Therefore, the binding site of benzolactone enamides cannot be formed exclusively by subunit c. Apparently, subunit a substantially contributes to the binding of the benzolactone enamides.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Enzyme Inhibitors/pharmacology , Macrolides/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Thiazoles/chemistry , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Binding Sites , Binding, Competitive , Macrolides/pharmacology , Manduca , Molecular Conformation , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/chemistry
11.
J Biol Chem ; 286(42): 36291-6, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21890633

ABSTRACT

PA1b (for pea albumin 1 subunit b) is a plant bioinsecticide lethal to several pests that are important in agriculture or human health. PA1b belongs to the inhibitory cystine knot family or knottin family. Originating from a plant (the garden pea) commonly eaten by humans without any known toxic or allergic effects, PA1b is a candidate for transgenic applications and is one of the most promising biopesticides for pest control. Using whole-cell patch-clamp techniques on Sf9 PA1b-sensitive lepidopteran insect cells, we discovered that PA1b reversibly blocked ramp membrane currents in a dose-dependent manner (EC(50) = 0.52 µM). PA1b had the same effect as bafilomycin, a specific inhibitor of the vacuolar proton pump (V-type H(+)-ATPase), and the PA1b-sensitive current depended on the internal proton concentration. Biochemical assays on purified V-ATPase from the lepidopteran model Manduca sexta showed that PA1b inhibited the V(1)V(0)-type H(+)-ATPase holoenzyme activity (IC(50) ∼ 70 nM) by interacting with the membrane-bound V(0) part of the V-ATPase. V-ATPase is a complex protein that has been studied increasingly because of its numerous physiological roles. In the midgut of insects, V-ATPase activity is essential for energizing nutrient absorption, and the results reported in this work explain the entomotoxic properties of PA1b. Targeting V-ATPase is a promising means of combating insect pests, and PA1b represents the first peptidic V-ATPase inhibitor. The search for V-ATPase inhibitors is currently of great importance because it has been demonstrated that V-ATPase plays a role in so many physiological processes.


Subject(s)
Cystine-Knot Miniproteins/pharmacology , Insect Proteins/antagonists & inhibitors , Insecticides/pharmacology , Manduca/enzymology , Plant Proteins/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Cell Line , Humans , Spodoptera/enzymology
12.
Eur J Cell Biol ; 90(9): 688-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21640428

ABSTRACT

V-ATPases in eukaryotes are heteromultimeric, H(+)-transporting proteins. They are localized in a multitude of different membranes and energize many different transport processes. Unique features of V-ATPases are, on the one hand, their ability to regulate enzymatic and ion transporting activity by the reversible dissociation of the catalytic V(1) complex from the membrane bound proton translocating V(0) complex and, on the other hand, their high sensitivity to specific macrolides such as bafilomycin and concanamycin from streptomycetes or archazolid and apicularen from myxomycetes. Both features require distinct intramolecular as well as intermolecular interactions. Here we will summarize our own results together with newer developments in both of these research areas.


Subject(s)
Vacuolar Proton-Translocating ATPases/metabolism , Humans , Protein Binding , Vacuoles/enzymology
13.
J Nat Prod ; 74(5): 1100-5, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21513292

ABSTRACT

Two structurally novel analogues of the macrolides archazolids A and B, archazolid A-15-O-ß-D-glucopyranoside (archazolid E, 5) and iso-archazolid B (archazolid F, 6), were isolated from the myxobacterium Cystobacter violaceus and Archangium gephyra, respectively. Macrolactone 5 represents the first 15-O-glycoside of the archazolids. iso-Archazolid B (6) incorporates a C-3 alkene and presents the first constitutional isomer reported for this natural product class. The structures of these polyketides were determined by spectroscopic analysis, in particular by HMBC, HMQC, and ROESY NMR investigations and by chemical degradation. iso-Archazolid B (6) demonstrated extremely high antiproliferative and V-ATPase inhibitory effects, with IC(50) values in the picomolar range, while only moderate activity was observed for glycoside 5. iso-Archazolid B presents the most potent archazolid known.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Glucosides/isolation & purification , Glucosides/pharmacology , Macrolides/isolation & purification , Macrolides/pharmacology , Myxococcales/chemistry , Thiazoles/isolation & purification , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Female , Glucosides/chemistry , Humans , Inhibitory Concentration 50 , Macrolides/chemistry , Male , Mice , Molecular Structure , Thiazoles/chemistry
14.
J Biol Chem ; 285(49): 38304-14, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20884613

ABSTRACT

The macrolactone archazolid is a novel, highly specific V-ATPase inhibitor with an IC(50) value in the low nanomolar range. The binding site of archazolid is presumed to overlap with the binding site of the established plecomacrolide V-ATPase inhibitors bafilomycin and concanamycin in subunit c of the membrane-integral V(O) complex. Using a semi-synthetic derivative of archazolid for photoaffinity labeling of the V(1)V(O) holoenzyme we confirmed binding of archazolid to the V(O) subunit c. For the plecomacrolide binding site a model has been published based on mutagenesis studies of the c subunit of Neurospora crassa, revealing 11 amino acids that are part of the binding pocket at the interface of two adjacent c subunits (Bowman, B. J., McCall, M. E., Baertsch, R., and Bowman, E. J. (2006) J. Biol. Chem. 281, 31885-31893). To investigate the contribution of these amino acids to the binding of archazolid, we established in Saccharomyces cerevisiae mutations that in N. crassa had changed the IC(50) value for bafilomycin 10-fold or more and showed that out of the amino acids forming the plecomacrolide binding pocket only one amino acid (tyrosine 142) contributes to the binding of archazolid. Using a fluorescent derivative of N,N'-dicyclohexylcarbodiimide, we found that the binding site for archazolid comprises the essential glutamate within helix 4 of subunit c. In conclusion the archazolid binding site resides within the equatorial region of the V(O) rotor subunit c. This hypothesis was supported by an additional subset of mutations within helix 4 that revealed that leucine 144 plays a role in archazolid binding.


Subject(s)
Enzyme Inhibitors/metabolism , Macrolides/metabolism , Saccharomyces cerevisiae/enzymology , Thiazoles/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Binding Sites , Enzyme Inhibitors/pharmacology , Macrolides/pharmacology , Mutation , Neurospora crassa/enzymology , Protein Binding/genetics , Protein Structure, Secondary , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
15.
J Exp Biol ; 212(Pt 11): 1611-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19448071

ABSTRACT

Active transepithelial cation transport in insects was initially discovered in Malpighian tubules, and was subsequently also found in other epithelia such as salivary glands, labial glands, midgut and sensory sensilla. Today it appears to be established that the cation pump is a two-component system of a H(+)-transporting V-ATPase and a cation/nH(+) antiporter. After tracing the discovery of the V-ATPase as the energizer of K(+)/nH(+) antiport in the larval midgut of the tobacco hornworm Manduca sexta we show that research on the tobacco hornworm V-ATPase delivered important findings that emerged to be of general significance for our knowledge of V-ATPases, which are ubiquitous and highly conserved proton pumps. We then discuss the V-ATPase in Malpighian tubules of the fruitfly Drosophila melanogaster where the potential of post-genomic biology has been impressively illustrated. Finally we review an integrated physiological approach in Malpighian tubules of the yellow fever mosquito Aedes aegypti which shows that the V-ATPase delivers the energy for both transcellular and paracellular ion transport.


Subject(s)
Epithelium/metabolism , Insecta/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Epithelium/ultrastructure , Gene Expression Regulation, Enzymologic , Vacuolar Proton-Translocating ATPases/chemistry , Vacuolar Proton-Translocating ATPases/genetics
16.
Am J Physiol Renal Physiol ; 296(4): F730-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19193723

ABSTRACT

The goal of this study was to identify and characterize the hypothesized apical cation/H(+) exchanger responsible for K(+) and/or Na(+) secretion in the renal (Malpighian) tubules of the yellow fever mosquito Aedes aegypti. From Aedes Malpighian tubules, we cloned "AeNHE8," a full-length cDNA encoding an ortholog of mammalian Na(+)/H(+) exchanger 8 (NHE8). The expression of AeNHE8 transcripts is ubiquitous among mosquito tissues and is not enriched in Malpighian tubules. Western blots of Malpighian tubules suggest that AeNHE8 is expressed primarily as an intracellular protein, which was confirmed by immunohistochemical localizations in Malpighian tubules. AeNHE8 immunoreactivity is expressed in principal cells of the secretory, distal segments, where it localizes to a subapical compartment (e.g., vesicles or endosomes), but not in the apical brush border. Furthermore, feeding mosquitoes a blood meal or treating isolated tubules with dibutyryl-cAMP, both of which stimulate a natriuresis by Malpighian tubules, do not influence the intracellular localization of AeNHE8 in principal cells. When expressed heterologously in Xenopus laevis oocytes, AeNHE8 mediates EIPA-sensitive Na/H exchange, in which Li(+) partially and K(+) poorly replace Na(+). The expression of AeNHE8 in Xenopus oocytes is associated with the development of a conductive pathway that closely resembles the known endogenous nonselective cation conductances of Xenopus oocytes. In conclusion, AeNHE8 does not mediate cation/H(+) exchange in the apical membrane of Aedes Malpighian tubules; it is more likely involved with an intracellular function.


Subject(s)
Aedes/metabolism , Insect Proteins/metabolism , Intracellular Membranes/metabolism , Malpighian Tubules/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium/metabolism , Yellow Fever/virology , Aedes/virology , Amino Acid Sequence , Animals , Blotting, Western , Cloning, Molecular , Computational Biology , Female , Hydrogen-Ion Concentration , Immunohistochemistry , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Intracellular Membranes/drug effects , Malpighian Tubules/drug effects , Membrane Potentials , Molecular Sequence Data , Oocytes , RNA, Messenger/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Xenopus laevis
17.
J Mol Biol ; 386(4): 989-99, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19244615

ABSTRACT

The vacuolar H+-ATPase (V-ATPase) is an ATP-driven rotary molecular motor that is a transmembrane proton pump in all eukaryotic cells. Although its activity is fundamental to many physiological processes, our understanding of the structure and mechanism of the V-ATPase is poor. Using cryo-electron microscopy of the tobacco hornworm (Manduca sexta) enzyme, we have calculated the first 3D reconstruction of the intact pump in its native state. The resolution of 16.5 A is significantly higher than that of previous cryo-electron microscopy models of either V-ATPase or the related F1F0-ATPase. A network of four stalk structures connecting the V1 catalytic domain and the V0 membrane domain is now fully resolved, demonstrating substantially greater complexity than that found in the F-ATPase. Three peripheral stator stalks connect these domains to a horizontal collar that partly encircles the region between V1 and V0. The fourth stalk is a central axle that connects to V0 but makes minimal contact with V1. Several subunit crystal structures can be fit accurately into the reconstruction. The model thus provides new insights into the organisation of key components involved in mechanical coupling between the domains and regulation of activity.


Subject(s)
Cryoelectron Microscopy , Manduca/enzymology , Molecular Motor Proteins/ultrastructure , Vacuolar Proton-Translocating ATPases/ultrastructure , Animals , Catalytic Domain , Image Processing, Computer-Assisted , Models, Molecular , Molecular Motor Proteins/isolation & purification , Protein Subunits/chemistry , Vacuolar Proton-Translocating ATPases/isolation & purification
18.
J Exp Biol ; 212(Pt 3): 341-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19151208

ABSTRACT

V-ATPases constitute a ubiquitous family of heteromultimeric, proton translocating proteins. According to their localization in a multitude of eukaryotic endomembranes and plasma membranes, they energize many different transport processes. Currently, a handful of specific inhibitors of the V-ATPase are known, which represent valuable tools for the characterization of transport processes on the level of tissues, single cells or even purified proteins. The understanding of how these inhibitors function may provide a basis to develop new drugs for the benefit of patients suffering from diseases such as osteoporosis or cancer. For this purpose, it appears absolutely essential to determine the exact inhibitor binding site in a target protein on the one side and to uncover the crucial structural elements of an inhibitor on the other side. However, even for some of the most popular and long known V-ATPase inhibitors, such as bafilomycin or concanamycin, the authentic structures of their binding sites are elusive. The aim of this review is to summarize the recent advances for the old players in the inhibition game, the plecomacrolides bafilomycin and concanamycin, and to introduce some of the new players, the macrolacton archazolid, the benzolactone enamides salicylihalamide, lobatamide, apicularen, oximidine and cruentaren, and the indolyls.


Subject(s)
Binding Sites/genetics , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Alkenes/chemistry , Alkenes/metabolism , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Lactones/chemistry , Lactones/metabolism , Macrolides/chemistry , Macrolides/metabolism , Molecular Structure , Salicylates/chemistry , Salicylates/metabolism , Thiazoles/chemistry , Thiazoles/metabolism
19.
FEBS Lett ; 581(29): 5566-72, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-17997985

ABSTRACT

Although the reversible dissociation of the V(1)V(O) holoenzyme into its V(1) and V(O) complexes is a general mechanism for the regulation of V-ATPases, important aspects are still not understood. By analyzing the endogenous nucleotide content of the V(1)V(O) holoenzyme and of the V(1) complex, both purified from Manduca sexta larval midgut, we found that the V(1) complex contained 1.7 molec. of ADP, whereas only 0.3 molec. of ADP were bound to the V(1)V(O) holoenzyme. By contrast, both proteins contained only negligible amounts of ATP. Incubation of the V(1)V(O) holoenzyme with various adenine nucleotides revealed that ATP hydrolysis, leading to a state containing tightly bound ADP is necessary for its dissociation.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Holoenzymes/chemistry , Holoenzymes/metabolism , Hydrolysis , Larva/enzymology , Manduca/enzymology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/chemistry
20.
FEBS Lett ; 581(18): 3523-7, 2007 Jul 24.
Article in English | MEDLINE | ID: mdl-17624334

ABSTRACT

Cruentaren A, a new antifungal benzolactone produced by the myxobacterium Byssovorax cruenta, proved to be highly cytotoxic against various human cell lines. It inhibited the proliferation of different cancer cell lines including a multidrug-resistant KB line at low nanomolar levels. It arrested human histocytic lymphoma cells (U-937) in G(0/1) phase, but did not trigger an apoptotic process. Studies to uncover the molecular target of cruentaren A showed that the novel compound, despite its structural similarity to the benzolactone enamides apicularen and salicylihalamide, was no V-ATPase inhibitor. In contrast, cruentaren specifically inhibited mitochondrial F(O)F(1)-ATPases with IC50 values of 15-30 nM. Although the exact binding site of cruentaren remains undefined, inhibition was shown to occur by interaction with the catalytic F(1) domain. Since mitochondrial ATPases play a crucial role in the pathophysiology of several human disorders including cancer, cruentaren or synthetic derivatives thereof could form the basis of future therapeutic strategies.


Subject(s)
Benzene/chemistry , Enzyme Inhibitors/toxicity , Macrolides/chemistry , Macrolides/toxicity , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/metabolism , Myxococcales/chemistry , Animals , Bridged Bicyclo Compounds, Heterocyclic/toxicity , Cattle , Cell Line , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Heart/drug effects , Humans , Kinetics , Macrolides/classification , Molecular Structure , Myocardium/enzymology , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...