Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 388(2): 333-346, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37770203

ABSTRACT

Synthesis of the acetylcholinesterase inhibitor paraoxon (POX) as a carbon-11 positron emission tomography tracer ([11C]POX) and profiling in live rats is reported. Naïve rats intravenously injected with [11C]POX showed a rapid decrease in parent tracer to ∼1%, with an increase in radiolabeled serum proteins to 87% and red blood cells (RBCs) to 9%. Protein and RBC leveled over 60 minutes, reflecting covalent modification of proteins by [11C]POX. Ex vivo biodistribution and imaging profiles in naïve rats had the highest radioactivity levels in lung followed by heart and kidney, and brain and liver the lowest. Brain radioactivity levels were low but observed immediately after injection and persisted over the 60-minute experiment. This showed for the first time that even low POX exposures (∼200 ng tracer) can rapidly enter brain. Rats given an LD50 dose of nonradioactive paraoxon at the LD50 20 or 60 minutes prior to [11C]POX tracer revealed that protein pools were blocked. Blood radioactivity at 20 minutes was markedly lower than naïve levels due to rapid protein modification by nonradioactive POX; however, by 60 minutes the blood radioactivity returned to near naïve levels. Live rat tissue imaging-derived radioactivity values were 10%-37% of naïve levels in nonradioactive POX pretreated rats at 20 minutes, but by 60 minutes the area under the curve (AUC) values had recovered to 25%-80% of naïve. The live rat imaging supported blockade by nonradioactive POX pretreatment at 20 minutes and recovery of proteins by 60 minutes. SIGNIFICANCE STATEMENT: Paraoxon (POX) is an organophosphorus (OP) compound and a powerful prototype and substitute for OP chemical warfare agents (CWAs) such as sarin, VX, etc. To study the distribution and penetration of POX into the central nervous system (CNS) and other tissues, a positron emission tomography (PET) tracer analog, carbon-11-labeled paraoxon ([11C]POX), was prepared. Blood and tissue radioactivity levels in live rats demonstrated immediate penetration into the CNS and persistent radioactivity levels in tissues indicative of covalent target modification.


Subject(s)
Acetylcholinesterase , Carbon Radioisotopes , Paraoxon , Rats , Animals , Tissue Distribution , Positron-Emission Tomography , Organophosphorus Compounds
2.
Neurochem Res ; 46(3): 494-503, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33398639

ABSTRACT

The vesicular glutamate transporter (VGLUT) facilitates the uptake of glutamate (Glu) into neuronal vesicles. VGLUT has not yet been fully characterized pharmacologically but a body of work established that certain azo-dyes bearing two Glu isosteres via a linker were potent inhibitors. However, the distance between the isostere groups that convey potent inhibition has not been delineated. This report describes the synthesis and pharmacologic assessment of Congo Red analogs that contain one or two glutamate isostere or mimic groups; the latter varied in the interatomic distance and spacer properties to probe strategic binding interactions within VGLUT. The more potent inhibitors had two glutamate isosteres symmetrically linked to a central aromatic group and showed IC50 values ~ 0.3-2.0 µM at VGLUT. These compounds contained phenyl, diphenyl ether (PhOPh) or 1,2-diphenylethane as the linker connecting 4-aminonaphthalene sulfonic acid groups. A homology model for VGLUT2 using D-galactonate transporter (DgoT) to dock and identify R88, H199 and F219 as key protein interactions with Trypan Blue, Congo Red and selected potent analogs prepared and tested in this report.


Subject(s)
Congo Red/analogs & derivatives , Congo Red/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Animals , Congo Red/pharmacology , Drug Design , Molecular Docking Simulation , Molecular Structure , Protein Binding , Rats , Structure-Activity Relationship , Vesicular Glutamate Transport Proteins/antagonists & inhibitors
3.
Chem Res Toxicol ; 34(1): 63-69, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33373198

ABSTRACT

Organophosphorus esters (OPs) were originally developed as pesticides but were repurposed as easily manufactured, inexpensive, and highly toxic chemical warfare agents. Acute OP toxicity is primarily due to inhibition of acetylcholinesterase (AChE), an enzyme in the central and peripheral nervous system. OP inhibition of AChE can be reversed using oxime reactivators but many show poor CNS penetration, indicating a need for new clinically viable reactivators. However, challenges exist on how to best measure restored AChE activity in vivo and assess the reactivating agent efficacy. This work reports the development of molecular imaging tools using radiolabeled OP analog tracers that are less toxic to handle in the laboratory, yet inhibit AChE in a similar fashion to the actual OPs. Carbon-11 and fluorine-18 radiolabeled analog tracers of VX and sarin OP agents were prepared. Following intravenous injection in normal Sprague-Dawley rats (n = 3-4/tracer), the tracers were evaluated and compared using noninvasive microPET/CT imaging, biodistribution assay, and arterial blood analyses. All showed rapid uptake and stable retention in brain, heart, liver, and kidney tissues determined by imaging and biodistribution. Lung uptake of the sarin analog tracers was elevated, 2-fold and 4-fold higher uptake at 5 and 30 min, respectively, compared to that for the VX analog tracers. All tracers rapidly bound to red blood cells (RBC) and blood proteins as measured in the biodistribution and arterial blood samples. Analysis of the plasma soluble activity (nonprotein/cell bound activity) showed only 1-6% parent tracer and 88-95% of the activity in the combined solid fractions (RBC and protein bound) as early as 0.5 min post injection. Multivariate analysis of tracer production yield, molar activity, brain uptake, brain area under the curve over 0-15 min, and the amount of parent tracer in the plasma at 5 min revealed the [18F]VX analog tracer had the most favorable values for each metric. This tracer was considered the more optimal tracer relative to the other tracers studied and suitable for future in vivo OP exposure and reactivation studies.


Subject(s)
Chemical Warfare Agents/pharmacology , Cholinesterase Inhibitors/pharmacology , Organothiophosphorus Compounds/pharmacology , Sarin/pharmacology , Acetylcholinesterase/metabolism , Animals , Carbon Radioisotopes , Chemical Warfare Agents/chemistry , Cholinesterase Inhibitors/chemistry , Fluorine Radioisotopes , Male , Molecular Structure , Organothiophosphorus Compounds/chemistry , Rats , Rats, Sprague-Dawley , Sarin/chemistry , Tissue Distribution
4.
Food Chem Toxicol ; 146: 111785, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33011351

ABSTRACT

The drug transporter P-glycoprotein (P-gp) is often investigated in drug-interaction studies because the activity is modulated by a wide variety of xenobiotics including drugs, herbal products, and food components. In this study, we tested six common arylsulfonate food dyes-allura red, carmoisine, ponceau 4R, quinolone yellow, sunset yellow, and tartrazine-as activators and inhibitors of P-gp activity in vitro. The dyes were studied as P-gp activators by measuring ATPase activity in P-gp-expressing membranes. Compared to verapamil, a known activator of P-gp, the six food dyes showed no stimulatory activity. The potential for these six food dyes to act as P-gp inhibitors was tested in an intracellular efflux assay with P-gp-expressing cells. Compared to GF120918, a known P-gp inhibitor, there was no inhibitory activity for these six food dyes. The six food dyes tested do not interact with P-gp in vitro and, therefore, are unlikely cause clinical drug-food dye interactions. Further investigation is necessary to determine whether these food dyes could interact with other drug transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Food Coloring Agents/pharmacology , Adenosine Triphosphatases/metabolism , Biological Transport , Drug Interactions , Food Coloring Agents/chemistry , Food-Drug Interactions , Humans , Verapamil/pharmacology
5.
Chem Res Toxicol ; 33(9): 2455-2466, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32833441

ABSTRACT

Studies with acetylcholinesterase (AChE) inhibited by organophosphorus (OP) compounds with two chiral centers can serve as models or surrogates for understanding the rate, orientation, and postinhibitory mechanisms by the nerve agent soman that possesses dual phosphorus and carbon chiral centers. In the current approach, stereoisomers of O-methyl, [S-(succinic acid, diethyl ester), O-(4-nitrophenyl) phosphorothiolate (MSNPs) were synthesized, and the inhibition, reactivation, and aging mechanisms were studied with electric eel AChE (eeAChE) and recombinant mouse brain AChE (rmAChE). The MSNP RPRC isomer was the strongest inhibitor of both eeAChE and rmAChE at 8- and 24-fold greater potency, respectively, than the weakest SPSC isomer. eeAChE inhibited by the RPRC- or RPSC-MSNP isomer underwent spontaneous reactivation ∼10- to 20-fold faster than the enzyme inhibited by SPRC- and SPSC-MSNP, and only 4% spontaneous reactivation was observed from the SPRC-eeAChE adduct. Using 2-pyridine aldoxime methiodide (2-PAM) or trimedoxime (TMB-4), eeAChE inhibited by RPRC- or SPRC-MSNP reactivated up to 90% and 3- to 4-fold faster than eeAChE inhibited by the RPSC- or SPSC-MSNP isomer. Spontaneous reactivation rates for rmAChE were 1.5- to 10-fold higher following inhibition by RPSC- and SPSC-MSNPs than inhibition by either RC isomer, a trend opposite to that found for eeAChE. Oxime reactivation of rmAChE following inhibition by RPRC- and SPRC-MSNPs was 2.5- to 5-fold faster than inhibition by RPSC- or SPSC-MSNPs. Due to structural similarities, MSNPs that phosphylate AChE with the loss of the p-nitrophenoxy (PNP) group form identical, nonreactivatable adducts to those formed from SP-isomalathion; however, all the MSNP isomers inhibited AChE to form adducts that reactivated. Thus, MSNPs inactivate AChE via the ejection of either PNP or thiosuccinyl groups to form a combination of reactivatable and nonreactivatable adducts, and this differs from the mechanism of AChE inhibition by isomalathion.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Esters/pharmacology , Nitrophenols/pharmacology , Organophosphorus Compounds/pharmacology , Sulfhydryl Compounds/pharmacology , Animals , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Esters/chemistry , Mice , Molecular Structure , Nitrophenols/chemistry , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/chemistry , Sulfhydryl Compounds/chemistry
6.
Ann N Y Acad Sci ; 1479(1): 180-195, 2020 11.
Article in English | MEDLINE | ID: mdl-32436233

ABSTRACT

Oxime antidotes regenerate organophosphate-inhibited acetylcholinesterase (AChE). Although they share a common mechanism of AChE reactivation, the rate and amount of oxime that enters the brain are critical to the efficacy, a process linked to the oxime structure and charge. Using a platform based on the organophosphate [18 F]-VXS as a positron emission tomography tracer for active AChE, the in vivo distribution of [18 F]-VXS was evaluated after an LD50 dose (250 µg/kg) of the organophosphate paraoxon (POX) and following oximes as antidotes. Rats given [18 F]-VXS tracer alone had significantly higher radioactivity (two- to threefold) in the heart and lung than rats given LD50 POX at 20 or 60 min prior to [18 F]-VXS. When rats were given LD50 POX followed by 2-PAM (cationic), RS194b (ionizable), or monoisonitrosoacetone (MINA) (neutral), central nervous system (CNS) radioactivity returned to levels at or above untreated naive rats (no POX), whereas CNS radioactivity did not increase in rats given the dication oximes HI-6 or MMB-4. MINA showed a significant, pairwise increase in CNS brain radioactivity compared with POX-treated rats. This new in vivo dynamic platform using [18 F]-VXS tracer measures and quantifies peripheral and CNS relative changes in AChE availability after POX exposure and is suitable for comparing oxime delivery and AChE reactivation in rats.


Subject(s)
Acetylcholinesterase , Antidotes/pharmacology , Contrast Media/pharmacology , Heart , Lung , Oximes/pharmacology , Paraoxon/toxicity , Positron-Emission Tomography , Acetylcholinesterase/metabolism , Animals , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Heart/diagnostic imaging , Heart/physiopathology , Lung/diagnostic imaging , Lung/metabolism , Lung/physiopathology , Male , Organophosphorus Compounds/pharmacology , Radioactive Tracers , Rats , Rats, Sprague-Dawley
7.
Biochim Biophys Acta Biomembr ; 1862(12): 183175, 2020 12 01.
Article in English | MEDLINE | ID: mdl-31923412

ABSTRACT

The vesicular glutamate transporters (VGLUTs) bind and move glutamate (Glu) from the cytosol into the lumen of synaptic vesicles using a H+-electrochemical gradient (ΔpH and Δψ) generated by the vesicular H+-ATPase. VGLUTs show very low Glu binding and to date, no three-dimensional structure has been elucidated. Prior studies have attempted to identify the key residues involved in binding VGLUT substrates and inhibitors using homology models and docking experiments. Recently, the inward and outward oriented crystal structures of d-galactonate transporter (DgoT) emerged as possible structure templates for VGLUT. In this review, a new homology model for VGLUT2 based on DgoT has been developed and used to conduct docking experiments to identify and differentiate residues and binding orientations involved in ligand interactions. This review describes small molecule-ligand interactions including docking using a VGLUT2 homology model derived from DgoT.


Subject(s)
Molecular Docking Simulation , Vesicular Glutamate Transport Proteins/metabolism , Binding Sites , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Glutamic Acid/analogs & derivatives , Glutamic Acid/metabolism , Humans , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Substrate Specificity , Thermodynamics , Vesicular Glutamate Transport Proteins/antagonists & inhibitors
8.
Neurobiol Dis ; 133: 104455, 2020 01.
Article in English | MEDLINE | ID: mdl-31022458

ABSTRACT

There is a unique in vivo interplay involving the mechanism of inactivation of acetylcholinesterase (AChE) by toxic organophosphorus (OP) compounds and the restoration of AChE activity by oxime antidotes. OP compounds form covalent adducts to this critical enzyme target and oximes are introduced to directly displace the OP from AChE. For the most part, the in vivo inactivation of AChE leading to neurotoxicity and antidote-based therapeutic reversal of this mechanism are well understood, however, these molecular-level events have not been evaluated by dynamic imaging in living systems at millimeter resolution. A deeper understanding of these critically, time-dependent mechanisms is needed to develop new countermeasures. To address this void and to help accelerate the development of new countermeasures, positron-emission tomography (PET) has been investigated as a unique opportunity to create platform technologies to directly examine the interdependent toxicokinetic/pharmacokinetic and toxicodynamic/pharmacodynamic features of OPs and oximes in real time within live animals. This review will cover two first-in-class PET tracers representing an OP and an oxime antidote, including their preparation, requisite pharmacologic investigations, mechanistic interpretations, biodistribution and imaging.


Subject(s)
Cholinesterase Reactivators/pharmacokinetics , Nerve Agents , Organophosphorus Compounds , Positron-Emission Tomography/methods , Radiopharmaceuticals , Animals , Antidotes/pharmacokinetics , Humans , Nerve Agents/pharmacokinetics , Nerve Agents/toxicity , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/toxicity , Oximes/pharmacokinetics
9.
J Physiol ; 597(22): 5495-5514, 2019 11.
Article in English | MEDLINE | ID: mdl-31541561

ABSTRACT

KEY POINTS: Triheteromeric NMDA receptors contain two GluN1 and two distinct GluN2 subunits and mediate excitatory neurotransmission in the CNS. Triheteromeric GluN1/2B/2D receptors have functional properties intermediate to those of diheteromeric GluN1/2B and GluN1/2D receptors. GluN1/2B/2D receptors are more sensitive to channel blockade by ketamine and memantine compared to GluN1/2B receptors in the presence of physiological Mg2+ . GluN2B-selective antagonists produce robust inhibition of GluN1/2B/2D receptors, and the GluN2B-selective positive allosteric modulator spermine enhances responses from GluN1/2B/2D but not GluN1/2A/2B receptors. These insights into the properties of triheteromeric GluN1/2B/2D receptors are necessary to appreciate their physiological roles in neural circuit function and the actions of therapeutic agents targeting NMDA receptors. ABSTRACT: Triheteromeric NMDA-type glutamate receptors that contain two GluN1 and two different GluN2 subunits contribute to excitatory neurotransmission in the adult CNS. In the present study, we report properties of the triheteromeric GluN1/2B/2D NMDA receptor subtype that is expressed in distinct neuronal populations throughout the CNS. We show that neither GluN2B, nor GluN2D dominate the functional properties of GluN1/2B/2D receptors because agonist potencies, open probability and the glutamate deactivation time course of GluN1/2B/2D receptors are intermediate to those of diheteromeric GluN1/2B and GluN1/2D receptors. Furthermore, channel blockade of GluN1/2B/2D by extracellular Mg2+ is intermediate compared to GluN1/2B and GluN1/2D, although GluN1/2B/2D is more sensitive to blockade by ketamine and memantine compared to GluN1/2B in the presence of physiological Mg2+ . Subunit-selective allosteric modulators have distinct activity at GluN1/2B/2D receptors, including GluN2B-selective antagonists, ifenprodil, EVT-101 and CP-101-606, which inhibit with similar potencies but with different efficacies at GluN1/2B/2D (∼65% inhibition) compared to GluN1/2B (∼95% inhibition). Furthermore, the GluN2B-selective positive allosteric modulator spermine enhances responses from GluN1/2B/2D but not GluN1/2A/2B receptors. We show that these key features of allosteric modulation of recombinant GluN1/2B/2D receptors are also observed for NMDA receptors in hippocampal interneurons but not CA1 pyramidal cells, which is consistent with the expression of GluN1/2B/2D receptors in interneurons and GluN1/2A/2B receptors in pyramidal cells. Altogether, we uncover previously unknown functional and pharmacological properties of triheteromeric GluN1/2B/2D receptors that can facilitate advances in our understanding of their physiological roles in neural circuit function and therapeutic drug actions.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Interneurons/drug effects , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
10.
Environ Toxicol Pharmacol ; 71: 103218, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31302432

ABSTRACT

A novel panel of oximes were synthesized, which have displayed varying degree of reactivation ability towards different organophosphorus (OP) modified cholinesterases. In the present article, we report a comparative reactivation profile of a series of quaternary pyridinium-oximes for electric eel acetylcholinesterase (EEAChE) inhibited by the organophosphorus (OP) inhibitors methyl paraoxon (MePOX), ethyl paraoxon (POX; paraoxon) and diisopropyl fluorophosphate (DFP) that are distinguishable as dimethoxyphosphoryl, diethoxyphosphoryl and diisopropoxyphosphoryl AChE-OP-adducts. Most of the 59-oximes tested led to faster and more extensive reactivation of MePOX- and POX-inhibited EEAChE as compared to DFP-modified EEAChE. All were effective reactivators of three OP-modified EEAChE conjugates showing 18-21% reactivation for DFP-inhibited AChE and ≥45% reactivation for MePOX- and POX-inhibited EEAChE. Oximes 7 and 8 showed kr values better than pralidoxime (1) for DFP-inhibited EEAChE. Reactivation rates determined at different inhibition times showed no significant change in kr values during 0-90 min incubation with three OPs. However, a 34-72% decrease in kr for MePOX and POX and > 95% decrease in kr for DFP-inhibited EEAChE was observed after 24 h of OP-exposure (aging).


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Cholinesterase Reactivators/pharmacology , Organophosphorus Compounds/chemistry , Oximes/pharmacology , Pyridinium Compounds/pharmacology , Animals , Cholinesterase Reactivators/chemical synthesis , Cholinesterase Reactivators/chemistry , Drug Design , Electrophorus , Molecular Docking Simulation , Molecular Structure , Nerve Agents/chemistry , Organophosphate Poisoning/enzymology , Organophosphate Poisoning/prevention & control , Oximes/chemical synthesis , Oximes/chemistry , Pyridinium Compounds/chemical synthesis , Pyridinium Compounds/chemistry
11.
Appl Radiat Isot ; 151: 182-186, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31202183

ABSTRACT

Radiolabeled 1-[11C]ethyl, 4-nitrophenyl methylphosphonate (VX surrogate) and 2-[11C]-propanyl, 4-nitrophenyl methylphosphonate (sarin surrogate) were developed as organophosphate (OP) tracers. The [11C]ethyl- and [11C]isopropyl-iodide radiolabeled synthons were obtained by temperature controlled, in loop reactions of [11C]CO2 with MeMgBr followed by reduction with LiAlH4, then reaction with HI. Distillation of the [11C]alkyl iodides into a solution of hydrogen (4-nitrophenyl)methylphosphonate and cesium carbonate afforded the desired tracers in >95% radiochemical purity, yields from [11C]CO2 of 1-3% and 1.7-15.1 GBq/mmol molar activities.

12.
J Labelled Comp Radiopharm ; 61(14): 1089-1094, 2018 12.
Article in English | MEDLINE | ID: mdl-30347484

ABSTRACT

O-(1-Fluoropropan-2-yl)-O-(4-nitrophenyl) methylphosphonate is a reactive organophosphate ester (OP) developed as a surrogate of the chemical warfare agent sarin that forms a similar covalent adduct at the active site serine of acetylcholinesterase. The radiolabeled O-(1-[18 F]fluoropropan-2-yl)-O-(4-nitrophenyl) methylphosphonate ([18 F] fluorosarin surrogate) has not been previously prepared. In this paper, we report the first radiosynthesis of this tracer from the reaction of bis-(4-nitrophenyl) methylphosphonate with 1-[18 F]fluoro-2-propanol in the presence of DBU. The 1-[18 F]fluoro-2-propanol was prepared by reaction of propylene sulfite with Kryptofix 2.2.2 and [18 F] fluoride ion. The desired tracer O-(1-[18 F]fluoropropan-2-yl)-O-(4-nitrophenyl) methylphosphonate was obtained in a >98% radiochemical purity with a 2.4% ± 0.6% yield (n = 5, 65 minutes from start of synthesis) based on starting [18 F] fluoride ion and a molar activity of 49.9 GBq/µmol (1.349 ± 0.329 Ci/µmol, n = 3). This new facile radiosynthesis routinely affords sufficient quantities of [18 F] fluorosarin surrogate in high radiochemical purity, which will further enable the tracer development as a novel radiolabeled OP acetylcholinesterase inhibitor for assessment of OP modes of action with PET imaging in vivo.


Subject(s)
Nitro Compounds/chemistry , Nitro Compounds/chemical synthesis , Organophosphonates/chemistry , Organophosphonates/chemical synthesis , Positron-Emission Tomography , Sarin , Chemistry Techniques, Synthetic , Radioactive Tracers , Radiochemistry
13.
ACS Chem Neurosci ; 9(12): 3007-3014, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30071719

ABSTRACT

2-Pyridinealdoxime methiodide (2-PAM) is a widely used antidote for the treatment of organophosphorus (OP) exposure that reactivates the target protein acetylcholinesterase. Carbon-11 2-PAM was prepared to more fully understand the in vivo mode of action, distribution, and dynamic qualities of this important countermeasure. Alkylation of 2-pyridinealdoxime with [11C]CH3I provided the first-in-class [11C]2-PAM tracer in 3.5% decay corrected radiochemical yield from [11C]CH3I, >99% radiochemical purity, and 4831 Ci/mmol molar activity. [11C]2-PAM tracer distribution was evaluated by ex vivo biodistribution and in vivo dynamic positron emission tomography (PET) imaging in naïve (OP exposure deficient) rats. Tracer alone and tracer coinjected with a body mass-scaled human therapeutic dose of 30 mg/kg nonradioactive 2-PAM demonstrated statistically similar tissue and blood distribution profiles with the greatest uptake in kidney and significantly lower levels in liver, heart, and lung with lesser amounts in blood and brain. The imaging and biodistribution data show that radioactivity uptake in brain and peripheral organs is rapid and characterized by differential tissue radioactivity washout profiles. Analysis of arterial blood samples taken 5 min after injection showed ∼82% parent [11C]2-PAM tracer. The imaging and biodistribution data are now established, enabling future comparisons to outcomes acquired in OP intoxicated rodent models.


Subject(s)
Antidotes/pharmacokinetics , Carbon Radioisotopes/pharmacokinetics , Organophosphate Poisoning , Pralidoxime Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/chemistry , Heart/diagnostic imaging , Kidney/diagnostic imaging , Kidney/metabolism , Liver/diagnostic imaging , Liver/metabolism , Lung/diagnostic imaging , Lung/metabolism , Myocardium/metabolism , Positron-Emission Tomography , Pralidoxime Compounds/chemical synthesis , Radioactive Tracers , Radiopharmaceuticals/chemical synthesis , Rats , Tissue Distribution
14.
Chem Biol Interact ; 291: 220-227, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29920286

ABSTRACT

In this study, the mechanisms of HuAChE and HuBChE inhibition by Me-P(O) (OPNP) (OR) [PNP = p-nitrophenyl; R = CH2CH3, CH2CH2F, OCH(CH3)2, OCH(CH3) (CH2F)] representing surrogates and fluoro-surrogates of VX and sarin were studied by in vitro kinetics and mass spectrometry. The in vitro measures showed that the VX- and fluoro-VX surrogates were relatively strong inhibitors of HuAChE and HuBChE (ki ∼ 105-106 M-1min-1) and underwent spontaneous and 2-PAM-mediated reactivation within 30 min. The sarin surrogates were weaker inhibitors of HuAChE and HuBChE (ki ∼ 104-105 M-1min-1), and in general did not undergo spontaneous reactivation, although HuAChE adducts were partially reactivatable at 18 h using 2-PAM. The mechanism of HuAChE and HuBChE inhibition by the surrogates was determined by Q-TOF and MALDI-TOF mass spectral analyses. The surrogate-adducted proteins were trypsin digested and the active site-containing peptide bearing the OP-modified serine identified by Q-TOF as triply- and quadruply-charged ions representing the respective increase in mass of the attached OP moiety. Correspondingly, monoisotopic ions of the tryptic peptides representing the mass increase of the OP-adducted peptide was identified by MALDI-TOF. The mass spectrometry analyses validated the identity of the OP moiety attached to HuAChE or HuBChE as MeP(O) (OR)-O-serine peptides (loss of the PNP leaving group) via mechanisms consistent with those found with chemical warfare agents. MALDI-TOF MS analyses of the VX-modified peptides versus time showed a steady reduction in adduct versus parent peptide (reactivation), whereas the sarin-surrogate-modified peptides remained largely intact over the course of the experiment (24 h). Overall, the presence of a fluorine atom on the surrogate modestly altered the rate constants of inhibition and reactivation, however, the mechanism of inhibition (ejection of PNP group) did not change.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Reactivators/pharmacology , Organothiophosphorus Compounds/toxicity , Sarin/toxicity , Chemical Warfare Agents/toxicity , Enzyme Activation/drug effects , Fluorescence , Humans , Kinetics , Organophosphorus Compounds/toxicity , Paraoxon/toxicity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
J Labelled Comp Radiopharm ; 60(7): 337-342, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28406525

ABSTRACT

O-(2-Fluoroethyl)-O-(p-nitrophenyl) methylphosphonate 1 is an organophosphate cholinesterase inhibitor that creates a phosphonyl-serine covalent adduct at the enzyme active site blocking cholinesterase activity in vivo. The corresponding radiolabeled O-(2-[18 F]fluoroethyl)-O-(p-nitrophenyl) methylphosphonate, [18 F]1, has been previously prepared and found to be an excellent positron emission tomography imaging tracer for assessment of cholinesterases in live brain, peripheral tissues, and blood. However, the previously reported [18 F]1 tracer synthesis was slow even with microwave acceleration, required high-performance liquid chromatography separation of the tracer from impurities, and gave less optimal radiochemical yields. In this paper, we report a new synthetic approach to circumvent these shortcomings that is reliant on the facile reactivity of bis-(O,O-p-nitrophenyl) methylphosphonate, 2, with 2-fluoroethanol in the presence of DBU. The cold synthesis was successfully translated to provide a more robust radiosynthesis. Using this new strategy, the desired tracer, [18 F]1, was obtained in a non-decay-corrected radiochemical yield of 8 ± 2% (n = 7) in >99% radiochemical and >95% chemical purity with a specific activity of 3174 ± 345 Ci/mmol (EOS). This new facile radiosynthesis routinely affords highly pure quantities of [18 F]1, which will further enable tracer development of OP cholinesterase inhibitors and their evaluation in vivo.


Subject(s)
Chemistry Techniques, Synthetic/methods , Cholinesterases/analysis , Organophosphonates/chemical synthesis , Positron-Emission Tomography , Organophosphonates/chemistry , Radioactive Tracers
16.
Chem Res Toxicol ; 29(11): 1810-1817, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27551891

ABSTRACT

The organophosphate O-(2-fluoroethyl)-O-(p-nitrophenyl) methyphosphonate 1 is the first-in-class, fluorine-18 radiolabeled organophosphate inhibitor ([18F]1) of acetylcholinesterase (AChE). In rats, [18F]1 localizes in AChE rich regions of the brain and other tissues where it likely exists as the (CH3)(18FCH2CH2O)P(O)-AChE adduct (ChE-1). Characterization of this adduct would define the inhibition mechanism and subsequent postinhibitory pathways and reactivation rates. To validate this adduct, the stability (hydrolysis) of 1 and ChE-1 reactivation rates were determined. Base hydrolysis of 1 yields p-nitrophenol and (CH3) (FCH2CH2O)P(O)OH with pseudo first order rate constants (kobsd) at pH 7.4 (PBS) of 3.25 × 10-4 min-1 (t1/2 = 35.5 h) at 25 °C and 8.70 × 10-4 min-1 (t1/2 = 13.3 h) at 37 °C. Compound 1 was a potent inhibitor of human acetylcholinesterase (HuAChE; ki = 7.5 × 105 M-1 min-1), electric eel acetylcholinesterase (EEAChE) (ki = 3.0 × 106 M-1 min-1), and human serum butyrylcholinesterase (HuBChE; 1.95 × 105 M-1 min-1). Spontaneous and oxime-mediated reactivation rates for the (CH3) (FCH2CH2O)P(O)-serine ChE adducts using 2-PAM (10 µM) were (a) HuAChE 8.8 × 10-5 min-1 (t1/2 = 131.2 h) and 2.41 × 10-2 min-1 (t1/2 = 0.48 h), (b) EEAChE 9.32 × 10-3 min-1 (t1/2 = 1.24 h) and 3.33 × 10-2 min-1 (t1/2 = 0.35 h), and (c) HuBChE 1.16 × 10-4 min-1 (t1/2 = 99.6 h) and 4.19 × 10-2 min-1 (t1/2 = 0.27 h). All ChE-1 adducts undergo rapid and near complete restoration of enzyme activity following addition of 2-PAM (30 min), and no aging was observed for either reactivation process. The fast reactivation rates and absence of aging of ChE-1 adducts are explained on the basis of the electron-withdrawing fluorine group that favors the nucleophilic reactivation processes but disfavors cation-based dealkylation aging mechanisms. Therefore, the likely fate of radiolabeled compound 1 in vivo is the formation of (CH3)(FCH2CH2O)P(O)-serine adducts and monoacid (CH3)(FCH2CH2O)P(O)OH from hydrolysis and reactivation.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Cholinesterase Reactivators/pharmacology , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacology , Animals , Carbon-13 Magnetic Resonance Spectroscopy , Cholinesterases/drug effects , Cholinesterases/metabolism , Humans , Hydrolysis , Ligands , Organophosphorus Compounds/chemistry , Proton Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization
17.
Chem Biol Interact ; 223: 134-40, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25261769

ABSTRACT

In this study, the first mechanism-based monoclonal antibodies have been produced that recognize and differentiate diethoxy- and monoethoxyphosphorylated serine residues. Haptens were synthesized as the stable phosphonate form of phosphoserine esters to improve the immunoresponse. Following condensation with a glutaric anhydride to link the phosphoserine moieties to carrier protein, the hapten densities attached to bovine serum albumin and keyhole limpet henocyanin were determined by partial trypsin digestion and MALDI mass spectrometry, and confirmed using a fluorescent assay (FITC) to quantify unmodified lysine residues. The conjugation reactions were pH optimized to improve hapten density. Screening of subclones led to the identification of two monoclonal antibodies: (a) N257/25.11 that specifically recognizes (EtO)2P(O)-Ser as the phosphylated or inhibited form, and (b) N262/16 that recognizes (EtO)(HO)P(O)-Ser as the 'aged' form. Analysis of blood samples treated with paraoxon (EtO)2P(O)-OPhNO2 showed a concentration dependent recognition of the phosphylated form.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Haptens/chemistry , Insecticides/chemistry , Insecticides/immunology , Organophosphates/chemistry , Organophosphates/immunology , Animals , Cattle , Enzyme-Linked Immunosorbent Assay , Haptens/isolation & purification , Hemocyanins/chemistry , Hemocyanins/immunology , Humans , Insecticides/toxicity , Male , Mice , Organophosphates/toxicity , Paraoxon/chemistry , Paraoxon/immunology , Paraoxon/toxicity , Phosphoserine/analogs & derivatives , Phosphoserine/chemistry , Phosphoserine/immunology , Rats , Rats, Inbred SHR , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Tetrahedron Asymmetry ; 25(6-7): 529-533, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24839353

ABSTRACT

Malathion, diethyl 2-[(dimethoxyphosphorothioyl)sulfanyl]butanedioate, is an organophosphate used to control insect pests. Malathion contains a diethyl succinate moiety that is a known functional group susceptible to desymmetrizing enzymes such as esterases that selectively react with a single enantiomer. Purified rac-malathion was subjected to hydrolysis at the diethyl succinate moiety of malathion under various conditions using wild type pig liver esterase to form (S)-malathion (12 % ee) and ~ 3:2 mixture of α- and ß-monoacids of (R)-malathion. Technical malathion could not be enriched due to the presence of esterase inhibitors. Further investigation of this resolution using a panel of six PLE isoenzymes also demonstrated formation of (S)-malathion, however, an improvement of up to 56 % ee was obtained.

19.
ACS Chem Neurosci ; 5(7): 519-24, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-24716794

ABSTRACT

Radiosynthesis of a fluorine-18 labeled organophosphate (OP) inhibitor of acetylcholinesterase (AChE) and subsequent positron emission tomography (PET) imaging using the tracer in the rat central nervous system are reported. The tracer structure, which contains a novel ß-fluoroethoxy phosphoester moiety, was designed as an insecticide-chemical nerve agent hybrid to optimize handling and the desired target reactivity. Radiosynthesis of the ß-fluoroethoxy tracer is described that utilizes a [(18)F]prosthetic group coupling approach. The imaging utility of the [(18)F]tracer is demonstrated in vivo within rats by the evaluation of its brain penetration and cerebral distribution qualities in the absence and presence of a challenge agent. The tracer effectively penetrates brain and localizes to cerebral regions known to correlate with the expression of the AChE target. Brain pharmacokinetic properties of the tracer are consistent with the formation of an OP-adducted acetylcholinesterase containing the fluoroethoxy tracer group. Based on the initial favorable in vivo qualities found in rat, additional [(18)F]tracer studies are ongoing to exploit the technology to dynamically probe organophosphate mechanisms of action in mammalian live tissues.


Subject(s)
Acetylcholinesterase/metabolism , Brain/diagnostic imaging , Fluorine Radioisotopes , Organophosphates , Radiopharmaceuticals , Spinal Cord/diagnostic imaging , Animals , Brain/metabolism , Fluorine Radioisotopes/chemistry , Male , Organophosphates/chemical synthesis , Organophosphates/pharmacokinetics , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Skull/diagnostic imaging , Skull/metabolism , Spinal Cord/metabolism , Spine/diagnostic imaging , Spine/metabolism , Tomography, X-Ray Computed
20.
Toxicol In Vitro ; 28(5): 715-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24486155

ABSTRACT

Organophosphate (OP) compounds are used as insecticides, acaricides, and chemical agents and share a common neurotoxic mechanism of action. The biochemical alterations leading to many of the deleterious effects have been studied in neuronal cell lines, however, non-neuronal toxic effects of OPs are far less well characterized in vitro, and specifically in cell lines representing oral routes of exposure. To address this void, the human salivary gland (HSG) cell line, representing likely interactions in the oral cavity, was exposed to the representative OP paraoxon (PX; O,O-diethyl-p-nitrophenoxy phosphate) over a range of concentrations (0.01-100 µM) and analyzed for cytotoxicity. PX induced cytotoxicity in HSG cells at most of the exposure concentrations as revealed by MTT assay, however, the release of LDH only occurred at the highest concentration of PX tested (100 µM) at 48 h. Slight increases in cellular ATP levels were measured in PX-exposed (10 µM) HSG cells at 24 h. Exposing HSG cells to 10 µM PX also led to an increase in DNA fragmentation prior to loss of cellular membrane integrity implicating reactive oxygen species (ROS) as a trigger of toxicity. The ROS genes gss, gstm2, gstt2 and sod2 were upregulated, and the presence of superoxide following 10 µM PX exposure was determined via dihydroethidium fluorescence studies further implicating PX-induced oxidative stress in HSG cells.


Subject(s)
Cholinesterase Inhibitors/toxicity , Insecticides/toxicity , Oxidative Stress , Paraoxon/toxicity , Salivary Glands/cytology , Acetylcholinesterase/metabolism , Adenosine Triphosphate/metabolism , Cell Line , Cell Survival/drug effects , DNA Fragmentation , Eating , Glutathione Synthase/genetics , Glutathione Transferase/genetics , Humans , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...