Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 867
Filter
1.
Biol Chem ; 404(4): 279-289, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36215695

ABSTRACT

GluN2B-NMDA receptors play a key role in several neurological and neurodegenerative disorders. In order to develop novel negative allosteric GluN2B-NMDA receptor modulators, the concept of conformational restriction was pursued, i.e. the flexible aminoethanol substructure of ifenprodil was embedded into a more rigid tetrahydro-3-benzazepine system. The resulting tetrahydro-3-benzazepine-1,7-diol (±)-2 (WMS-1410) showed promising receptor affinity in receptor binding studies (K i = 84 nM) as well as pharmacological activity in two-electrode-voltage-clamp experiments (IC 50 = 116 nM) and in cytoprotective assays (IC 50 = 18.5 nM). The interactions of (R)-2 with the ifenprodil binding site of GluN2B-NMDA receptors were analyzed on the molecular level and the "foot-in-the-door" mechanism was developed. Due to promising pharmacokinetic parameters (logD7.4 = 1.68, plasma protein binding of 76-77%, sufficient metabolic stability) F-substituted analogs were prepared and evaluated as tracers for positron emission tomography (PET). Both fluorine-18-labeled PET tracers [18F]11 and [18F]15 showed high brain uptake, specific accumulation in regions known for high GluN2B-NMDA receptor expression, but no interactions with σ 1 receptors. Radiometabolites were not observed in the brain. Both PET tracers might be suitable for application in humans.


Subject(s)
Positron-Emission Tomography , Receptors, N-Methyl-D-Aspartate , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Positron-Emission Tomography/methods , Benzazepines/pharmacology , Benzazepines/chemistry , Benzazepines/metabolism
2.
J Labelled Comp Radiopharm ; 65(14): 354-360, 2022 12.
Article in English | MEDLINE | ID: mdl-36261868

ABSTRACT

Diabetes mellitus (DM) and insulinoma are mainly affected by the status of pancreatic ß-cell mass (BCM). Development of imaging agents for BCM allows to study pancreatic ß cells and the relationship between ß cells and DM or insulinoma. In this study, we investigated the density of dopamine D1 receptor on the ß cells and measured BCM by statistical image processing. The pancreatic uptakes of [125 I]I-R-(+)-7-chloro-8-hydroxy-1-(3'-iodopheny1)-3-methyl-2,3,4,5-tetrahydro-1H-3-benzazepine ([125 I]I-R-(+)-TISCH), dopamine D1 receptor tracer, in normal and diabetic rats displayed significant differences at 30 min (1.11 ± 0.08% ID/g vs. 0.63 ± 0.09% ID/g, p < 0.0001). In the presence of SCH23390, the pancreatic uptake of [125 I]I-R-(+)-TISCH at 30 min in normal rats was lower (1.01 ± 0.04% ID/g, p < 0.05). Although the blocking was not complete, [125 I]I-R-(+)-TISCH showed specific binding signals to the pancreas. Furthermore, the uptakes of [125 I]I-R-(+)-TISCH in INS-1 cells were reduced in the presence of SCH23390 at different concentrations. [125 I]I-R-(+)-TISCH displayed a respectable uptake in insulinoma. Overall, [125 I]I-R-(+)-TISCH provided specific binding signals to pancreatic ß cells. Although the specific signal may not be sufficient for imaging in vivo, the dopamine D1 receptor can still be considered as a potential target for studying BCM. Further investigation will be required to optimize the ligand.


Subject(s)
Diabetes Mellitus, Experimental , Insulin-Secreting Cells , Insulinoma , Pancreatic Neoplasms , Animals , Rats , Dopamine , Receptors, Dopamine D1/metabolism , Ligands , Insulin-Secreting Cells/metabolism , Benzazepines/metabolism
3.
Arch Pharm (Weinheim) ; 355(6): e2200020, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35224754

ABSTRACT

To improve the metabolic stability and receptor selectivity of ifenprodil (1), the benzoxazolone moiety of besonprodil (2) and the 3-benzazepone moiety of WMS-1410 (3) were merged to obtain oxazolobenzazepines of type 4. The 5-(hydroxyethyl)benzoxazolone 7 representing the first key intermediate was prepared in four steps starting with the 4-(2-hydroxyethyl)phenol (8). Mitsunobu reaction of primary alcohol 7 with N-sulfonylated glycine esters established the necessary side chain. The intramolecular Friedel-Crafts acylation of acid 12a containing the N-tosyl protective group led upon decarbonylation exclusively to the tricyclic tetrahydroisoquinoline 14. Protection of the amino moiety by the stronger electron-withdrawing triflyl group resulted in the desired 3-benzazepine 15 without the formation of analogous isoquinoline. The triflyl protective group was cleaved off by K2 CO3 -induced elimination of trifluoromethanesulfinate. In a one-pot three-step procedure, various oxazolobenzazepinediones 15 were obtained, which were reduced to afford the desired secondary alcohols 18.


Subject(s)
Benzazepines , Receptors, N-Methyl-D-Aspartate , Benzazepines/metabolism , Benzazepines/pharmacology , Phenols , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship
4.
Int J Mol Sci ; 22(23)2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34884931

ABSTRACT

Neuroblastoma is a severe childhood disease, accounting for ~10% of all infant cancers. The amplification of the MYCN gene, coding for the N-Myc transcription factor, is an essential marker correlated with tumor progression and poor prognosis. In neuroblastoma cells, the mitotic kinase Aurora-A (AURKA), also frequently overexpressed in cancer, prevents N-Myc degradation by directly binding to a highly conserved N-Myc region. As a result, elevated levels of N-Myc are observed. During recent years, it has been demonstrated that some ATP competitive inhibitors of AURKA also cause essential conformational changes in the structure of the activation loop of the kinase that prevents N-Myc binding, thus impairing the formation of the AURKA/N-Myc complex. In this study, starting from a screening of crystal structures of AURKA in complexes with known inhibitors, we identified additional compounds affecting the conformation of the kinase activation loop. We assessed the ability of such compounds to disrupt the interaction between AURKA and N-Myc in vitro, using Surface Plasmon Resonance competition assays, and in tumor cell lines overexpressing MYCN, by performing Proximity Ligation Assays. Finally, their effects on N-Myc cellular levels and cell viability were investigated. Our results identify PHA-680626 as an amphosteric inhibitor both in vitro and in MYCN overexpressing cell lines, thus expanding the repertoire of known conformational disrupting inhibitors of the AURKA/N-Myc complex and confirming that altering the conformation of the activation loop of AURKA with a small molecule is an effective strategy to destabilize the AURKA/N-Myc interaction in neuroblastoma cancer cells.


Subject(s)
Aurora Kinase A/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrroles/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/chemistry , Azepines/metabolism , Azepines/pharmacology , Benzazepines/metabolism , Benzazepines/pharmacology , Binding Sites , Binding, Competitive , Cell Line , Drug Evaluation, Preclinical/methods , Humans , N-Myc Proto-Oncogene Protein/chemistry , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Pyrimidines/pharmacology , Pyrroles/metabolism , Surface Plasmon Resonance
5.
J Pharmacol Exp Ther ; 379(3): 235-244, 2021 11.
Article in English | MEDLINE | ID: mdl-34593560

ABSTRACT

Participation of N-methyl-d-aspartate (NMDA) receptors (NMDARs) in the failure of pancreatic ß cells during development of type 2 diabetes mellitus is discussed. Our study investigates whether ß cell mass and function can be preserved by selectively addressing the GluN2B subunit of the NMDAR. NMDAR activation by NMDA and its coagonist glycine moderately influenced electrical activity and Ca2+ handling in islet cells at a threshold glucose concentration (4-5 mM) without affecting glucose-mediated insulin secretion. Exposure of islet cells to NMDA/glycine or a glucolipotoxic milieu increased apoptosis by 5% and 8%, respectively. The GluN2B-specific NMDAR antagonist WMS-1410 (0.1 and 1 µM) partly protected against this. In addition, WMS-1410 completely prevented the decrease in insulin secretion of about 32% provoked by a 24-hour-treatment with NMDA/glycine. WMS-1410 eliminated NMDA-induced changes in the oxidation status of the islet cells and elevated the sensitivity of intracellular calcium to 15 mM glucose. By contrast, WMS-1410 did not prevent the decline in glucose-stimulated insulin secretion occurring after glucolipotoxic culture. This lack of effect was due to a decrease in insulin content to 18% that obviously could not be compensated by the preservation of cell mass or the higher percentage of insulin release in relation to insulin content. In conclusion, the negative effects of permanent NMDAR activation were effectively counteracted by WMS-1410 as well as the apoptotic cell death induced by high glucose and lipid concentrations. Modulation of NMDARs containing the GluN2B subunit is suggested to preserve ß cell mass during development of type 2 diabetes mellitus. SIGNIFICANCE STATEMENT: Addressing NMDA receptors containing the GluN2B subunit in pancreatic islet cells has the potential to protect the ß cell mass that progressively declines during the development of type 2 diabetes. Furthermore, this study shows that harmful effects of permanent NMDAR activation can be effectively counteracted by the compound WMS-1410, a selective modulator for NMDARs containing the GluN2B subunit.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Benzazepines/pharmacology , Cell Death/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Apoptosis/physiology , B-Lymphocytes/metabolism , Benzazepines/metabolism , Cell Death/physiology , Female , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Protein Subunits/metabolism , Protein Subunits/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism
6.
Sci Rep ; 11(1): 1535, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452311

ABSTRACT

The Toll-like receptor 8 (TLR8) agonist VTX-2337 (motolimod) is an anti-cancer immunotherapeutic agent that is believed to augment natural killer (NK) and dendritic cell (DC) activity. The goal of this work is to examine the role of TLR8 expression/activity in head and neck squamous cell carcinoma (HNSCC) to facilitate the prediction of responders to VTX-2337-based therapy. The prognostic role of TLR8 expression in HNSCC patients was assessed by TCGA and tissue microarray analyses. The anti-tumor effect of VTX-2337 was determined in SCCVII/C3H, mEERL/C57Bl/6 and TUBO-human EGFR/BALB/c syngeneic mouse models. The effect of combined VTX-2337 and cetuximab treatment on tumor growth, survival and immune cell recruitment was assessed. TLR8 expression was associated with CD8+ T cell infiltration and favorable survival outcomes. VTX-2337 delayed tumor growth in all 3 syngeneic mouse models and significantly increased the survival of cetuximab-treated mice. The anti-tumor effects of VTX-2337+ cetuximab were accompanied by increased splenic lymphoid DCs and IFNγ+ CD4+ and tumor-specific CD8+ T cells. Depletion of CD4+ T cells, CD8+ T cells and NK cells were all able to abolish the anti-tumor effect of VTX-2337+ cetuximab. Altogether, VTX-2337 remains promising as an adjuvant for cetuximab-based therapy however patients with high TLR8 expression may be more likely to derive benefit from this drug combination compared to patients with low TLR8 expression.


Subject(s)
Benzazepines/pharmacology , Cetuximab/pharmacology , T-Lymphocytes/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/pharmacology , Benzazepines/metabolism , Biomarkers, Pharmacological , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cetuximab/metabolism , Cytokines/metabolism , Databases, Genetic , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 8/drug effects , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism
7.
Int J Mol Sci ; 21(18)2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32899459

ABSTRACT

Extended-access methamphetamine self-administration results in unregulated intake of the drug; however, the role of dorsal striatal dopamine D1-like receptors (D1Rs) in the reinforcing properties of methamphetamine under extended-access conditions is unclear. Acute (ex vivo) and chronic (in vivo) methamphetamine exposure induces neuroplastic changes in the dorsal striatum, a critical region implicated in instrumental learning. For example, methamphetamine exposure alters high-frequency stimulation (HFS)-induced long-term depression in the dorsal striatum; however, the effect of methamphetamine on HFS-induced long-term potentiation (LTP) in the dorsal striatum is unknown. In the current study, dorsal striatal infusion of SCH23390, a D1R antagonist, prior to extended-access methamphetamine self-administration reduced methamphetamine addiction-like behavior. Reduced behavior was associated with reduced expression of PSD-95 in the dorsal striatum. Electrophysiological findings demonstrate that superfusion of methamphetamine reduced basal synaptic transmission and HFS-induced LTP in dorsal striatal slices, and SCH23390 prevented this effect. These results suggest that alterations in synaptic transmission and synaptic plasticity induced by acute methamphetamine via D1Rs could assist with methamphetamine-induced modification of corticostriatal circuits underlying the learning of goal-directed instrumental actions and formation of habits, mediating escalation of methamphetamine self-administration and methamphetamine addiction-like behavior.


Subject(s)
Benzazepines/pharmacology , Methamphetamine/adverse effects , Receptors, Dopamine D1/metabolism , Animals , Benzazepines/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Methamphetamine/metabolism , Neuronal Plasticity/drug effects , Rats , Rats, Long-Evans , Reinforcement, Psychology , Self Administration/methods , Synapses/physiology , Synaptic Transmission/drug effects
8.
Bioorg Med Chem ; 28(15): 115601, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32631570

ABSTRACT

Bromodomain-containing protein 4 (BRD4) is a key epigenetic regulator in cancer, and inhibitors targeting BRD4 exhibit great anticancer activity. By replacing the methyltriazole ring of the BRD4 inhibitor I-BET-762 with an N-methylthiazolidone heterocyclic ring, fifteen novel BRD4 inhibitors were designed and synthesized. Compound 13f had a hydrophobic acetylcyclopentanyl side chain, showing the most potent BRD4 inhibitory activity in the BRD4-BD1 inhibition assay (IC50 value of 110 nM), it also significantly suppressed the proliferation of MV-4-11 cells with high BRD4 level (IC50 value of 0.42 µM). Furthermore, the potent apoptosis-promoting and G0/G1 cycle-arresting activity of compound 13f were indicated by flow cytometry. As the downstream-protein of BRD4, c-Myc was in significantly low expression by compound 13f treatment in a dose-dependent manner. All the findings supported that this novel compound 13f provided a perspective for developing effective BRD4 inhibitors.


Subject(s)
Benzazepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Thiazoles/pharmacology , Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzazepines/chemical synthesis , Benzazepines/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Protein Binding , Thiazoles/chemical synthesis , Thiazoles/metabolism , Transcription Factors/metabolism
9.
Epilepsy Behav ; 105: 106989, 2020 04.
Article in English | MEDLINE | ID: mdl-32169824

ABSTRACT

OBJECTIVE: Adjunctive fenfluramine hydrochloride, classically described as acting pharmacologically through a serotonergic mechanism, has demonstrated a unique and robust clinical response profile with regard to its magnitude, consistency, and durability of effect on seizure activity in patients with pharmacoresistant Dravet syndrome. Recent findings also support long-term improvements in executive functions (behavior, emotion, cognition) in these patients. The observed clinical profile is inconsistent with serotonergic activity alone, as other serotonergic medications have not been demonstrated to have these clinical effects. This study investigated a potential role for σ1 receptor activity in complementing fenfluramine's serotonergic pharmacology. METHODS: Radioligand binding assays tested the affinity of fenfluramine for 47 receptors associated with seizures in the literature, including σ receptors. Cellular function assays tested fenfluramine and norfenfluramine (its major metabolite) activity at various receptors, including adrenergic, muscarinic, and serotonergic receptors. The σ1 receptor activity was assessed by the mouse vas deferens isometric twitch and by an assay of dissociation of the σ1 receptor from the endoplasmic reticulum stress protein binding immunoglobulin protein (BiP). In vivo mouse models assessed fenfluramine activity at σ1 receptors in ameliorating dizocilpine-induced learning deficits in spatial and nonspatial memory tasks, alone or in combination with the reference σ1 receptor agonist PRE-084. RESULTS: Fenfluramine and norfenfluramine bound ≥30% to ß2-adrenergic, muscarinic M1, serotonergic 5-HT1A, and σ receptors, as well as sodium channels, with a Ki between 266 nM (σ receptors) and 17.5 µM (ß-adrenergic receptors). However, only σ1 receptor isometric twitch assays showed a positive functional response, with weak stimulation by fenfluramine and inhibition by norfenfluramine. Fenfluramine, but not the 5-HT2C agonist lorcaserin, showed a positive modulation of the PRE-084-induced dissociation of σ1 protein from BiP. Fenfluramine also showed dose-dependent antiamnesic effects against dizocilpine-induced learning deficits in spontaneous alternation and passive avoidance responses, which are models of σ1 activation. Moreover, low doses of fenfluramine synergistically potentiated the low-dose effect of PRE-084, confirming a positive modulatory effect at the σ1 receptor. Finally, all in vivo effects were blocked by the σ1 receptor antagonist NE-100. SIGNIFICANCE: Fenfluramine demonstrated modulatory activity at σ1 receptors in vitro and in vivo in addition to its known serotonergic activity. These studies identify a possible new σ1 receptor mechanism underpinning fenfluramine's central nervous system effects, which may contribute to its antiseizure activity in Dravet syndrome and positive effects observed on executive functions in clinical studies.


Subject(s)
Fenfluramine/metabolism , Fenfluramine/pharmacology , Receptors, sigma/metabolism , Seizures/drug therapy , Seizures/metabolism , Animals , Benzazepines/metabolism , Benzazepines/pharmacology , CHO Cells , Cricetinae , Cricetulus , Fenfluramine/therapeutic use , HEK293 Cells , Humans , Male , Mice , Morpholines/metabolism , Morpholines/pharmacology , Protein Binding/physiology , Radioligand Assay/methods , Rats , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Sigma-1 Receptor
10.
J Biol Chem ; 295(10): 3285-3300, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31911436

ABSTRACT

Genetic and biochemical evidence points to an association between mitochondrial dysfunction and Parkinson's disease (PD). PD-associated mutations in several genes have been identified and include those encoding PTEN-induced putative kinase 1 (PINK1) and parkin. To identify genes, pathways, and pharmacological targets that modulate the clearance of damaged or old mitochondria (mitophagy), here we developed a high-content imaging-based assay of parkin recruitment to mitochondria and screened both a druggable genome-wide siRNA library and a small neuroactive compound library. We used a multiparameter principal component analysis and an unbiased parameter-agnostic machine-learning approach to analyze the siRNA-based screening data. The hits identified in this analysis included specific genes of the ubiquitin proteasome system, and inhibition of ubiquitin-conjugating enzyme 2 N (UBE2N) with a specific antagonist, Bay 11-7082, indicated that UBE2N modulates parkin recruitment and downstream events in the mitophagy pathway. Screening of the compound library identified kenpaullone, an inhibitor of cyclin-dependent kinases and glycogen synthase kinase 3, as a modulator of parkin recruitment. Validation studies revealed that kenpaullone augments the mitochondrial network and protects against the complex I inhibitor MPP+. Finally, we used a microfluidics platform to assess the timing of parkin recruitment to depolarized mitochondria and its modulation by kenpaullone in real time and with single-cell resolution. We demonstrate that the high-content imaging-based assay presented here is suitable for both genetic and pharmacological screening approaches, and we also provide evidence that pharmacological compounds modulate PINK1-dependent parkin recruitment.


Subject(s)
Mitochondria/metabolism , RNA, Small Interfering/metabolism , Small Molecule Libraries/metabolism , Ubiquitin-Protein Ligases/metabolism , Benzazepines/chemistry , Benzazepines/metabolism , Benzazepines/pharmacology , HeLa Cells , Humans , Hydrazones/chemistry , Hydrazones/metabolism , Hydrazones/pharmacology , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitophagy/drug effects , Principal Component Analysis , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
11.
Mini Rev Med Chem ; 20(9): 768-778, 2020.
Article in English | MEDLINE | ID: mdl-30961494

ABSTRACT

This review refers to the all-inclusive details of Lorcaserin Hydrochloride on comprehensive information about the synthesis, analytical methods, pharmacodynamics, pharmacokinetics, drug interactions and adverse effects. Lorcaserin Hydrochloride is chemically (R)-8-Chloro-1-methyl-2,3,4,5- tetrahydro-1H-3-benzazepine hydrochloride. Lorcaserin HCl is a novel, synthetic, centrally-acting selective serotonin C (5-HT2c) receptor, l agonist, which results in increased satiety and decreased food consumption in patients. Headache, dizziness and nausea are the most common side effects associated with this drug. Lorcaserin HCl has two major metabolites, one conjugated with glucuronide called N-carbamoyl glucuronide which is excreted in urine and the second Lorcaserin N-sulfamate, which is circulated in the blood. Lorcaserin HCl is synthesized using four different schemes of which a six-step method that resulted in 92.3% yield with 99.8% of purity is employed for scale-up production. It is analyzed quantitatively in the plasma and brain tissue matrix of rats by Ultra Performance Liquid chromatographic (UPLC) method using MS-MS (Mass Spectrometric) detection.


Subject(s)
Benzazepines/chemistry , Serotonin 5-HT2 Receptor Agonists/chemistry , Appetite Depressants/adverse effects , Appetite Depressants/chemistry , Appetite Depressants/metabolism , Appetite Depressants/therapeutic use , Benzazepines/adverse effects , Benzazepines/metabolism , Benzazepines/therapeutic use , Clinical Trials as Topic , Drug Interactions , Half-Life , Headache/etiology , Humans , Obesity/drug therapy , Obesity/pathology , Serotonin 5-HT2 Receptor Agonists/adverse effects , Serotonin 5-HT2 Receptor Agonists/metabolism , Serotonin 5-HT2 Receptor Agonists/therapeutic use
12.
J Hum Genet ; 65(2): 143-153, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31645655

ABSTRACT

Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. The aim of the present study was to elucidate the relative contributions of CYP3A4 and CYP3A5 to the metabolism of ASV, DCV, and BCV, as well as the effect of CYP3A5*3 genetic variant in vitro. The amount of each drug and their major metabolites were determined using LC-MS/MS. Recombinant CYP3As and CYP3A5*3-genotyped human liver microsomes (CYP3A5 expressers or non-expressers) were used for the determination of their metabolic activities. The contribution of CYP3A5 to ASV metabolism was considerable compared to that of CYP3A4. Consistently, ASV metabolic activity in CYP3A5 expressers was higher than those in CYP3A5 non-expresser. Moreover, CYP3A5 expression level was significantly correlated with ASV metabolism. In contrast, these observations were not found in DCV and BCV metabolism. To our knowledge, this is the first study to directly demonstrate the effect of CYP3A5*3 genetic variants on the metabolism of ASV. The findings of the present study may provide basic information on ASV, DCV, and BCV metabolisms.


Subject(s)
Antiviral Agents/metabolism , Benzazepines/metabolism , Cytochrome P-450 CYP3A/genetics , Imidazoles/metabolism , Indoles/metabolism , Isoquinolines/metabolism , Sulfonamides/metabolism , Benzazepines/chemistry , Carbamates , Chromatography, Liquid , Cytochrome P-450 CYP3A/metabolism , Genetic Variation , Genotype , Humans , Imidazoles/chemistry , Indoles/chemistry , Isoquinolines/chemistry , Liver/metabolism , Microsomes, Liver , Pyrrolidines , Recombinant Proteins , Sulfonamides/chemistry , Tandem Mass Spectrometry , Valine/analogs & derivatives
13.
J Med Chem ; 62(21): 9450-9470, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31657559

ABSTRACT

Aspiring to develop a positron emission tomography (PET) imaging agent for the GluN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), a key therapeutic target for drug development toward several neurological disorders, we synthesized a series of 2,3,4,5-tetrahydro-1H-3-benzazepine and 6,7,8,9-tetrahydro-5H-benzo[7]annulen-7-amine analogues. After in vitro testing via competition binding assay and autoradiography, [18F]PF-NB1 emerged as the best performing tracer with respect to specificity and selectivity over σ1 and σ2 receptors and was thus selected for further in vivo evaluation. Copper-mediated radiofluorination was accomplished in good radiochemical yields and high molar activities. Extensive in vivo characterization was performed in Wistar rats comprising PET imaging, biodistribution, receptor occupancy, and metabolites studies. [18F]PF-NB1 binding was selective to GluN2B-rich forebrain regions and was specifically blocked by the GluN2B antagonist, CP-101,606, in a dose-dependent manner with no brain radiometabolites. [18F]PF-NB1 is a promising fluorine-18 PET tracer for imaging the GluN2B subunits of the NMDAR and has utility for receptor occupancy studies.


Subject(s)
Amines/chemistry , Amines/metabolism , Benzazepines/chemistry , Benzazepines/metabolism , Halogenation , Positron-Emission Tomography/methods , Receptors, N-Methyl-D-Aspartate/metabolism , Amines/pharmacokinetics , Animals , Benzazepines/pharmacokinetics , Male , Protein Binding , Radiography , Rats , Rats, Wistar , Structure-Activity Relationship
14.
J Pharm Biomed Anal ; 167: 90-99, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30763883

ABSTRACT

Motolimod (VTX-2337) is an agonist of toll-like receptor 8 (TLR8) with potential immune-stimulating and antineoplastic activities. The purpose of this study was to investigate the in vitro metabolic profiles of VTX-2337. The average in vitro T1/2 values were 6.93, 8.71, 7.39, 2.85, and 10.58 min in the liver microsomes of mouse, rat, dog, monkey and human respectively, suggesting that VTX-2337 suffered from extensive metabolism. The metabolites were further profiled and identified by using ultra-high performance liquid chromatography coupled with diode array detector and Q-Exactive-Orbitrap tandem mass spectrometer (UHPLC-DAD-Q-Exactive-Orbitrap-MS) operated in positive ion mode. A total of 20 metabolites were detected and their identities were characterized based on their accurate masses, fragment ions and retention times. M13 (depropylation) was the most abundant metabolite in all species. M14 (oxygenation) was also the major metabolite in the liver microsomes of mouse, rat, monkey and human. M1, M5, M10, M15, and M16 were specifically detected in mouse, while M6 and M17 were monkey-specific. All the metabolites present in human could be found in animal species. The metabolic pathways of VTX-2337 referred to oxygenation, hydrolysis, depropylation, and dehydrogenation. Rat had the similar metabolic profiles to humans. The current study provided overall metabolic profiles of VTX-2337, which would be of great help in predicting in vivo pharmacokinetic profiles and in understanding the effectiveness and safety of this drug.


Subject(s)
Benzazepines/metabolism , Microsomes, Liver/metabolism , Toll-Like Receptor 8/agonists , Animals , Dogs , Drug Stability , Female , Humans , In Vitro Techniques , Macaca fascicularis , Male , Mice , Rats, Sprague-Dawley , Species Specificity
15.
Addict Biol ; 24(3): 376-387, 2019 05.
Article in English | MEDLINE | ID: mdl-29498158

ABSTRACT

Varenicline, a nicotinic acetylcholine receptor partial agonist, is used to treat nicotine dependence. Lorcaserin, a 5-HT2C receptor agonist has been approved in some countries to treat obesity. Based on preclinical and preliminary clinical evidence, lorcaserin may have potential to treat nicotine dependence. These experiments examined in rats the effects of combining varenicline (0.5 or 1 mg/kg) and lorcaserin (0.3, 0.6 and 1 mg/kg) on nicotine self-administration, reinstatement of nicotine seeking, responding for food and impulsive action. Both drugs alone reduced nicotine self-administration. Combining varenicline and 0.6 mg/kg lorcaserin reduced responding to a greater extent than either drug alone. In a relapse model, extinguished nicotine seeking was reinstated by a priming injection of nicotine and nicotine-associated cues. Reinstatement was reduced by varenicline (1 mg/kg) and by lorcaserin (0.3 mg/kg). Combining lorcaserin (0.3 mg/kg) with varenicline (0.5 or 1 mg/kg) reduced reinstatement to a greater degree than either drug alone. Both drugs had minimal effects on responding for food, alone or in combination. In the five-choice serial reaction time test, varenicline (0.5 or 1 mg/kg) increased impulsivity, measured as increased premature responding. This effect was reduced by lorcaserin (0.3 mg/kg). Plasma levels of varenicline or lorcaserin were not altered by co-administration of the other drug. Varenicline and lorcaserin have additive effects on nicotine self-administration, and on nicotine seeking. Lorcaserin prevents impulsivity induced by varenicline. This pattern of effects suggests that co-administration of varenicline and lorcaserin has potential as a treatment for nicotine dependence that may exceed the value of either drug alone.


Subject(s)
Benzazepines/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Smoking Cessation Agents/pharmacology , Tobacco Use Disorder/drug therapy , Varenicline/pharmacology , Animals , Benzazepines/metabolism , Conditioning, Operant/drug effects , Drug Combinations , Drug-Seeking Behavior/drug effects , Feeding Behavior/drug effects , Impulsive Behavior/drug effects , Male , Rats, Long-Evans , Reinforcement, Psychology , Smoking Cessation Agents/metabolism , Varenicline/metabolism
16.
J Med Chem ; 62(1): 128-140, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30525590

ABSTRACT

The five dopamine receptor subtypes (D1-5) are activated by the endogenous catecholamine dopamine. Sustained research has sought to identify selective ligands for receptor subtypes. In particular, activation of the D1 receptor has attracted attention due to its promising role in neurological diseases. Initial attempts to identify agonists yielded catechol derivatives, mimicking dopamine, with suboptimal DMPK parameters and low selectivity over the D5 subtype. However, more recent efforts to identify ligands capable of activating the D1 receptor have made substantial progress with the identification of non-catechol agonists with suitable properties to progress to clinical studies. In addition, several research groups have identified positive allosteric modulators that offer new potential. Furthermore, structural studies have surprisingly uncovered two potential allosteric binding sites, the most characterized of which appears to be on intracellular loop 2 (ICL2). This review highlights the recent progress in the field, covering both orthosteric and allosteric modes of activation, discusses the elucidation of the allosteric binding sites, and summarizes the clinical development status of various compounds.


Subject(s)
Receptors, Dopamine/chemistry , Allosteric Regulation , Animals , Benzazepines/chemistry , Benzazepines/metabolism , Binding Sites , Dopamine Agonists/chemistry , Dopamine Agonists/metabolism , Humans , Molecular Docking Simulation , Piperazine/analogs & derivatives , Piperazine/metabolism , Protein Structure, Tertiary , Receptors, Dopamine/metabolism
17.
Cell Metab ; 28(4): 619-630.e5, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30146485

ABSTRACT

To meet the challenge to human health posed by obesity, a better understanding of the regulation of feeding is essential. Medications targeting 5-hydroxytryptamine (5-HT; serotonin) 2C receptors (htr2c; 5-HT2CR) improve obesity. Here we probed the functional significance of 5-HT2CRs specifically within the brainstem nucleus of the solitary tract (5-HT2CRNTS) in feeding behavior. Selective activation of 5-HT2CRNTS decreased feeding and was sufficient to mediate acute food intake reductions elicited by the 5-HT2CR agonist obesity medication lorcaserin. Similar to pro-opiomelanocortin neurons expressed within the hypothalamic arcuate nucleus (POMCARC), a subset of POMCNTS neurons co-expressed 5-HT2CRs and were activated by 5-HT2CR agonists. Knockdown of POMCNTS prevented the acute appetite-suppressive effect of lorcaserin, whereas POMCARC knockdown prevented the full anorectic effect. These data identify 5-HT2CRNTS as a sufficient subpopulation of 5-HT2CRs in reducing food intake when activated and reveal that 5-HT2CR agonist obesity medications require POMC within the NTS and ARC to reduce food intake.


Subject(s)
Appetite Depressants/therapeutic use , Benzazepines/therapeutic use , Eating/physiology , Obesity/drug therapy , Serotonin 5-HT2 Receptor Agonists/therapeutic use , Solitary Nucleus/metabolism , Analysis of Variance , Animals , Appetite Depressants/metabolism , Appetite Regulation/drug effects , Arcuate Nucleus of Hypothalamus/cytology , Benzazepines/metabolism , Cell Line, Tumor , Feeding Behavior/physiology , Male , Mice , Mice, Knockout , Neurons/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/metabolism , Statistics, Nonparametric , Transfection
18.
Behav Brain Res ; 336: 145-150, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28842271

ABSTRACT

The sensorimotor cortex and the striatum are interconnected by the corticostriatal pathway, suggesting that cortical injury alters the striatal function that is associated with skilled movements and motor learning, which are functions that may be modulated by dopamine (DA). In this study, we explored motor coordination and balance in order to investigate whether the activation of D1 receptors (D1Rs) modulates functional recovery after cortical injury. The results of the beam-walking test showed motor deficit in the injured group at 24, 48 and 96h post-injury, and the recovery time was observed at 192h after cortical injury. In the sham and injured rats, systemic administration of the D1R antagonist SCH-23390 (1mg/kg) alone at 24, 48, 96 and 192h significantly (P<0.01) increased the motor deficit, while administration of the D1R agonist SKF-38393 alone (2, 3 and 4mg/kg) at 24, 48, 96 and 192h post-injury did not produce a significant difference; however, the co-administration of SKF-38393 and SCH-23390 prevented the antagonist-induced increase in the motor deficit. The cortical+striatal injury showed significantly increased the motor deficit at 24, 48, 96 and 192h post-injury (P<0.01) but did not show recovery at 192h. In conclusion, the administration of the D1R agonist did not accelerate the motor recovery, but the activation of D1Rs maintained motor coordination, confirming that an intact striatum may be necessary for achieving recovery.


Subject(s)
Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/physiology , Sensorimotor Cortex/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/metabolism , Benzazepines/pharmacology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Male , Motor Cortex/physiopathology , Neostriatum/metabolism , Rats , Rats, Wistar , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/metabolism , Sensorimotor Cortex/metabolism
19.
J Nucl Med ; 59(4): 698-703, 2018 04.
Article in English | MEDLINE | ID: mdl-29191857

ABSTRACT

Clinical and preclinical research with modulators at the N-methyl-d-aspartate (NMDA) receptor GluN2B N-terminal domain (NTD) aims for the treatment of various neurologic diseases. The interpretation of the results is hampered by the lack of a suitable NMDA PET tracer for assessing the receptor occupancy of potential drugs. We have developed 11C-Me-NB1 as a PET tracer for imaging GluN1/GluN2B-containing NMDA receptors and used it to investigate in rats the dose-dependent receptor occupancy of eliprodil, a GluN2B NTD modulator. Methods:11C-Me-NB1 was synthesized and characterized by in vitro displacement binding experiments with rat brain membranes, in vitro autoradiography, and blocking and displacement experiments by PET and PET kinetic modeling. Receptor occupancy by eliprodil was studied by PET with 11C-Me-NB1. Results:11C-Me-NB1 was synthesized at 290 ± 90 GBq/µmol molar activity, 7.4 ± 1.9 GBq total activity at the end of synthesis (n = 17), and more than 99% radiochemical purity. 11C-Me-NB1 binding in rat brain was blocked in vitro and in vivo by the NTD modulators Ro-25-6981 and eliprodil. Half-maximal receptor occupancy by eliprodil occurred at 1.5 µg/kg. At 1 mg/kg of eliprodil, a dose with reported neuroprotective effects, more than 99.5% of binding sites were occupied. In vitro, 11C-Me-NB1 binding was independent of the σ-1 receptor (Sigma1R), and the Sigma1R agonist (+)-pentazocine did not compete for high-affinity binding. In vivo, a 2.5 mg/kg dose of (+)-pentazocine abolished 11C-Me-NB1-specific binding, indicating an indirect effect of Sigma1R on 11C-Me-NB1 binding. Conclusion:11C-Me-NB1 is suitable for the in vivo imaging of NMDA GluN1/GluN2B receptors and the assessment of receptor occupancy by NTD modulators. GluN1/GluN2B NMDA receptors are fully occupied at neuroprotective doses of eliprodil. Furthermore, 11C-Me-NB1 enables imaging of GluN1/GluN2B NMDA receptor cross talk.


Subject(s)
Benzazepines/pharmacology , Positron-Emission Tomography/methods , Receptor Cross-Talk , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Benzazepines/metabolism , Image Processing, Computer-Assisted , Ketamine/pharmacology , Ligands , Male , Piperidines/pharmacology , Radioactive Tracers , Rats , Rats, Wistar , Receptor Cross-Talk/drug effects , Tissue Distribution
20.
Mol Metab ; 6(10): 1092-1102, 2017 10.
Article in English | MEDLINE | ID: mdl-29031711

ABSTRACT

OBJECTIVE: The increasing prevalence of type 2 diabetes (T2D) and associated morbidity and mortality emphasizes the need for a more complete understanding of the mechanisms mediating glucose homeostasis to accelerate the identification of new medications. Recent reports indicate that the obesity medication lorcaserin, a 5-hydroxytryptamine (5-HT, serotonin) 2C receptor (5-HT2CR) agonist, improves glycemic control in association with weight loss in obese patients with T2D. Here we evaluate whether lorcaserin has an effect on glycemia without body weight loss and how this effect is achieved. METHODS: Murine models of common and genetic T2D were utilized to probe the direct effect of lorcaserin on glycemic control. RESULTS: Lorcaserin dose-dependently improves glycemic control in mouse models of T2D in the absence of reductions in food intake or body weight. Examining the mechanism of this effect, we reveal a necessary and sufficient neurochemical mediator of lorcaserin's glucoregulatory effects, brain pro-opiomelanocortin (POMC) peptides. To clarify further lorcaserin's therapeutic brain circuit, we examined the receptor target of POMC peptides. We demonstrate that lorcaserin requires functional melanocortin4 receptors on cholinergic preganglionic neurons (MC4RChAT) to exert its effects on glucose homeostasis. In contrast, MC4RChAT signaling did not impact lorcaserin's effects on feeding, indicating a divergence in the neurocircuitry underpinning lorcaserin's therapeutic glycemic and anorectic effects. Hyperinsulinemic-euglycemic clamp studies reveal that lorcaserin reduces hepatic glucose production, increases glucose disposal and improves insulin sensitivity. CONCLUSIONS: These data suggest that lorcaserin's action within the brain represents a mechanistically novel treatment for T2D: findings of significance to a prevalent global disease.


Subject(s)
Benzazepines/pharmacology , Blood Glucose/drug effects , Receptor, Serotonin, 5-HT2C/drug effects , Animals , Benzazepines/metabolism , Body Weight/drug effects , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Eating/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Glucose Tolerance Test , Homeostasis/physiology , Humans , Insulin Resistance/physiology , Melanocortins/pharmacology , Mice , Mice, Transgenic , Obesity/drug therapy , Receptors, Melanocortin/drug effects , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...