Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Emerg Infect Dis ; 30(4): 757-760, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526137

ABSTRACT

Analyzing vaccine stability under different storage and transportation conditions is critical to ensure that effectiveness and safety are not affected by distribution. In a simulation of the last mile in the supply chain, we found that shock and vibration had no effect on Ad26.ZEBOV/MVA-BN-Filo Ebola vaccine regimen quality under refrigerated conditions.


Subject(s)
Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Hemorrhagic Fever, Ebola/prevention & control , Vibration , Computer Simulation , Antibodies, Viral
2.
Methods Mol Biol ; 2576: 189-199, 2023.
Article in English | MEDLINE | ID: mdl-36152187

ABSTRACT

Cannabinoid receptor 1 (CB1R) and cannabinoid receptor 2 (CB2R) are G protein-coupled receptors (GPCRs) that activate a variety of pathways upon activation by (partial) agonists including the G protein pathway and the recruitment of ß-arrestins. Differences in the activation level of these pathways lead to biased signaling. Here, we describe a detailed protocol to characterize the potency and efficacy of ligands to induce or inhibit ß-arrestin recruitment to the human CB1R and CB2R using the PathHunter® assay. This is a cellular assay that uses a ß-galactosidase complementation system which has a chemiluminescent read-out and can be performed in 384-well plates. We have successfully used this assay to characterize a set of reference ligands (both agonists, antagonists, and an inverse agonist) on human CB1R and CB2R, of which some examples will be presented here.


Subject(s)
GTP-Binding Proteins , Receptors, G-Protein-Coupled , GTP-Binding Proteins/metabolism , Humans , Ligands , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism , beta-Galactosidase/metabolism
3.
Nat Chem Biol ; 16(6): 667-675, 2020 06.
Article in English | MEDLINE | ID: mdl-32393901

ABSTRACT

N-acylethanolamines (NAEs), which include the endocannabinoid anandamide, represent an important family of signaling lipids in the brain. The lack of chemical probes that modulate NAE biosynthesis in living systems hamper the understanding of the biological role of these lipids. Using a high-throughput screen, chemical proteomics and targeted lipidomics, we report here the discovery and characterization of LEI-401 as a CNS-active N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor. LEI-401 reduced NAE levels in neuroblastoma cells and in the brain of freely moving mice, but not in NAPE-PLD KO cells and mice, respectively. LEI-401 activated the hypothalamus-pituitary-adrenal axis and impaired fear extinction, thereby emulating the effect of a cannabinoid CB1 receptor antagonist, which could be reversed by a fatty acid amide hydrolase inhibitor. Our findings highlight the distinctive role of NAPE-PLD in NAE biosynthesis in the brain and suggest the presence of an endogenous NAE tone controlling emotional behavior.


Subject(s)
Behavior, Animal/drug effects , Enzyme Inhibitors/chemistry , Lipid Metabolism/drug effects , Phosphatidylethanolamines/metabolism , Phospholipase D/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Blood Proteins/metabolism , Brain/metabolism , Cannabinoid Receptor Antagonists/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Fear/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Receptors, Cannabinoid/metabolism , Signal Transduction
4.
J Med Chem ; 61(17): 7892-7901, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30080404

ABSTRACT

Using activity-based protein profiling (ABPP), functional proteins can be interrogated in their native environment. Despite their pharmaceutical relevance, G protein-coupled receptors (GPCRs) have been difficult to address through ABPP. In the current study, we took the prototypical human adenosine A2A receptor (hA2AR) as the starting point for the construction of a chemical toolbox allowing two-step affinity-based labeling of GPCRs. First, we equipped an irreversibly binding hA2AR ligand with a terminal alkyne to serve as probe. We showed that our probe irreversibly and concentration-dependently labeled purified hA2AR. Click-ligation with a sulfonated cyanine-3 fluorophore allowed us to visualize the receptor on SDS-PAGE. We further demonstrated that labeling of the purified hA2AR by our probe could be inhibited by selective antagonists. Lastly, we showed successful labeling of the receptor in cell membranes overexpressing hA2AR, making our probe a promising affinity-based tool compound that sets the stage for the further development of probes for GPCRs.


Subject(s)
Adenosine/metabolism , Cell Membrane/metabolism , Molecular Probes/chemistry , Molecular Probes/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenosine/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , HEK293 Cells , Humans , Ligands , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Receptors, G-Protein-Coupled/chemistry
5.
Cannabis Cannabinoid Res ; 3(1): 136-151, 2018.
Article in English | MEDLINE | ID: mdl-29992186

ABSTRACT

Introduction: Δ9-Tetrahydrocannabinol (THC), the principle psychoactive ingredient in Cannabis, is widely used for its therapeutic effects in a large variety of diseases, but it also has numerous neurological side effects. The cannabinoid receptors (CBRs) are responsible to a large extent for these, but not all biological responses are mediated via the CBRs. Objectives: The identification of additional target proteins of THC to enable a better understanding of the (adverse) physiological effects of THC. Methods: In this study, a chemical proteomics approach using a two-step photoaffinity probe is applied to identify potential proteins that may interact with THC. Results: Photoaffinity probe 1, containing a diazirine as a photocrosslinker, and a terminal alkyne as a ligation handle, was synthesized in 14 steps. It demonstrated high affinity for both CBRs. Subsequently, two-step photoaffinity labeling in neuroblastoma cells led to identification of four potential novel protein targets of THC. The identification of these putative protein hits is a first step towards a better understanding of the protein interaction profile of THC, which could ultimately lead to the development of novel therapeutics based on THC.

6.
Biochem Pharmacol ; 152: 129-142, 2018 06.
Article in English | MEDLINE | ID: mdl-29574067

ABSTRACT

A decade ago, the drug-target residence time model has been (re-)introduced, which describes the importance of binding kinetics of ligands on their protein targets. Since then, it has been applied successfully for multiple protein targets, including GPCRs, for the development of lead compounds with slow dissociation kinetics (i.e. long target residence time) to increase in vivo efficacy or with short residence time to prevent on-target associated side effects. To date, this model has not been applied in the design and pharmacological evaluation of novel selective ligands for the cannabinoid CB2 receptor (CB2R), a GPCR with therapeutic potential in the treatment of tissue injury and inflammatory diseases. Here, we have investigated the relationships between physicochemical properties, binding kinetics and functional activity in two different signal transduction pathways, G protein activation and ß-arrestin recruitment. We synthesized 24 analogues of 3-cyclopropyl-1-(4-(6-((1,1-dioxidothiomorpholino)methyl)-5-fluoropyridin-2-yl)benzyl)imidazoleidine-2,4-dione (LEI101), our previously reported in vivo active and CB2R-selective agonist, with varying basicity and lipophilicity. We identified a positive correlation between target residence time and functional potency due to an increase in lipophilicity on the alkyl substituents, which was not the case for the amine substituents. Basicity of the agonists did not show a relationship with affinity, residence time or functional activity. Our findings provide important insights about the effects of physicochemical properties of the specific substituents of this scaffold on the binding kinetics of agonists and their CB2R pharmacology. This work therefore shows how CB2R agonists can be designed to have optimal kinetic profiles, which could aid the lead optimization process in drug discovery for the study or treatment of inflammatory diseases.


Subject(s)
Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/pharmacology , Imidazolidines/chemistry , Imidazolidines/pharmacology , Morpholines/chemistry , Morpholines/pharmacology , Receptor, Cannabinoid, CB2/physiology , Animals , Cannabinoid Receptor Agonists/chemistry , Cell Line , Cricetinae , Humans , Kinetics , Ligands , Molecular Structure , Protein Binding , Structure-Activity Relationship
7.
J Am Chem Soc ; 140(19): 6067-6075, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29420021

ABSTRACT

Chemical tools and methods that report on G protein-coupled receptor (GPCR) expression levels and receptor occupancy by small molecules are highly desirable. We report the development of LEI121 as a photoreactive probe to study the type 2 cannabinoid receptor (CB2R), a promising GPCR to treat tissue injury and inflammatory diseases. LEI121 is the first CB2R-selective bifunctional probe that covalently captures CB2R upon photoactivation. An incorporated alkyne serves as ligation handle for the introduction of reporter groups. LEI121 enables target engagement studies and visualization of endogenously expressed CB2R in HL-60 as well as primary human immune cells using flow cytometry. Our findings show that strategically functionalized probes allow monitoring of endogenous GPCR expression and engagement in human cells using tandem photoclick chemistry and hold promise as biomarkers in translational drug discovery.


Subject(s)
Morpholines/chemistry , Photoaffinity Labels/chemistry , Pyridines/chemistry , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/metabolism , Alkynes/chemistry , HL-60 Cells , Humans , Ligands , Molecular Structure , Morpholines/chemical synthesis , Photoaffinity Labels/chemical synthesis , Pyridines/chemical synthesis
8.
Science ; 356(6342): 1084-1087, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28596366

ABSTRACT

A recent phase 1 trial of the fatty acid amide hydrolase (FAAH) inhibitor BIA 10-2474 led to the death of one volunteer and produced mild-to-severe neurological symptoms in four others. Although the cause of the clinical neurotoxicity is unknown, it has been postulated, given the clinical safety profile of other tested FAAH inhibitors, that off-target activities of BIA 10-2474 may have played a role. Here we use activity-based proteomic methods to determine the protein interaction landscape of BIA 10-2474 in human cells and tissues. This analysis revealed that the drug inhibits several lipases that are not targeted by PF04457845, a highly selective and clinically tested FAAH inhibitor. BIA 10-2474, but not PF04457845, produced substantial alterations in lipid networks in human cortical neurons, suggesting that promiscuous lipase inhibitors have the potential to cause metabolic dysregulation in the nervous system.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Anti-Anxiety Agents/pharmacology , Cyclic N-Oxides/pharmacology , Neurons/drug effects , Pyridines/pharmacology , Analgesics/adverse effects , Analgesics/chemistry , Analgesics/metabolism , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/metabolism , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Cross Reactions , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/metabolism , Humans , Neurons/metabolism , Protein Interaction Maps , Pyridazines/pharmacology , Pyridazines/therapeutic use , Pyridines/adverse effects , Pyridines/chemistry , Pyridines/metabolism , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
9.
Nat Commun ; 8: 13958, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045021

ABSTRACT

The cannabinoid CB2 receptor (CB2R) represents a promising therapeutic target for various forms of tissue injury and inflammatory diseases. Although numerous compounds have been developed and widely used to target CB2R, their selectivity, molecular mode of action and pharmacokinetic properties have been poorly characterized. Here we report the most extensive characterization of the molecular pharmacology of the most widely used CB2R ligands to date. In a collaborative effort between multiple academic and industry laboratories, we identify marked differences in the ability of certain agonists to activate distinct signalling pathways and to cause off-target effects. We reach a consensus that HU910, HU308 and JWH133 are the recommended selective CB2R agonists to study the role of CB2R in biological and disease processes. We believe that our unique approach would be highly suitable for the characterization of other therapeutic targets in drug discovery research.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Neurons/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction , Animals , Bridged Bicyclo Compounds/pharmacology , CHO Cells , Cannabinoids/pharmacology , Cell Line, Tumor , Cricetulus , Gene Expression , HEK293 Cells , High-Throughput Screening Assays , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Kinetics , Ligands , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Protein Binding , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics
10.
Methods Mol Biol ; 1412: 103-11, 2016.
Article in English | MEDLINE | ID: mdl-27245896

ABSTRACT

Cannabinoid CB1 and CB2 receptors are G-protein-coupled receptors (GPCRs) that recruit ß-arrestins upon activation by (partial) agonists. ß-Arrestin recruitment is induced by phosphorylation of their C-terminal tails, and is associated with the termination of GPCR signaling; yet, it may also activate cellular signaling pathways independent of G-proteins. Here, we describe a detailed protocol to characterize the potency and efficacy of ligands to induce or inhibit ß-arrestin recruitment to the human CB1 and CB2 receptors, by using the PathHunter(®) assay. The latter is a cellular assay that can be performed in plates with 384-wells. The PathHunter(®) assay makes use of ß-galactosidase complementation, and has a chemiluminescent readout. We used this assay to characterize a set of reference ligands (both agonists and antagonists) on human CB1 and CB2 receptors.


Subject(s)
Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , beta-Arrestins/metabolism , Animals , Biological Assay/methods , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Gene Expression , Genes, Reporter , Ligands , Protein Binding , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
11.
J Biol Chem ; 291(6): 3053-62, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26601956

ABSTRACT

Peroxiredoxin 2 (Prx2) is a thiol protein that functions as an antioxidant, regulator of cellular peroxide concentrations, and sensor of redox signals. Its redox cycle is widely accepted to involve oxidation by a peroxide and reduction by thioredoxin/thioredoxin reductase. Interactions of Prx2 with other thiols are not well characterized. Here we show that the active site Cys residues of Prx2 form stable mixed disulfides with glutathione (GSH). Glutathionylation was reversed by glutaredoxin 1 (Grx1), and GSH plus Grx1 was able to support the peroxidase activity of Prx2. Prx2 became glutathionylated when its disulfide was incubated with GSH and when the reduced protein was treated with H2O2 and GSH. The latter reaction occurred via the sulfenic acid, which reacted sufficiently rapidly (k = 500 m(-1) s(-1)) for physiological concentrations of GSH to inhibit Prx disulfide formation and protect against hyperoxidation to the sulfinic acid. Glutathionylated Prx2 was detected in erythrocytes from Grx1 knock-out mice after peroxide challenge. We conclude that Prx2 glutathionylation is a favorable reaction that can occur in cells under oxidative stress and may have a role in redox signaling. GSH/Grx1 provide an alternative mechanism to thioredoxin and thioredoxin reductase for Prx2 recycling.


Subject(s)
Glutaredoxins , Glutathione , Peroxiredoxins , Protein Processing, Post-Translational/physiology , Animals , Catalytic Domain , Cell Line , Cysteine , Glutaredoxins/chemistry , Glutaredoxins/genetics , Glutaredoxins/metabolism , Glutathione/chemistry , Glutathione/genetics , Glutathione/metabolism , Humans , Hydrogen Peroxide/chemistry , Mice , Mice, Knockout , Oxidative Stress/drug effects , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/chemistry , Thioredoxins/genetics , Thioredoxins/metabolism
12.
Br J Pharmacol ; 173(3): 446-58, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26398481

ABSTRACT

BACKGROUND AND PURPOSE: Here, we have characterized 3-cyclopropyl-1-(4-(6-((1,1-dioxidothiomorpholino)methyl)-5-fluoropyridin-2-yl)benzyl)imidazolidine-2,4-dione hydrochloride (LEI-101) as a novel, peripherally restricted cannabinoid CB2 receptor agonist, using both in vitro and in vivo models. EXPERIMENTAL APPROACH: We investigated the effects of LEI-101 on binding and functional activity. We assessed its in vitro and in vivo selectivity. Efficacy of LEI-101 was determined in a mouse model of cisplatin-induced nephrotoxicity. KEY RESULTS: LEI-101 behaved as a partial agonist at CB2 receptors using ß-arrestin and GTPγS assays and was ~100-fold selective in CB2 /CB1 receptor-binding assays. It did not display any activity on endocannabinoid hydrolases and nor did it react with serine hydrolases in an activity-based protein profiling assay. In mice, LEI-101 had excellent oral bioavailability reaching high concentrations in the kidney and liver with minimal penetration into the brain. LEI-101 up to a dose of 60 mg·kg(-1) (p.o.) did not exert any CNS-mediated effects in the tetrad assay, in mice. LEI-101 (p.o. or i.p.) at 3 or 10 mg·kg(-1) dose-dependently prevented kidney dysfunction and/or morphological damage induced by cisplatin in mice. These protective effects were associated with improved renal histopathology, attenuated oxidative stress and inflammation in the kidney. These effects were absent in CB2 receptor knockout mice. CONCLUSION AND IMPLICATIONS: These results indicate that LEI-101 is a selective, largely peripherally restricted, orally available CB2 receptor agonist with therapeutic potential in diseases that are associated with inflammation and/or oxidative stress, including kidney disease.


Subject(s)
Imidazolidines/pharmacology , Kidney Diseases/drug therapy , Morpholines/pharmacology , Protective Agents/therapeutic use , Receptor, Cannabinoid, CB2/agonists , Administration, Oral , Animals , Apoptosis/drug effects , CHO Cells , Cisplatin/adverse effects , Cricetulus , DNA Fragmentation , Imidazolidines/chemistry , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lipid Peroxidation/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Morpholines/chemistry , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Receptor, Cannabinoid, CB2/genetics
13.
Proc Natl Acad Sci U S A ; 113(1): 26-33, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26668358

ABSTRACT

Diacylglycerol lipases (DAGLα and DAGLß) convert diacylglycerol to the endocannabinoid 2-arachidonoylglycerol. Our understanding of DAGL function has been hindered by a lack of chemical probes that can perturb these enzymes in vivo. Here, we report a set of centrally active DAGL inhibitors and a structurally related control probe and their use, in combination with chemical proteomics and lipidomics, to determine the impact of acute DAGL blockade on brain lipid networks in mice. Within 2 h, DAGL inhibition produced a striking reorganization of bioactive lipids, including elevations in DAGs and reductions in endocannabinoids and eicosanoids. We also found that DAGLα is a short half-life protein, and the inactivation of DAGLs disrupts cannabinoid receptor-dependent synaptic plasticity and impairs neuroinflammatory responses, including lipopolysaccharide-induced anapyrexia. These findings illuminate the highly interconnected and dynamic nature of lipid signaling pathways in the brain and the central role that DAGL enzymes play in regulating this network.


Subject(s)
Arachidonic Acids/metabolism , Brain/drug effects , Diglycerides/metabolism , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Glycerides/metabolism , Lipoprotein Lipase/antagonists & inhibitors , Neuronal Plasticity/drug effects , Animals , Brain/enzymology , Brain/metabolism , Enzyme Inhibitors/chemistry , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Cannabinoid/metabolism , Signal Transduction/drug effects
14.
Eur J Med Chem ; 101: 681-91, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26210506

ABSTRACT

We report the synthesis and biological evaluation of new derivatives of Capadenoson, a former drug candidate that was previously advanced to phase IIa clinical trials. 19 of the 20 ligands show an affinity below 100 nM at the human adenosine A1 receptor (hA1AR) and display a wide range of residence times at this target (from approx. 5 min (compound 10) up to 132 min (compound 5)). Structure-affinity and structure-kinetics relationships were established, and computational studies of a homology model of the hA1AR revealed crucial interactions for both the affinity and dissociation kinetics of this family of ligands. These results were also combined with global metrics (Ligand Efficiency, cLogP), showing the importance of binding kinetics as an additional way to better select a drug candidate amongst seemingly similar leads.


Subject(s)
Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/pharmacology , Aminopyridines/chemistry , Receptor, Adenosine A1/metabolism , Thiazoles/chemistry , Adenosine A1 Receptor Agonists/chemical synthesis , Aminopyridines/pharmacology , Dose-Response Relationship, Drug , Humans , Kinetics , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Thiazoles/pharmacology
15.
J Am Chem Soc ; 137(27): 8851-7, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26083464

ABSTRACT

Diacylglycerol lipase (DAGL)-α and -ß are enzymes responsible for the biosynthesis of the endocannabinoid 2-arachidonoylglycerol (2-AG). Selective and reversible inhibitors are required to study the function of DAGLs in neuronal cells in an acute and temporal fashion, but they are currently lacking. Here, we describe the identification of a highly selective DAGL inhibitor using structure-guided and a chemoproteomics strategy to characterize the selectivity of the inhibitor in complex proteomes. Key to the success of this approach is the use of comparative and competitive activity-based proteome profiling (ABPP), in which broad-spectrum and tailor-made activity-based probes are combined to report on the inhibition of a protein family in its native environment. Competitive ABPP with broad-spectrum fluorophosphonate-based probes and specific ß-lactone-based probes led to the discovery of α-ketoheterocycle LEI105 as a potent, highly selective, and reversible dual DAGL-α/DAGL-ß inhibitor. LEI105 did not affect other enzymes involved in endocannabinoid metabolism including abhydrolase domain-containing protein 6, abhydrolase domain-containing protein 12, monoacylglycerol lipase, and fatty acid amide hydrolase and did not display affinity for the cannabinoid CB1 receptor. Targeted lipidomics revealed that LEI105 concentration-dependently reduced 2-AG levels, but not anandamide levels, in Neuro2A cells. We show that cannabinoid CB1-receptor-mediated short-term synaptic plasticity in a mouse hippocampal slice model can be reduced by LEI105. Thus, we have developed a highly selective DAGL inhibitor and provide new pharmacological evidence to support the hypothesis that "on demand biosynthesis" of 2-AG is responsible for retrograde signaling.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/metabolism , Neurons/drug effects , Neurons/enzymology , Animals , Cell Line , Drug Discovery , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Mice , Synaptic Transmission/drug effects
16.
Free Radic Biol Med ; 77: 331-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25289458

ABSTRACT

The diterpenoid, adenanthin, represses tumor growth and prolongs survival in mouse promyelocytic leukemia models (Liu et al., Nat. Chem. Biol. 8, 486, 2012). It was proposed that this was done by inactivating peroxiredoxins (Prxs) 1 and 2 through the formation of an adduct specifically on the resolving Cys residue. We confirmed that adenanthin underwent Michael addition to isolated Prx2, thereby inhibiting oxidation to a disulfide-linked dimer. However, contrary to the original report, both the peroxidatic and the resolving Cys residues could be derivatized. Glutathione also formed an adenanthin adduct, reacting with a second-order rate constant of 25±5 M(-1) s(-1). With 50 µM adenanthin, the peroxidatic and resolving Cys of Prx2 reacted with half-times of 7 and 40 min, respectively, compared with 10 min for GSH. When erythrocytes or Jurkat T cells were treated with adenanthin, we saw no evidence for a reaction with Prxs 1 or 2. Instead, adenanthin caused time- and concentration-dependent loss of GSH followed by dimerization of the Prxs. Prxs undergo continuous oxidation in cells and are normally recycled by thioredoxin reductase and thioredoxin. Our results indicate that Prx reduction was inhibited. We observed rapid inhibition of purified thioredoxin reductase (half-time 5 min with 2 µM adenanthin) and in cells, thioredoxin reductase was much more sensitive than GSH and loss of both preceded accumulation of oxidized Prxs. Thus, adenanthin is not a specific Prx inhibitor, and its reported antitumor and anti-inflammatory effects are more likely to involve more general inhibition of thioredoxin and/or glutathione redox pathways.


Subject(s)
Diterpenes, Kaurane/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Homeodomain Proteins/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Diterpenes, Kaurane/chemistry , Enzyme Inhibitors/chemistry , Erythrocytes/drug effects , Erythrocytes/enzymology , Glutaredoxins/chemistry , Glutathione/chemistry , Homeodomain Proteins/chemistry , Humans , Jurkat Cells , Kinetics , Oxidation-Reduction , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/metabolism
17.
J Med Chem ; 56(23): 9427-40, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24224763

ABSTRACT

Cardiotoxicity is a side effect that plagues modern drug design and is very often due to the off-target blockade of the human ether-à-go-go related gene (hERG) potassium channel. To better understand the structural determinants of this blockade, we designed and synthesized a series of 40 derivatives of clofilium, a class III antiarrhythmic agent. These were evaluated in radioligand binding and patch-clamp assays to establish structure-affinity relationships (SAR) for this potassium channel. Efforts were especially focused on studying the influence of the structural rigidity and the nature of the linkers composing the clofilium scaffold. It was shown that introducing triple bonds and oxygen atoms in the n-butyl linker of the molecule greatly reduced affinity without significantly modifying the pKa of the essential basic nitrogen. These findings could prove useful in the first stages of drug discovery as a systematic way of reducing the risk of hERG K(+) channel blockade-induced cardiotoxicity.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Ether-A-Go-Go Potassium Channels/drug effects , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Quaternary Ammonium Compounds/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...