Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Biochem Pharmacol ; 180: 114144, 2020 10.
Article in English | MEDLINE | ID: mdl-32653590

ABSTRACT

Partial agonists for G protein-coupled receptors (GPCRs) provide opportunities for novel pharmacotherapies with enhanced on-target safety compared to full agonists. For the human adenosine A1 receptor (hA1AR) this has led to the discovery of capadenoson, which has been in phase IIa clinical trials for heart failure. Accordingly, the design and profiling of novel hA1AR partial agonists has become an important research focus. In this study, we report on LUF7746, a capadenoson derivative bearing an electrophilic fluorosulfonyl moiety, as an irreversibly binding hA1AR modulator. Meanwhile, a nonreactive ligand bearing a methylsulfonyl moiety, LUF7747, was designed as a control probe in our study. In a radioligand binding assay, LUF7746's apparent affinity increased to nanomolar range with longer pre-incubation time, suggesting an increasing level of covalent binding over time. Moreover, compared to the reference full agonist CPA, LUF7746 was a partial agonist in a hA1AR-mediated G protein activation assay and resistant to blockade with an antagonist/inverse agonist. An in silico structure-based docking study combined with site-directed mutagenesis of the hA1AR demonstrated that amino acid Y2717.36 was the primary anchor point for the covalent interaction. Additionally, a label-free whole-cell assay was set up to identify LUF7746's irreversible activation of an A1 receptor-mediated cell morphological response. These results led us to conclude that LUF7746 is a novel covalent hA1AR partial agonist and a valuable chemical probe for further mapping the receptor activation process. It may also serve as a prototype for a therapeutic approach in which a covalent partial agonist may cause less on-target side effects, conferring enhanced safety compared to a full agonist.


Subject(s)
Adenosine A1 Receptor Agonists/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Drug Design , Drug Partial Agonism , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Protein Structure, Secondary , Radioligand Assay/methods , Receptor, Adenosine A1/chemistry
2.
J Comput Aided Mol Des ; 34(6): 697-707, 2020 06.
Article in English | MEDLINE | ID: mdl-32112287

ABSTRACT

Among still comparatively few G protein-coupled receptors, the adenosine A2A receptor has been co-crystallized with several ligands, agonists as well as antagonists. It can thus serve as a template with a well-described orthosteric ligand binding region for adenosine receptors. As not all subtypes have been crystallized yet, and in order to investigate the usability of homology models in this context, multiple adenosine A1 receptor (A1AR) homology models had been previously obtained and a library of lead-like compounds had been docked. As a result, a number of potent and one selective ligand toward the intended target have been identified. However, in in vitro experimental verification studies, many ligands also bound to the A2AAR and the A3AR subtypes. In this work we asked the question whether a classification of the ligands according to their selectivity was possible based on docking scores. Therefore, we built an A3AR homology model and docked all previously found ligands to all three receptor subtypes. As a metric, we employed an in vitro/in silico selectivity ranking system based on taxicab geometry and obtained a classification model with reasonable separation. In the next step, the method was validated with an external library of, selective ligands with similarly good performance. This classification system might also be useful in further screens.


Subject(s)
Protein Conformation , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A3/chemistry , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Antagonists/chemistry , Binding Sites/drug effects , Humans , Ligands , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding/drug effects , Protein Conformation/drug effects , Receptor, Adenosine A1/ultrastructure , Receptor, Adenosine A2A/ultrastructure , Receptor, Adenosine A3/ultrastructure , Structure-Activity Relationship
3.
Pharmacol Biochem Behav ; 181: 110-116, 2019 06.
Article in English | MEDLINE | ID: mdl-31054946

ABSTRACT

Repetitive behaviors are diagnostic for autism spectrum disorder (ASD) and commonly observed in other neurodevelopmental disorders. Currently, there are no effective pharmacological treatments for repetitive behavior in these clinical conditions. This is due to the lack of information about the specific neural circuitry that mediates the development and expression of repetitive behavior. Our previous work in mouse models has linked repetitive behavior to decreased activation of the subthalamic nucleus, a brain region in the indirect and hyperdirect pathways in the basal ganglia circuitry. The present experiments were designed to further test our hypothesis that pharmacological activation of the indirect pathway would reduce repetitive behavior. We used a combination of adenosine A1 and A2A receptor agonists that have been shown to alter the firing frequency of dorsal striatal neurons within the indirect pathway of the basal ganglia. This drug combination markedly and selectively reduced repetitive behavior in both male and female C58 mice over a six-hour period, an effect that required both A1 and A2A agonists as neither alone reduced repetitive behavior. The adenosine A1 and A2A receptor agonist combination also significantly increased the number of Fos transcripts and Fos positive cells in dorsal striatum. Fos induction was found in both direct and indirect pathway neurons suggesting that the drug combination restored the balance of activation across these complementary basal ganglia pathways. The adenosine A1 and A2A receptor agonist combination also maintained its effectiveness in reducing repetitive behavior over a 7-day period. These findings point to novel potential therapeutic targets for development of drug therapies for repetitive behavior in clinical disorders.


Subject(s)
Adenosine A1 Receptor Agonists/therapeutic use , Adenosine A2 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Compulsive Behavior/drug therapy , Phenethylamines/therapeutic use , Stereotyped Behavior/drug effects , Adenosine/administration & dosage , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/administration & dosage , Adenosine A2 Receptor Agonists/chemistry , Analysis of Variance , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Behavior, Animal/drug effects , Corpus Striatum/cytology , Drug Therapy, Combination , Female , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neurons/metabolism , Peanut Oil/chemistry , Peanut Oil/pharmacology , Phenethylamines/administration & dosage , Phenethylamines/chemistry , Phenotype , Proto-Oncogene Proteins c-fos/metabolism
4.
Sci Rep ; 8(1): 16836, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442899

ABSTRACT

Despite intense interest in designing positive allosteric modulators (PAMs) as selective drugs of the adenosine A1 receptor (A1AR), structural binding modes of the receptor PAMs remain unknown. Using the first X-ray structure of the A1AR, we have performed all-atom simulations using a robust Gaussian accelerated molecular dynamics (GaMD) technique to determine binding modes of the A1AR allosteric drug leads. Two prototypical PAMs, PD81723 and VCP171, were selected. Each PAM was initially placed at least 20 Å away from the receptor. Extensive GaMD simulations using the AMBER and NAMD simulation packages at different acceleration levels captured spontaneous binding of PAMs to the A1AR. The simulations allowed us to identify low-energy binding modes of the PAMs at an allosteric site formed by the receptor extracellular loop 2 (ECL2), which are highly consistent with mutagenesis experimental data. Furthermore, the PAMs stabilized agonist binding in the receptor. In the absence of PAMs at the ECL2 allosteric site, the agonist sampled a significantly larger conformational space and even dissociated from the A1AR alone. In summary, the GaMD simulations elucidated structural binding modes of the PAMs and provided important insights into allostery in the A1AR, which will greatly facilitate the receptor structure-based drug design.


Subject(s)
Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/pharmacology , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Antagonists/chemistry , Adenosine A1 Receptor Antagonists/pharmacology , Allosteric Regulation , Allosteric Site , Binding Sites , Molecular Dynamics Simulation , Structure-Activity Relationship
5.
J Med Chem ; 61(22): 9966-9975, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30359014

ABSTRACT

Central adenosine A1 receptor (A1R) is implicated in pain, sleep, substance use disorders, and neurodegenerative diseases, and is an important target for pharmaceutical development. Radiotracers for A1R positron emission tomography (PET) would enable measurement of the dynamic interaction of endogenous adenosine and A1R during the sleep-awake cycle. Although several human A1R PET tracers have been developed, most are xanthine-based antagonists that failed to demonstrate competitive binding against endogenous adenosine. Herein, we explored non-nucleoside (3,5-dicyanopyridine and 5-cyanopyrimidine) templates for developing an agonist A1R PET radiotracer. We synthesized novel analogues, including 2-amino-4-(3-methoxyphenyl)-6-(2-(6-methylpyridin-2-yl)ethyl)pyridine-3,5-dicarbonitrile (MMPD, 22b), a partial A1R agonist of sub-nanomolar affinity. [11C]22b showed suitable blood-brain barrier (BBB) permeability and test-retest reproducibility. Regional brain uptake of [11C]22b was consistent with known brain A1R distribution and was blocked significantly by A1R but not A2AR ligands. [11C]22b is the first BBB-permeable A1R partial agonist PET radiotracer with the promise of detecting endogenous adenosine fluctuations.


Subject(s)
Adenosine A1 Receptor Agonists/metabolism , Positron-Emission Tomography , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/chemistry , Blood-Brain Barrier/metabolism , HEK293 Cells , Humans , Ligands , Structure-Activity Relationship
6.
Comput Biol Chem ; 75: 74-81, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29747078

ABSTRACT

Organophosphorus nerve agents (NAs) irreversibly inhibit acetylcholinesterase (AChE), the enzyme responsible for breaking down the neurotransmitter acetylcholine (ACh). The over accumulation of ACh after NA exposure leads to cholinergic toxicity, seizure, and death. Current medical countermeasures effectively mitigate peripheral symptoms, however; the brain is often unprotected. Alternative acute treatment with the adenosine A1 receptor agonist N6-cyclopentyladensosine (CPA) has previously been demonstrated to prevent AChE inhibition as well as to suppress neuronal activity. The mechanism of AChE protection is unknown. To elucidate the feasibility of potential CPA-AChE interaction mechanisms, we applied a truncated molecular model approach and density functional theory. The candidate mechanisms studied are reversible enzyme inhibition, enzyme reactivation, and NA blocking prior to enzyme conjugation. Our thermodynamic data suggest that CPA can compete with the NAs sarin and soman for the active site of AChE, but may, in contrast to NAs, undergo back-reaction. We found a strong interaction between CPA and NA conjugated AChE, making enzyme reactivation unlikely but possibly allowing for CPA protection through the prevention of NA aging. The data also indicates that there is an affinity between CPA and unbound NAs. The results from this study support the hypothesis that CPA counters NA toxicity via multiple mechanisms and is a promising therapeutic strategy that warrants further development.


Subject(s)
Acetylcholinesterase/metabolism , Adenosine A1 Receptor Agonists/metabolism , Adenosine/analogs & derivatives , Nerve Agents/metabolism , Organophosphorus Compounds/metabolism , Sarin/metabolism , Soman/metabolism , Adenosine/chemistry , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/pharmacology , Animals , Molecular Structure , Nerve Agents/chemistry , Nerve Agents/pharmacology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Quantum Theory , Rats , Sarin/chemistry , Sarin/pharmacology , Soman/chemistry , Soman/pharmacology , Thermodynamics
7.
J Med Chem ; 61(1): 305-318, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29257884

ABSTRACT

Our previous work discovered that combining the appropriate 5'- and N6-substitution in adenosine derivatives leads to the highly selective human A1 adenosine receptor (hA1AR) agonists or highly potent dual hA1AR agonists and hA3AR antagonists. In order to explore novel dual adenosine receptor ligands, a series of N6-substituted-5'-pyrazolyl-adenosine and 2-chloro-adenosine derivatives were synthesized and assayed in vitro at all ARs. The N6-(±)-endo-norbornyl derivative 12 was the most potent and selective at A1AR and effective as an analgesic in formalin test in mice, but none of the 5'-pyrazolyl series compounds showed a dual behavior at hA1 and hA3AR. Molecular modeling studies rationalized the structure-activity relationships and the selectivity profiles of the new series of A1AR agonists. Interestingly, an unexpected inverted binding mode of the N6-tetrahydrofuranyl derivative 14 was hypothesized to explain its low affinity at A1AR.


Subject(s)
Adenosine/chemical synthesis , Adenosine/pharmacology , Drug Design , Receptor, Adenosine A1/metabolism , Adenosine/chemistry , Adenosine/metabolism , Adenosine A1 Receptor Agonists/chemical synthesis , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenylyl Cyclases/metabolism , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/metabolism , Analgesics/pharmacology , Animals , Chemistry Techniques, Synthetic , Male , Mice , Molecular Docking Simulation , Protein Conformation , Receptor, Adenosine A1/chemistry , Structure-Activity Relationship
8.
ChemMedChem ; 12(10): 728-737, 2017 05 22.
Article in English | MEDLINE | ID: mdl-28488817

ABSTRACT

Adenosine is known to be released under a variety of physiological and pathophysiological conditions to facilitate the protection and regeneration of injured ischemic tissues. The activation of myocardial adenosine A1 receptors (A1 Rs) has been shown to inhibit myocardial pathologies associated with ischemia and reperfusion injury, suggesting several options for new cardiovascular therapies. When full A1 R agonists are used, the desired protective and regenerative cardiovascular effects are usually overshadowed by unintended pharmacological effects such as induction of bradycardia, atrioventricular (AV) blocks, and sedation. These unwanted effects can be overcome by using partial A1 R agonists. Starting from previously reported capadenoson we evaluated options to tailor A1 R agonists to a specific partiality range, thereby optimizing the therapeutic window. This led to the identification of the potent and selective agonist neladenoson, which shows the desired partial response on the A1 R, resulting in cardioprotection without sedative effects or cardiac AV blocks. To circumvent solubility and formulation issues for neladenoson, a prodrug approach was pursued. The dipeptide ester neladenoson bialanate hydrochloride showed significantly improved solubility and exposure after oral administration. Neladenoson bialanate hydrochloride is currently being evaluated in clinical trials for the treatment of heart failure.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Dipeptides/pharmacology , Heart Diseases/drug therapy , Prodrugs/pharmacology , Pyridines/pharmacology , Receptor, Adenosine A1/metabolism , Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A1 Receptor Agonists/chemistry , Administration, Oral , Animals , Chronic Disease , Dipeptides/administration & dosage , Dipeptides/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Injections, Intravenous , Molecular Structure , Prodrugs/administration & dosage , Prodrugs/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Rats , Solubility , Structure-Activity Relationship
9.
J Med Chem ; 60(10): 4327-4341, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28447789

ABSTRACT

Structural determinants of affinity of N6-substituted-5'-C-(ethyltetrazol-2-yl)adenosine and 2-chloroadenosine derivatives at adenosine receptor (AR) subtypes were studied with binding and molecular modeling. Small N6-cycloalkyl and 3-halobenzyl groups furnished potent dual acting A1AR agonists and A3AR antagonists. 4 was the most potent dual acting human (h) A1AR agonist (Ki = 0.45 nM) and A3AR antagonist (Ki = 0.31 nM) and highly selective versus A2A; 11 and 26 were most potent at both h and rat (r) A3AR. All N6-substituted-5'-C-(ethyltetrazol-2-yl)adenosine derivatives proved to be antagonists at hA3AR but agonists at the rA3AR. Analgesia of 11, 22, and 26 was evaluated in the mouse formalin test (A3AR antagonist blocked and A3AR agonist strongly potentiated). N6-Methyl-5'-C-(ethyltetrazol-2-yl)adenosine (22) was most potent, inhibiting both phases, as observed combining A1AR and A3AR agonists. This study demonstrated for the first time the advantages of a single molecule activating two AR pathways both leading to benefit in this acute pain model.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Analgesics/chemistry , Analgesics/pharmacology , Purinergic P1 Receptor Agonists/chemistry , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/chemistry , Purinergic P1 Receptor Antagonists/pharmacology , Acute Pain/drug therapy , Adenosine/therapeutic use , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Agonists/therapeutic use , Adenosine A3 Receptor Antagonists/chemistry , Adenosine A3 Receptor Antagonists/pharmacology , Adenosine A3 Receptor Antagonists/therapeutic use , Analgesics/therapeutic use , Animals , Humans , Mice , Models, Molecular , Purinergic P1 Receptor Agonists/therapeutic use , Purinergic P1 Receptor Antagonists/therapeutic use , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Purinergic P1/metabolism
10.
Cell ; 168(5): 867-877.e13, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235198

ABSTRACT

The adenosine A1 receptor (A1-AR) is a G-protein-coupled receptor that plays a vital role in cardiac, renal, and neuronal processes but remains poorly targeted by current drugs. We determined a 3.2 Å crystal structure of the A1-AR bound to the selective covalent antagonist, DU172, and identified striking differences to the previously solved adenosine A2A receptor (A2A-AR) structure. Mutational and computational analysis of A1-AR revealed a distinct conformation of the second extracellular loop and a wider extracellular cavity with a secondary binding pocket that can accommodate orthosteric and allosteric ligands. We propose that conformational differences in these regions, rather than amino-acid divergence, underlie drug selectivity between these adenosine receptor subtypes. Our findings provide a molecular basis for AR subtype selectivity with implications for understanding the mechanisms governing allosteric modulation of these receptors, allowing the design of more selective agents for the treatment of ischemia-reperfusion injury, renal pathologies, and neuropathic pain.


Subject(s)
Receptor, Adenosine A1/chemistry , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Antagonists/chemistry , Allosteric Site , Crystallography, X-Ray , Drug Design , Humans , Receptor, Adenosine A1/genetics , Receptor, Adenosine A2A/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL