Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Comput Biol Chem ; 77: 272-278, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30396154

ABSTRACT

Organophosphate compounds bioremediation by use of organophosphorus degradation enzymes such as DFPase is a developing interest in industry and medicine. The most important problem with the bio-catalytic enzymes is their instability on high temperatures. This work carried out to find suitable locations for introducing disulfide bridges in DFPase enzyme. We employed some computational approaches to design the disulfide bridges and evaluate their roles in the enzyme structural thermostability. According to the in silico results, mutant 6 (V24C, C76) increased the enzyme thermostability relative to wild-type.


Subject(s)
Loligo/enzymology , Phosphoric Triester Hydrolases/chemistry , Animals , Catalytic Domain , Databases, Protein , Disulfides/chemistry , Enzyme Stability , Hot Temperature , Loligo/chemistry , Loligo/genetics , Molecular Dynamics Simulation , Phosphoric Triester Hydrolases/genetics , Point Mutation , Protein Conformation
2.
J Phys Chem B ; 118(17): 4479-89, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24720808

ABSTRACT

Organophosphorus (OP) nerve agents such as (S)-sarin are among the most highly toxic compounds that have been synthesized. Engineering enzymes that catalyze the hydrolysis of nerve agents ("bioscavengers") is an emerging prophylactic approach to diminish their toxic effects. Although its native function is not known, diisopropyl fluorophosphatase (DFPase) from Loligo vulgaris catalyzes the hydrolysis of OP compounds. Here, we investigate the mechanisms of diisopropylfluorophosphate (DFP) and (S)-sarin hydrolysis by DFPase with quantum mechanical/molecular mechanical umbrella sampling simulations. We find that the mechanism for hydrolysis of DFP involves nucleophilic attack by Asp229 on phosphorus to form a pentavalent intermediate. P-F bond dissociation then yields a phosphoacyl enzyme intermediate in the rate-limiting step. The simulations suggest that a water molecule, coordinated to the catalytic Ca(2+), donates a proton to Asp121 and then attacks the tetrahedral phosphoacyl intermediate to liberate the diisopropylphosphate product. In contrast, the calculated free energy barrier for hydrolysis of (S)-sarin by the same mechanism is highly unfavorable, primarily because of the instability of the pentavalent phosphoenzyme species. Instead, simulations suggest that hydrolysis of (S)-sarin proceeds by a mechanism in which Asp229 could activate an intervening water molecule for nucleophilic attack on the substrate. These findings may lead to improved strategies for engineering DFPase and related six-bladed ß-propeller folds for more efficient degradation of OP compounds.


Subject(s)
Chemical Warfare Agents/metabolism , Isoflurophate/metabolism , Phosphoric Triester Hydrolases/metabolism , Protein Engineering , Sarin/metabolism , Animals , Hydrolysis , Loligo/enzymology , Models, Molecular , Phosphoric Triester Hydrolases/chemistry , Phosphoric Triester Hydrolases/genetics , Protein Conformation , Thermodynamics
3.
Nanotechnology ; 23(29): 294009, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22743846

ABSTRACT

In the advancement of green syntheses and sustainable reactions, enzymatic biocatalysis offers extremely high reaction rates and selectivity that goes far beyond the reach of chemical catalysts; however, these enzymes suffer from typical environmental constraints, e.g. operational temperature, pH and tolerance to oxidative environments. A common hydrolase enzyme, diisopropylfluorophosphatase (DFPase, EC 3.1.8.2), has demonstrated a pronounced efficacy for the hydrolysis of a variety of substrates for potential toxin remediation, but suffers from the aforementioned limitations. As a means to enhance DFPase's stability in oxidative environments, enzymatic covalent immobilization within the polymeric matrix of poly(propylene sulfide) (PPS) nanoparticles was performed. By modifying the enzyme's exposed lysine residues via thiolation, DFPase is utilized as a comonomer/crosslinker in a mild emulsion polymerization. The resultant polymeric polysulfide shell acts as a 'sacrificial barrier' by first oxidizing to polysulfoxides and polysulfones, rendering DFPase in an active state. DFPase-PPS nanoparticles thus retain activity upon exposure to as high as 50 parts per million (ppm) of hypochlorous acid (HOCl), while native DFPase is observed as inactive at 500 parts per billion (ppb). This trend is also confirmed by enzyme-generated (chloroperoxidase (CPO), EC 1.11.1.10) reactive oxygen species (ROS) including both HOCl (3 ppm) and ClO(2) (100 ppm).


Subject(s)
Enzymes, Immobilized/chemistry , Loligo/enzymology , Nanoparticles/chemistry , Phosphoric Triester Hydrolases/chemistry , Polymers/chemistry , Sulfides/chemistry , Animals , Chlorine Compounds/metabolism , Enzyme Stability , Enzymes, Immobilized/metabolism , Loligo/chemistry , Models, Molecular , Oxides/metabolism , Phosphoric Triester Hydrolases/metabolism , Polymerization , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/chemistry
4.
Drug Test Anal ; 4(3-4): 262-70, 2012.
Article in English | MEDLINE | ID: mdl-22174192

ABSTRACT

Highly toxic organophosphorus compounds that irreversibly inhibit the enzyme acetycholinesterase (AChE), including nerve agents like tabun, sarin, or soman, still pose a credible threat to civilian populations and military personnel. New therapeutics that can be used as a pretreatment or after poisoning with these compounds, complementing existing treatment schemes such as the use of atropine and AChE reactivating oximes, are currently the subject of intense research. A prominent role among potential candidates is taken by enzymes that can detoxify nerve agents by hydrolysis. Diisopropyl fluorophosphatase (DFPase) from the squid Loligo vulgaris is known to effectively hydrolyze DFP and the range of G-type nerve agents including sarin and soman. In the present work, DFPase was PEGylated to increase biological half-life, and to lower or avoid an immunogenic reaction and proteolytic digest. Addition of linear polyethylene glycol (PEG) chains was achieved using mPEG-NHS esters and conjugates were characterized by electrospray ionization--time of flight--mass specrometry (ESI-ToF-MS). PEGylated wildtype DFPase and a mutant selective for the more toxic stereoisomers of the agents were tested in vivo with rats that were challenged with a subcutaneous 3x LD(50) dose of soman. While wildtype DFPase prevented death only at extremely high doses, the mutant was able keep the animals alive and to minimize or totally avoid symptoms of poisoning. The results serve as a proof of principle that engineered variants of DFPase are potential candidates for in vivo use if substrate affinity can be improved or the turnover rate enhanced to lower the required enzyme dose.


Subject(s)
Antidotes/therapeutic use , Chemical Warfare Agents/poisoning , Cholinesterase Inhibitors/poisoning , Loligo/enzymology , Phosphoric Triester Hydrolases/therapeutic use , Soman/poisoning , Animals , Antidotes/chemistry , Loligo/genetics , Male , Mass Spectrometry , Mutation , Phosphoric Triester Hydrolases/chemistry , Phosphoric Triester Hydrolases/genetics , Polyethylene Glycols/chemistry , Rats , Rats, Wistar
5.
Nat Struct Mol Biol ; 18(9): 999-1005, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822282

ABSTRACT

The enzyme phospholipase C-ß (PLCß) is a crucial regulator of intracellular calcium levels whose activity is controlled by heptahelical receptors that couple to members of the Gq family of heterotrimeric G proteins. We have determined atomic structures of two invertebrate homologs of PLCß (PLC21) from cephalopod retina and identified a helix from the C-terminal regulatory region that interacts with a conserved surface of the catalytic core of the enzyme. Mutations designed to disrupt the analogous interaction in human PLCß3 considerably increase basal activity and diminish stimulation by Gαq. Gαq binding requires displacement of the autoinhibitory helix from the catalytic core, thus providing an allosteric mechanism for activation of PLCß.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Loligo/enzymology , Phospholipase C beta/chemistry , Sepia/enzymology , Animals , Crystallography, X-Ray , Models, Molecular , Mutagenesis, Site-Directed , Phospholipase C beta/physiology , Protein Structure, Secondary/physiology , Protein Structure, Tertiary
6.
Eur Biophys J ; 40(6): 761-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21416312

ABSTRACT

The enzyme diisopropyl fluorophosphatase (DFPase) from the squid Loligo vulgaris is of great interest because of its ability to catalyze the hydrolysis of highly toxic organophosphates. In this work, the enzyme structure in solution (native state) was studied by use of different scattering methods. The results are compared with those from hydrodynamic model calculations based on the DFPase crystal structure. Bicontinuous microemulsions made of sugar surfactants are discussed as host systems for the DFPase. The microemulsion remains stable in the presence of the enzyme, which is shown by means of scattering experiments. Moreover, activity assays reveal that the DFPase still has high activity in this complex reaction medium. To complement the scattering experiments cryo-SEM was also employed to study the microemulsion structure.


Subject(s)
Carbohydrates/pharmacology , Loligo/metabolism , Phosphoric Triester Hydrolases/analysis , Surface-Active Agents/pharmacology , Animals , Carbohydrates/chemistry , Emulsions , Loligo/enzymology , Microscopy, Electron, Scanning , Neutron Diffraction , Phosphoric Triester Hydrolases/chemistry , Phosphoric Triester Hydrolases/metabolism , Scattering, Small Angle , Solutions/chemistry , Surface-Active Agents/chemistry
7.
Anal Bioanal Chem ; 396(3): 1213-21, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943158

ABSTRACT

The enzyme diisopropyl fluorophosphatase (DFPase, EC 3.1.8.2) from the squid Loligo vulgaris effectively catalyzes the hydrolysis of diisopropyl fluorophosphate (DFP) and a number of organophosphorus nerve agents, including sarin, soman, cyclosarin, and tabun. Until now, determination of kinetic data has been achieved by use of techniques such as pH-stat titration, ion-selective electrodes, and a recently introduced method based on in situ Fourier-transform infrared (FTIR) spectroscopy. We report the use of 1D (1)H-(31)P HSQC NMR spectroscopy as a new method for real-time quantification of the hydrolysis of toxic organophosphonates by DFPase. The method is demonstrated for the agents sarin (GB), soman (GD), and cyclosarin (GD) but can also be used for V-type nerve agents, for example VX. Besides buffered aqueous solutions the method was used to determine enzymatic activities in a biodiesel-based bicontinuous microemulsion that serves as an example of complex decontamination media, for which other established techniques often fail. The method is non-invasive and requires only limited manual handling of small volumes of liquid (700 microL), which adds to work safety when handling highly toxic organophosphorus compounds. Limits of detection are slightly below 100 micromol L(-1) on a 400 MHz spectrometer with 16 FIDs added for a single time frame. The method is not restricted to DFPase but can be used with other phosphotriesterases, for example paraxonase (PON), and even reactive chemicals, for example oximes and other nucleophiles, as long as the reaction components are compatible with the NMR experiment.


Subject(s)
Biosensing Techniques/methods , Chemical Warfare Agents/analysis , Cholinesterase Inhibitors/analysis , Magnetic Resonance Spectroscopy/methods , Organophosphorus Compounds/analysis , Phosphoric Triester Hydrolases/metabolism , Animals , Chemical Warfare Agents/metabolism , Cholinesterase Inhibitors/metabolism , Convulsants/analysis , Convulsants/metabolism , Hydrolysis , Limit of Detection , Loligo/enzymology , Organophosphorus Compounds/metabolism , Sarin/analysis , Sarin/metabolism , Soman/analysis , Soman/metabolism
8.
Anal Biochem ; 385(2): 187-93, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19084491

ABSTRACT

The enzyme diisopropyl fluorophosphatase (DFPase) from the squid Loligo vulgaris effectively catalyzes the hydrolysis of diisopropyl fluorophosphate (DFP) and a number of organophosphorus nerve agents, including sarin, soman, cyclosarin, and tabun. Up to now, the determination of kinetic data has been achieved by techniques such as pH-stat titration, ion-selective electrodes, and fluorogenic substrate analogs. We report a new assaying method using in situ Fourier transform infrared (FTIR) spectroscopy with attenuated total reflection (ATR) for the real-time determination of reaction rates. The method employs changes in the P-O-R stretching vibration of DFP and nerve agent substrates when hydrolyzed to their corresponding phosphoric and phosphonic acids. It is shown that the Lambert-Beer law holds and that changes in absorbance can be directly related to changes in concentration. Compared with other methods, the use of in situ FTIR spectroscopy results in a substantially reduced reaction volume that adds extra work safety when handling highly toxic substrates. In addition, the new method allows the noninvasive measurement of buffered solutions with varying ionic strengths complementing existing methods. Because the assay is independent of the used enzyme, it should also be applicable to other phosphotriesterase enzymes such as organophosphorus hydrolase (OPH), organophosphorus acid anhydrolase (OPAA), and paraoxonase (PON).


Subject(s)
Loligo/enzymology , Organophosphates/metabolism , Organophosphonates/metabolism , Phosphoric Triester Hydrolases/metabolism , Spectroscopy, Fourier Transform Infrared/methods , Animals , Hydrolysis , Kinetics , Osmolar Concentration
9.
Article in English | MEDLINE | ID: mdl-17183172

ABSTRACT

The enzyme diisopropyl fluorophosphatase (DFPase) from Loligo vulgaris is capable of decontaminating a wide variety of toxic organophosphorus nerve agents. DFPase is structurally related to a number of enzymes, such as the medically important paraoxonase (PON). In order to investigate the reaction mechanism of this phosphotriesterase and to elucidate the protonation state of the active-site residues, large-sized crystals of DFPase have been prepared for neutron diffraction studies. Available H atoms have been exchanged through vapour diffusion against D2O-containing mother liquor in the capillary. A neutron data set has been collected to 2.2 A resolution on a relatively small (0.43 mm3) crystal at the spallation source in Los Alamos. The sample size and asymmetric unit requirements for the feasibility of neutron diffraction studies are summarized.


Subject(s)
Loligo/enzymology , Neutron Diffraction/methods , Phosphoric Triester Hydrolases/chemistry , Animals , Feasibility Studies , Loligo/chemistry , Phosphoric Triester Hydrolases/isolation & purification
10.
J Am Chem Soc ; 128(39): 12750-7, 2006 Oct 04.
Article in English | MEDLINE | ID: mdl-17002369

ABSTRACT

A wide range of organophosphorus nerve agents, including Soman, Sarin, and Tabun is efficiently hydrolyzed by the phosphotriesterase enzyme diisopropyl fluorophosphatase (DFPase) from Loligo vulgaris. To date, the lack of available inhibitors of DFPase has limited studies on its mechanism. The de novo design, synthesis, and characterization of substrate analogues acting as competitive inhibitors of DFPase are reported. The 1.73 A crystal structure of O,O-dicyclopentylphosphoroamidate (DcPPA) bound to DFPase shows a direct coordination of the phosphoryl oxygen by the catalytic calcium ion. The binding mode of this substrate analogue suggests a crucial role for electrostatics in the orientation of the ligand in the active site. This interpretation is further supported by the crystal structures of double mutants D229N/N120D and D229N/N175D, designed to reorient the electrostatic environment around the catalytic calcium. The structures show no differences in their calcium coordinating environment, although they are enzymatically inactive. Additional double mutants E21Q/N120D and E21Q/N175D are also inactive. On the basis of these crystal structures and kinetic and mutagenesis data as well as isotope labeling we propose a new mechanism for DFPase activity. Calcium coordinating residue D229, in concert with direct substrate activation by the metal ion, renders the phosphorus atom of the substrate susceptible for attack of water, through generation of a phosphoenzyme intermediate. Our proposed mechanism may be applicable to the structurally related enzyme paraoxonase (PON), a component of high-density lipoprotein (HDL).


Subject(s)
Phosphoric Triester Hydrolases/chemistry , Phosphoric Triester Hydrolases/metabolism , Animals , Binding Sites , Calcium/chemistry , Calcium/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Ligands , Loligo/enzymology , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/metabolism , Oxygen Isotopes , Phosphoric Triester Hydrolases/antagonists & inhibitors , Protein Binding , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL