Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Int J Mol Sci ; 23(9)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35562894

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a degenerating disease involving the motor neurons, which causes a progressive loss of movement ability, usually leading to death within 2 to 5 years from the diagnosis. Much effort has been put into research for an effective therapy for its eradication, but still, no cure is available. The only two drugs approved for this pathology, Riluzole and Edaravone, are onlyable to slow down the inevitable disease progression. As assessed in the literature, drug targets such as protein kinases have already been extensively examined as potential drug targets for ALS, with some molecules already in clinical trials. Here, we focus on the involvement of another very important and studied class of biological entities, G protein-coupled receptors (GPCRs), in the onset and progression of ALS. This workaimsto give an overview of what has been already discovered on the topic, providing useful information and insights that can be used by scientists all around the world who are putting efforts into the fight against this very important neurodegenerating disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/drug therapy , Edaravone/therapeutic use , Humans , Motor Neurons , Receptors, G-Protein-Coupled , Riluzole/therapeutic use
2.
Int J Mol Sci ; 22(18)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34575906

ABSTRACT

Fragment-Based Drug Discovery (FBDD) has become, in recent years, a consolidated approach in the drug discovery process, leading to several drug candidates under investigation in clinical trials and some approved drugs. Among these successful applications of the FBDD approach, kinases represent a class of targets where this strategy has demonstrated its real potential with the approved kinase inhibitor Vemurafenib. In the Kinase family, protein kinase CK1 isoform δ (CK1δ) has become a promising target in the treatment of different neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In the present work, we set up and applied a computational workflow for the identification of putative fragment binders in large virtual databases. To validate the method, the selected compounds were tested in vitro to assess the CK1δ inhibition.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Casein Kinase Idelta/chemistry , Drug Discovery/methods , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship , Workflow
3.
Molecules ; 24(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614517

ABSTRACT

Human A3 adenosine receptor hA3AR has been implicated in gastrointestinal cancer, where its cellular expression has been found increased, thus suggesting its potential as a molecular target for novel anticancer compounds. Observation made in our previous work indicated the importance of the carbonyl group of amide in the indolylpyrimidylpiperazine (IPP) for its human A2A adenosine receptor (hA2AAR) subtype binding selectivity over the other AR subtypes. Taking this observation into account, we structurally modified an indolylpyrimidylpiperazine (IPP) scaffold, 1 (a non-selective adenosine receptors' ligand) into a modified IPP (mIPP) scaffold by switching the position of the carbonyl group, resulting in the formation of both ketone and tertiary amine groups in the new scaffold. Results showed that such modification diminished the A2A activity and instead conferred hA3AR agonistic activity. Among the new mIPP derivatives (3-6), compound 4 showed potential as a hA3AR partial agonist, with an Emax of 30% and EC50 of 2.89 ± 0.55 µM. In the cytotoxicity assays, compound 4 also exhibited higher cytotoxicity against both colorectal and liver cancer cells as compared to normal cells. Overall, this new series of compounds provide a promising starting point for further development of potent and selective hA3AR partial agonists for the treatment of gastrointestinal cancers.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Pyrimidinones/chemistry , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A3/genetics , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Humans , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Piperazine/chemical synthesis , Piperazine/chemistry , Piperazine/pharmacology , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacology , Receptor, Adenosine A2A/chemistry , Structure-Activity Relationship
4.
Bioorg Med Chem ; 22(5): 1751-65, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24518296

ABSTRACT

In the present study, a molecular simplification approach was employed to design novel bicyclic pyrazolo[3,4-d]pyrimidine (PP) derivatives from tricyclic pyrazolo[4,3-e]-1,2,4-triazolo-[1,5-c]pyrimidines (PTP) as promising human A3 adenosine receptor (hA3AR) antagonists. All the target compounds were synthesized using novel and efficient synthetic schemes and the structure-activity relationship studies of these PPs were explored through the synthesis of a series of PTP analogues with various substituents. Substituents with different lipophilicity and steric hindrance (e.g., alkyl and aryl-alkyl) functions were introduced at N(2) position of the pyrazole ring, while acyl groups with different electronic properties were introduced at C(6) position of the bicyclic nucleus to probe both electronic and positional effects. Most of the synthesized derivatives of the PP series presented good affinity at the hA3AR, as indicated by the low micromolar range of Ki values and among them, compound 63 with N(2) neopentyl substituents showed most potent hA3AR affinity with Ki value of 0.9 µM and high selectivity (hA1AR/hA3AR=>111 & hA2AAR/hA3AR=>111) towards other adenosine receptor subtypes. Interestingly, small isopropyl groups at N(2) position displayed high affinity at another receptor subtype (hA2AAR, e.g., compound 55, with Ki hA2AAR=0.8 µM), while they were less favorable at the hA3AR. Molecular docking analysis was also performed to predict the possible binding mode of target compounds inside the hA3AR and hA2AAR. Overall, PP derivatives represent promising starting points for new AR antagonists.


Subject(s)
Purinergic P1 Receptor Antagonists/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Models, Molecular , Molecular Docking Simulation , Structure-Activity Relationship
5.
ChemMedChem ; 19(10): e202300641, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38329692

ABSTRACT

Rare diseases are particular pathological conditions affecting a limited number of people and few drugs are known to be effective as therapeutic treatment. Gaucher disease, caused by a deficiency of the lysosomal enzyme glucocerebrosidase, belongs to this class of disorders, and it is considered the most common among the Lysosomal Storage Diseases. The two main therapeutic approaches are the Enzyme Replacement Therapy (ERT) and the Substrate Reduction Therapy (SRT). ERT, consisting in replacing the defective enzyme by administering a recombinant enzyme, is effective in alleviating the visceral symptoms, hallmarks of the most common subtype of the disease whereas it has no effects when symptoms involve CNS, since the recombinant protein is unable to significantly cross the Blood Brain Barrier. The SRT strategy involves inhibiting glucosylceramide synthase (GCS), the enzyme responsible for the production of the associated storage molecule. The rational design of new inhibitors of GCS has been hampered by the lack of either the crystal structure of the enzyme or an in-silico model of the active site which could provide important information regarding the interactions of potential inhibitors with the target, but, despite this, interesting results have been obtained and are herein reviewed.


Subject(s)
Enzyme Inhibitors , Gaucher Disease , Gaucher Disease/drug therapy , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Chemistry, Pharmaceutical , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/metabolism , Glucosylceramidase/chemistry , Enzyme Replacement Therapy , Molecular Structure
6.
Pharmaceuticals (Basel) ; 17(4)2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38675428

ABSTRACT

Protein kinase CK1δ (CK1δ) is a serine-threonine/kinase that modulates different physiological processes, including the cell cycle, DNA repair, and apoptosis. CK1δ overexpression, and the consequent hyperphosphorylation of specific proteins, can lead to sleep disorders, cancer, and neurodegenerative diseases. CK1δ inhibitors showed anticancer properties as well as neuroprotective effects in cellular and animal models of Parkinson's and Alzheimer's diseases and amyotrophic lateral sclerosis. To obtain new ATP-competitive CK1δ inhibitors, three sets of benzimidazole-2-amino derivatives were synthesized (1-32), bearing different substituents on the fused benzo ring (R) and diverse pyrazole-containing acyl moieties on the 2-amino group. The best-performing derivatives were those featuring the (1H-pyrazol-3-yl)-acetyl moiety on the benzimidazol-2-amino scaffold (13-32), which showed CK1δ inhibitor activity in the low micromolar range. Among the R substituents, 5-cyano was the most advantageous, leading to a compound endowed with nanomolar potency (23, IC50 = 98.6 nM). Molecular docking and dynamics studies were performed to point out the inhibitor-kinase interactions.

7.
Med Res Rev ; 33(2): 235-335, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22095687

ABSTRACT

Adenosine is an ubiquitous local modulator that regulates various physiological and pathological functions by stimulating four membrane receptors, namely A(1), A(2A), A(2B), and A(3). Among these G protein-coupled receptors, the A(3) subtype is found mainly in the lung, liver, heart, eyes, and brain in our body. It has been associated with cerebroprotection and cardioprotection, as well as modulation of cellular growth upon its selective activation. On the other hand, its inhibition by selective antagonists has been reported to be potentially useful in the treatment of pathological conditions including glaucoma, inflammatory diseases, and cancer. In this review, we focused on the pharmacology and the therapeutic implications of the human (h)A(3) adenosine receptor (AR), together with an overview on the progress of hA(3) AR agonists, antagonists, allosteric modulators, and radioligands, as well as on the recent advances pertaining to the computational approaches (e.g., quantitative structure-activity relationships, homology modeling, molecular docking, and molecular dynamics simulations) applied to the modeling of hA(3) AR and drug design.


Subject(s)
Chemistry, Pharmaceutical/methods , Molecular Targeted Therapy/methods , Purinergic P1 Receptor Agonists/pharmacology , Receptor, Adenosine A3/chemistry , Receptor, Adenosine A3/drug effects , Animals , Brain Diseases/diagnosis , Brain Diseases/drug therapy , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Drug Design , Humans , Inflammation/diagnosis , Inflammation/drug therapy , Receptor, Adenosine A3/metabolism , Sensitivity and Specificity , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 23(1): 26-36, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23200243

ABSTRACT

Interest is increasing in developing fluorescent ligands for characterization of adenosine receptors (ARs), which hold a promise of usefulness in the drug discovery process. The size of a strategically labeled AR ligand can be greatly increased after the attachment of a fluorophore. The choice of dye moiety (e.g. Alexa Fluor 488), attachment point and linker length can alter the selectivity and potency of the parent molecule. Fluorescent derivatives of adenosine agonists and antagonists (e.g. XAC and other heterocyclic antagonist scaffolds) have been synthesized and characterized pharmacologically. Some are useful AR probes for flow cytometry, fluorescence correlation spectroscopy, fluorescence microscopy, fluorescence polarization, fluorescence resonance energy transfer, and scanning confocal microscopy. Thus, the approach of fluorescent labeled GPCR ligands, including those for ARs, is a growing dynamic research field.


Subject(s)
Fluorescent Dyes/chemistry , Ligands , Receptors, Purinergic P1/chemistry , Boron Compounds/chemistry , Fluorescein-5-isothiocyanate/chemistry , Humans , Protein Binding , Purinergic P1 Receptor Agonists/chemistry , Purinergic P1 Receptor Agonists/metabolism , Purinergic P1 Receptor Antagonists/chemistry , Purinergic P1 Receptor Antagonists/metabolism , Receptors, Purinergic P1/metabolism
9.
J Chem Inf Model ; 53(7): 1620-37, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-23705857

ABSTRACT

The application of both structure- and ligand-based design approaches represents to date one of the most useful strategies in the discovery of new drug candidates. In the present paper, we investigated how the application of docking-driven conformational analysis can improve the predictive ability of 3D-QSAR statistical models. With the use of the crystallographic structure in complex with the high affinity antagonist ZM 241385 (4-(2-[7-amino-2-(2-furyl)[1,2,4]-triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol), we revisited a general pharmacophore hypothesis for the human A(2A) adenosine receptor of a set of 751 known antagonists, by applying an integrated ligand- and structure-based approach. Our novel pharmacophore hypothesis has been validated by using an external test set of 29 newly synthesized human adenosine receptor antagonists.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Drug Design , Quantitative Structure-Activity Relationship , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists/metabolism , Humans , Molecular Docking Simulation , Protein Conformation , Receptor, Adenosine A2A/chemistry , Static Electricity , Triazines/chemistry , Triazines/metabolism , Triazines/pharmacology , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
10.
ChemMedChem ; 18(14): e202300109, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37114338

ABSTRACT

Traditionally, molecular recognition between the orthosteric site of adenosine receptors and their endogenous ligand occurs with a 1 : 1 stoichiometry. Inspired by previous mechanistic insights derived from supervised molecular dynamics (SuMD) simulations, which suggested an alternative 2 : 1 binding stoichiometry, we synthesized BRA1, a bis-ribosyl adenosine derivative, tested its ability to bind to and activate members of the adenosine receptor family, and rationalized its activity through molecular modeling.


Subject(s)
Adenosine , Molecular Dynamics Simulation , Adenosine/chemistry , Receptors, Purinergic P1 , Ligands
11.
Biomolecules ; 13(11)2023 11 03.
Article in English | MEDLINE | ID: mdl-38002292

ABSTRACT

Adenosine receptors are largely distributed in our organism and are promising therapeutic targets for the treatment of many pathologies. In this perspective, investigating the structural features of the ligands leading to affinity and/or selectivity is of great interest. In this work, we have focused on a small series of pyrazolo-triazolo-pyrimidine antagonists substituted in positions 2, 5, and N8, where bulky acyl moieties at the N5 position and small alkyl groups at the N8 position are associated with affinity and selectivity at the A3 adenosine receptor even if a good affinity toward the A2B adenosine receptor has also been observed. Conversely, a free amino function at the 5 position induces high affinity at the A2A and A1 receptors with selectivity vs. the A3 subtype. A molecular modeling study suggests that differences in affinity toward A1, A2A, and A3 receptors could be ascribed to two residues: one in the EL2, E168 in human A2A/E172 in human A1, that is occupied by the hydrophobic residue V169 in the human A3 receptor; and the other in TM6, occupied by H250/H251 in human A2A and A1 receptors and by a less bulky S247 in the A3 receptor. In the end, these findings could help to design new subtype-selective adenosine receptor ligands.


Subject(s)
Purinergic P1 Receptor Antagonists , Receptors, Purinergic P1 , Humans , Structure-Activity Relationship , Purinergic P1 Receptor Antagonists/pharmacology , Models, Molecular , Pyrimidines/pharmacology , Pyrimidines/chemistry
12.
ChemMedChem ; 18(21): e202300299, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37675643

ABSTRACT

The A3 adenosine receptor is an interesting target whose role in cancer is controversial. In this work, a structural investigation at the 2-position of the [1,2,4]triazolo[1,5-c]pyrimidine nucleus was performed, finding new potent and selective A3 adenosine receptor antagonists such as the ethyl 2-(4-methoxyphenyl)-5-(methylamino)-[1,2,4]triazolo[1,5-c]pyrimidine-8-carboxylate (20, DZ123) that showed a Ki value of 0.47 nM and an exceptional selectivity profile over the other adenosine receptor subtypes. Computational studies were performed to rationalize the affinity and the selectivity profile of the tested compounds at the A3 adenosine receptor and the A1 and A2A adenosine receptors. Compound 20 was tested on both A3 adenosine receptor positive cell lines (CHO-A3 AR transfected, THP1 and HCT16) and on A3 negative cancer cell lines, showing no effect in the latter and a pro-proliferative effect at a low concentration in the former. These interesting results pave the way to further investigation on both the mechanism involved and potential therapeutic applications.


Subject(s)
Neoplasms , Receptor, Adenosine A3 , Cricetinae , Animals , Structure-Activity Relationship , Receptor, Adenosine A3/metabolism , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/metabolism , Cell Line , Pyrimidines/chemistry , Purinergic P1 Receptor Antagonists/pharmacology , Purinergic P1 Receptor Antagonists/chemistry , CHO Cells , Receptor, Adenosine A2A
13.
Pharmaceuticals (Basel) ; 16(2)2023 Jan 23.
Article in English | MEDLINE | ID: mdl-37259317

ABSTRACT

Based on a screening of a chemical library of A2A adenosine receptor (AR) antagonists, a series of di- and tri-substituted adenine derivatives were synthesized and tested for their ability to inhibit the activity of the enzyme casein kinase 1 delta (CK1δ) and to bind adenosine receptors (ARs). Some derivatives, here called "dual anta-inhibitors", demonstrated good CK1δ inhibitory activity combined with a high binding affinity, especially for the A2AAR. The N6-methyl-(2-benzimidazolyl)-2-dimethyamino-9-cyclopentyladenine (17, IC50 = 0.59 µM and KiA2A = 0.076 µM) showed the best balance of A2AAR affinity and CK1δ inhibitory activity. Computational studies were performed to simulate, at the molecular level, the protein-ligand interactions involving the compounds of our series. Hence, the dual anta-inhibitor 17 could be considered the lead compound of new therapeutic agents endowed with synergistic effects for the treatment of chronic neurodegenerative and cancer diseases.

14.
Mol Pharm ; 9(3): 591-604, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22292533

ABSTRACT

We propose a potential antiparkinsonian prodrug DP-L-A(2A)ANT (2) obtained by amidic conjugation of dopamine (1) via a succinic spacer to a new triazolo-triazine A(2A) adenosine receptor (AR) antagonist A(2A)ANT (3). The affinity of 2 and its hydrolysis products-1, 3, dopamine-linker DP-L (4) and A(2A)ANT-linker L-A(2A)ANT (5)-was evaluated for hA(1), hA(2A), hA(2B) and hA(3) ARs and rat striatum A(2A)ARs or D(2) receptors. The hydrolysis patterns of 2, 4 and 5 and the stabilities of 1 and 3 were evaluated by HPLC analysis in human whole blood and rat brain homogenates. High hA(2A) affinity was shown by compounds 2 (K(i) = 7.32 ± 0.65 nM), 3 (K(i) = 35 ± 3 nM) and 5 (K(i) = 72 ± 5 nM), whose affinity values were similar in rat striatum. These compounds were not able to change dopamine affinity for D(2) receptors but counteracted the CGS 21680-induced reduction of dopamine affinity. DP-L (4) was inactive on adenosine and dopaminergic receptors. As for stability studies, compounds 4 and 5 were not degraded in incubation media. In human blood, the prodrug 2 was hydrolyzed (half-life = 2.73 ± 0.23 h) mainly on the amidic bound coupling the A(2A)ANT (3), whereas in rat brain homogenates the prodrug 2 was hydrolyzed (half-life > eight hours) exclusively on the amidic bound coupling dopamine, allowing its controlled release and increasing its poor stability as characterized by half-life = 22.5 ± 1.5 min.


Subject(s)
Adenosine A2 Receptor Antagonists/chemistry , Dopamine/chemistry , Animals , CHO Cells , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cricetinae , Cyclic AMP/metabolism , Humans , Kinetics , Parkinsonian Disorders/drug therapy , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology , Rats
15.
Curr Med Chem ; 29(27): 4631-4697, 2022.
Article in English | MEDLINE | ID: mdl-35170406

ABSTRACT

BACKGROUND: GSK-3ß activity has been strictly related to neuroinflammation and neurodegeneration. Alzheimer's disease is the most studied neurodegenerative disease, but GSK-3ß seems to be involved in almost all neurodegenerative diseases, including Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia, Huntington's disease, and the autoimmune disease multiple sclerosis. OBJECTIVE: This review aims to help researchers both working on this research topic or not to have a comprehensive overview of GSK-3ß in the context of neuroinflammation and neurodegeneration. METHODS: Literature has been searched using PubMed and SciFinder databases by inserting specific keywords. A total of more than 500 articles have been discussed. RESULTS: First of all, the structure and regulation of the kinase were briefly discussed, and then, specific GSK-3ß implications in neuroinflammation and neurodegenerative diseases were illustrated with the help of figures, to conclude with a comprehensive overview on the most important GSK-3ß and multitarget inhibitors. The structure and IC50 values at the target kinase have been reported for all the discussed compounds. CONCLUSION: GSK-3ß is involved in several signaling pathways in neurons, glial cells and immune cells. The fine regulation and interconnection of all these pathways are at the base of the rationale use of GSK-ß inhibitors in neuroinflammation and neurodegeneration. Some compounds are now under clinical trials. Despite this, the compounds' pharmacodynamic and ADME/Tox profiles were often not fully characterized which is deleterious in such a complex system.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neuroinflammatory Diseases , Parkinson Disease/metabolism , Signal Transduction
16.
ACS Chem Neurosci ; 13(15): 2252-2260, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35868251

ABSTRACT

Polypharmacology is a new trend in amyotrophic lateral sclerosis (ALS) therapy and an effective way of addressing a multifactorial etiology involving excitotoxicity, mitochondrial dysfunction, oxidative stress, and microglial activation. Inspired by a reported clinical trial, we converted a riluzole (1)-rasagiline (2) combination into single-molecule multi-target-directed ligands. By a ligand-based approach, the highly structurally integrated hybrids 3-8 were designed and synthesized. Through a target- and phenotypic-based screening pipeline, we identified hit compound 6. It showed monoamine oxidase A (MAO-A) inhibitory activity (IC50 = 6.9 µM) rationalized by in silico studies as well as in vitro brain permeability. By using neuronal and non-neuronal cell models, including ALS-patient-derived cells, we disclosed for 6 a neuroprotective/neuroinflammatory profile similar to that of the parent compounds and their combination. Furthermore, the unexpected MAO inhibitory activity of 1 (IC50 = 8.7 µM) might add a piece to the puzzle of its anti-ALS molecular profile.


Subject(s)
Amyotrophic Lateral Sclerosis , Neuroprotective Agents , Amyotrophic Lateral Sclerosis/drug therapy , Humans , Indans , Ligands , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Riluzole/pharmacology , Riluzole/therapeutic use
17.
Bioorg Med Chem Lett ; 21(10): 2898-905, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21511471

ABSTRACT

A ligand-based pharmacophore was obtained for a new series of 2-unsubstituted and 2-(para-substituted)phenyl-pyrazolo-triazolo-pyrimidines as potent human A(3) adenosine receptor antagonists. Through comparative molecular field analysis-based quantitative structure-activity relationship studies, structural features at the N(5)-, N(8)- and C(2)-positions of the tricyclic nucleus were deeply investigated, with emphasis given to the unprecedentedly explored C(2)-position. The resulting model showed good correlation and predictability (r(2)=0.936; q(2)=0.703; r(pred)(2)=0.663). Overall, the contribution of steric effect was found relatively more predominant for the optimal interaction of these antagonists to the human A(3) receptor.


Subject(s)
Adenosine A3 Receptor Antagonists , Pyrazoles/chemistry , Pyrimidines , Triazoles/chemistry , Adenosine A3 Receptor Antagonists/chemistry , Adenosine A3 Receptor Antagonists/pharmacology , Humans , Models, Molecular , Molecular Structure , Protein Binding/drug effects , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quantitative Structure-Activity Relationship
18.
Bioorg Med Chem ; 19(20): 6120-34, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21908194

ABSTRACT

In an attempt to study the optimal combination of a phenyl ring at the C(2)-position and different substituents at the N(5)- and N(8)-positions towards the selective modulation of human A(3) adenosine receptors (hA(3)AR), we synthesized a new series of 2-para-(un)substituted-phenyl-pyrazolo-triazolo-pyrimidines bearing either a methyl or phenylethyl at N(8) and chains of variable length at N(5). Through biological evaluation, it was found that the majority of the compounds had good affinities towards the hA(3)AR in the low nanomolar range. Compound 16 possessed the best hA(3)AR affinity and selectivity profile (K(i)hA(3)=1.33 nM; hA(1)/hA(3)=4880; hA(2A)/hA(3)=1100) in the present series of 2-(substituted)phenyl-pyrazolo-triazolo-pyrimidine derivatives. In addition to pharmacological characterization, a molecular modeling investigation on these compounds further elucidated the effect of different substituents at the pyrazolo-triazolo-pyrimidine scaffold on affinity and selectivity to hA(3)AR.


Subject(s)
Pyrazoles/chemistry , Pyridines/chemistry , Receptor, Adenosine A3/chemistry , Triazoles/chemistry , Animals , CHO Cells , Cricetinae , Humans , Models, Molecular , Structure-Activity Relationship
19.
RSC Med Chem ; 12(2): 254-262, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-34046614

ABSTRACT

A3 adenosine receptors were found to have a role in different pathological states, such as glaucoma, renal fibrosis, neuropathic pain and cancer. Consequently, it is important to utilize any molecular tool which could help to study these conditions. In the present study we continue our search for potent A3 adenosine receptor ligands which could be successively conjugated to other molecules with the aim of obtaining more potent (e.g. allosteric ligand conjugation) or detectable ligands (e.g. fluorescent molecule or biotin conjugation). Specifically, different aminoester moieties were introduced at the 5 position of the pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine core. The ester functionalization represents the candidate for the subsequent conjugation. All the reported compounds are potent hA3 adenosine receptor antagonists and some of them exhibited high selectivity against the other adenosine receptors. The main structural terms of ligand recognition and selectivity were disclosed by molecular modelling studies. Molecular docking results led to the characterization of an alternative binding mode for antagonists at the orthosteric binding site of the hA3 adenosine receptor, evaluated and assessed by classical molecular dynamics simulations.

20.
Eur J Med Chem ; 216: 113331, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33721670

ABSTRACT

Protein kinase CK1δ expression and activity is involved in different pathological situations that include neuroinflammatory and neurodegenerative diseases. For this reason, protein kinase CK1δ has become a possible therapeutic target for these conditions. 5,6-fused bicyclic heteroaromatic systems that resemble adenine of ATP represent optimal scaffolds for the development of a new class of ATP competitive CK1δ inhibitors. In particular, a new series of [1,2,4]triazolo[1,5-c]pyrimidines and [1,2,4]triazolo[1,5-a][1,3,5]triazines was developed. Some crucial interactors have been identified, such as the presence of a free amino group able to interact with the residues of the hinge region at the 5- and 7- positions of the [1,2,4]triazolo[1,5-c]pyrimidine and [1,2,4]triazolo[1,5-a][1,3,5]triazine scaffolds, respectively; or the presence of a 3-hydroxyphenyl or 3,5-dihydroxyphenyl moiety at the 2- position of both nuclei. Molecular modeling studies identified the key interactions involved in the inhibitor-protein recognition process that appropriately fit with the outlined structure-activity relationship. Considering the fact that the CK1 protein kinase is involved in various pathologies in particular of the central nervous system, the interest in the development of new inhibitors permeable to the blood-brain barrier represents today an important goal in the pharmaceutical field. The best potent compound of the series is the 5-(7-amino-5-(benzylamino)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-2-yl)benzen-1,3-diol (compound 51, IC50 = 0.18 µM) that was predicted to have an intermediate ability to cross the membrane in our in vitro assay and represents an optimal starting point to both studies the therapeutic value of protein kinase CK1δ inhibition and to develop new more potent derivatives.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Triazoles/chemistry , Binding Sites , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Casein Kinase Idelta/metabolism , Cell Line , Cell Survival/drug effects , Drug Design , Humans , Kinetics , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Static Electricity , Structure-Activity Relationship , Thermodynamics , Triazines/chemistry , Triazoles/metabolism , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL