Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 65(19): 13305-13327, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36173355

RESUMO

Drugs targeting adenosine receptors (AR) can provide treatment for diseases. We report the identification of 7-(phenylamino)-pyrazolo[3,4-c]pyridines L2-L10, A15, and A17 as low-micromolar to low-nanomolar A1R/A3R dual antagonists, with 3-phenyl-5-cyano-7-(trimethoxyphenylamino)-pyrazolo[3,4-c]pyridine (A17) displaying the highest affinity at both receptors with a long residence time of binding, as determined using a NanoBRET-based assay. Two binding orientations of A17 produce stable complexes inside the orthosteric binding area of A1R in molecular dynamics (MD) simulations, and we selected the most plausible orientation based on the agreement with alanine mutagenesis supported by affinity experiments. Interestingly, for drug design purposes, the mutation of L2506.51 to alanine increased the binding affinity of A17 at A1R. We explored the structure-activity relationships against A1R using alchemical binding free energy calculations with the thermodynamic integration coupled with the MD simulation (TI/MD) method, applied on the whole G-protein-coupled receptor-membrane system, which showed a good agreement (r = 0.73) between calculated and experimental relative binding free energies.


Assuntos
Antagonistas do Receptor A3 de Adenosina , Receptor A3 de Adenosina , Antagonistas do Receptor A3 de Adenosina/química , Alanina , Mutagênese , Antagonistas de Receptores Purinérgicos P1/química , Piridinas/química , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Receptor A3 de Adenosina/metabolismo , Relação Estrutura-Atividade
2.
J Med Chem ; 64(17): 12525-12536, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34435786

RESUMO

Distinguishing compounds' agonistic or antagonistic behavior would be of great utility for the rational discovery of selective modulators. We synthesized truncated nucleoside derivatives and discovered 6c (Ki = 2.40 nM) as a potent human A3 adenosine receptor (hA3AR) agonist, and subtle chemical modification induced a shift from antagonist to agonist. We elucidated this shift by developing new hA3AR homology models that consider the pharmacological profiles of the ligands. Taken together with molecular dynamics (MD) simulation and three-dimensional (3D) structural network analysis of the receptor-ligand complex, the results indicated that the hydrogen bonding with Thr943.36 and His2727.43 could make a stable interaction between the 3'-amino group with TM3 and TM7, and the corresponding induced-fit effects may play important roles in rendering the agonistic effect. Our results provide a more precise understanding of the compounds' actions at the atomic level and a rationale for the design of new drugs with specific pharmacological profiles.


Assuntos
Agonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/metabolismo , Agonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/química , Animais , Células CHO , Domínio Catalítico , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ligantes , Modelos Químicos , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Relação Estrutura-Atividade
3.
Sci Rep ; 10(1): 20781, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33247159

RESUMO

The adenosine A3 receptor (A3R) belongs to a family of four adenosine receptor (AR) subtypes which all play distinct roles throughout the body. A3R antagonists have been described as potential treatments for numerous diseases including asthma. Given the similarity between (adenosine receptors) orthosteric binding sites, obtaining highly selective antagonists is a challenging but critical task. Here we screen 39 potential A3R, antagonists using agonist-induced inhibition of cAMP. Positive hits were assessed for AR subtype selectivity through cAMP accumulation assays. The antagonist affinity was determined using Schild analysis (pA2 values) and fluorescent ligand binding. Structure-activity relationship investigations revealed that loss of the 3-(dichlorophenyl)-isoxazolyl moiety or the aromatic nitrogen heterocycle with nitrogen at α-position to the carbon of carboximidamide group significantly attenuated K18 antagonistic potency. Mutagenic studies supported by molecular dynamic simulations combined with Molecular Mechanics-Poisson Boltzmann Surface Area calculations identified the residues important for binding in the A3R orthosteric site. We demonstrate that K18, which contains a 3-(dichlorophenyl)-isoxazole group connected through carbonyloxycarboximidamide fragment with a 1,3-thiazole ring, is a specific A3R (< 1 µM) competitive antagonist. Finally, we introduce a model that enables estimates of the equilibrium binding affinity for rapidly disassociating compounds from real-time fluorescent ligand-binding studies. These results demonstrate the pharmacological characterisation of a selective competitive A3R antagonist and the description of its orthosteric binding mode. Our findings may provide new insights for drug discovery.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacocinética , Animais , Sítios de Ligação/genética , Ligação Competitiva , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ensaio Radioligante , Ratos , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/genética , Receptor A3 de Adenosina/metabolismo , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade
4.
J Med Chem ; 63(5): 2577-2587, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31738058

RESUMO

Adenosine receptors participate in many physiological functions. Molecules that may selectively interact with one of the receptors are favorable multifunctional chemical entities to treat or decelerate the evolution of different diseases. 3-Arylcoumarins have already been studied as neuroprotective agents by our group. Here, differently 8-substituted 3-arylcoumarins are complementarily studied as ligands of adenosine receptors, performing radioligand binding assays. Among the synthesized compounds, selective A3 receptor antagonists were found. 3-(4-Bromophenyl)-8-hydroxycoumarin (compound 4) displayed the highest potency and selectivity as A3 receptor antagonist (Ki = 258 nM). An analysis of its X-ray diffraction provided detailed information on its structure. Further evaluation of a selected series of compounds indicated that it is the nature and position of the substituents that determine their activity and selectivity. Theoretical modeling calculations corroborate and explain the experimental data, suggesting this novel scaffold can be involved in the generation of candidates as multitarget drugs.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Cumarínicos/química , Cumarínicos/farmacologia , Receptor A3 de Adenosina/metabolismo , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Modelos Moleculares , Receptor A3 de Adenosina/química , Relação Estrutura-Atividade
5.
J Chem Inf Model ; 59(12): 5183-5197, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31725294

RESUMO

Adenosine A3 receptor (A3R) is a promising drug target cancer and for a number of other conditions like inflammatory diseases, including asthma and rheumatoid arthritis, glaucoma, chronic obstructive pulmonary disease, and ischemic injury. Currently, there is no experimentally determined structure of A3R. We explored the binding profile of O4-{[3-(2,6-dichlorophenyl)-5-methylisoxazol-4-yl]carbonyl}-2-methyl-1,3-thiazole-4-carbohydroximamide (K18), which is a new specific and competitive antagonist at the orthosteric binding site of A3R. MD simulations and MM-GBSA calculations of the WT A3R in complex with K18 combined with in vitro mutagenic studies show that the most plausible binding conformation for the dichlorophenyl group of K18 is oriented toward trans-membrane helices (TM) 5, 6 and reveal important residues for binding. Further, MM-GBSA calculations distinguish mutations that reduce or maintain or increase antagonistic activity. Our studies show that selectivity of K18 toward A3R is defined not only by direct interactions with residues within the orthosteric binding area but also by remote residues playing a significant role. Although V1695.30 is considered to be a selectivity filter for A3R binders, when it was mutated to glutamic acid, K18 maintained antagonistic potency, in agreement with our previous results obtained for agonists binding profile investigation. Mutation of the direct interacting residue L903.32 in the low region and the remote L2647.35 in the middle/upper region to alanine increases antagonistic potency, suggesting an empty space in the orthosteric area available for increasing antagonist potency. These results approve the computational model for the description of K18 binding at A3R, which we previously performed for agonists binding to A3R, and the design of more effective antagonists based on K18.


Assuntos
Antagonistas do Receptor A3 de Adenosina/farmacologia , Simulação de Dinâmica Molecular , Mutagênese , Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/metabolismo , Amidas/química , Amidas/metabolismo , Amidas/farmacologia , Melfalan/metabolismo , Melfalan/farmacologia , Simulação de Acoplamento Molecular , Distribuição de Poisson , Ligação Proteica , Conformação Proteica , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/genética , Especificidade por Substrato , Termodinâmica , gama-Globulinas/metabolismo , gama-Globulinas/farmacologia
6.
Int J Mol Sci ; 20(14)2019 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-31315296

RESUMO

Adenosine receptors are a family of G protein-coupled receptors with increased attention as drug targets on different indications. We investigate the thermodynamics of ligand binding to the A3 adenosine receptor subtype, focusing on a recently reported series of diarylacetamidopyridine inhibitors via molecular dynamics simulations. With a combined approach of thermodynamic integration and one-step perturbation, we characterize the impact of the charge distribution in a central heteroaromatic ring on the binding affinity prediction. Standard charge distributions according to the GROMOS force field yield values in good agreement with the experimental data and previous free energy calculations. Subsequently, we examine the thermodynamics of inhibitor binding in terms of the energetic and entropic contributions. The highest entropy penalties are found for inhibitors with methoxy substituents in meta position of the aryl groups. This bulky group restricts rotation of aromatic rings attached to the pyrimidine core which leads to two distinct poses of the ligand. Our predictions support the previously proposed binding pose for the o-methoxy ligand, yielding in this case a very good correlation with the experimentally measured affinities with deviations below 4 kJ/mol.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor A3 de Adenosina/química , Sítios de Ligação , Ligação Proteica , Receptor A3 de Adenosina/metabolismo
7.
Arch Pharm Res ; 42(9): 773-779, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30264323

RESUMO

Truncated 4'-thionucleosides 1-4 and 4'-oxonucleosides 5-8 as potent and selective A3AR antagonists were synthesized from D-mannose and D-erythronic acid γ-lactone, respectively. These nucleosides were evaluated for their anti-fibrotic renoprotective activity in TGF-ß1-treated murine proximal tubular (mProx) cells. Their antagonistic activities for A3AR were proportional to their inhibitory activities against TGF-ß1-induced collagen I upregulation in mProx cells. This result suggests that the binding affinity of A3AR antagonists is closely correlated with their anti-fibrotic activity. Thus, A3AR antagonists might be novel therapeutic candidates for treating chronic kidney disease.


Assuntos
Antagonistas do Receptor A3 de Adenosina/farmacologia , Adenosina/farmacologia , Fibrose/tratamento farmacológico , Nefropatias/tratamento farmacológico , Receptor A3 de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/química , Antagonistas do Receptor A3 de Adenosina/síntese química , Antagonistas do Receptor A3 de Adenosina/química , Animais , Relação Dose-Resposta a Droga , Fibrose/metabolismo , Humanos , Nefropatias/metabolismo , Estrutura Molecular , Ratos , Relação Estrutura-Atividade
8.
J Med Chem ; 60(17): 7502-7511, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28792759

RESUMO

We report the first family of 2-acetamidopyridines as potent and selective A3 adenosine receptor (AR) antagonists. The computer-assisted design was focused on the bioisosteric replacement of the N1 atom by a CH group in a previous series of diarylpyrimidines. Some of the generated 2-acetamidopyridines elicit an antagonistic effect with excellent affinity (Ki < 10 nM) and outstanding selectivity profiles, providing an alternative and simpler chemical scaffold to the parent series of diarylpyrimidines. In addition, using molecular dynamics and free energy perturbation simulations, we elucidate the effect of the second nitrogen of the parent diarylpyrimidines, which is revealed as a stabilizer of a water network in the binding site. The discovery of 2,6-diaryl-2-acetamidopyridines represents a step forward in the search of chemically simple, potent, and selective antagonists for the hA3AR, and exemplifies the benefits of a joint theoretical-experimental approach to identify novel hA3AR antagonists through succinct and efficient synthetic methodologies.


Assuntos
Acetamidas/química , Acetamidas/farmacologia , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Receptor A3 de Adenosina/metabolismo , Animais , Sítios de Ligação , Células CHO , Desenho Assistido por Computador , Cricetulus , Desenho de Fármacos , Humanos , Simulação de Acoplamento Molecular , Nitrogênio/química , Nitrogênio/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Receptor A3 de Adenosina/química , Relação Estrutura-Atividade
9.
J Med Chem ; 60(17): 7459-7475, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28799755

RESUMO

A3 adenosine receptor (AR) ligands including A3 AR agonist, N6-(3-iodobenzyl)adenosine-5'-N-methyluronamide (1a, IB-MECA) were examined for adiponectin production in human bone marrow mesenchymal stem cells (hBM-MSCs). In this model, 1a significantly increased adiponectin production, which is associated with improved insulin sensitivity. However, A3 AR antagonists also promoted adiponectin production in hBM-MSCs, indicating that the A3 AR pathway may not be directly involved in the adiponectin promoting activity. In a target deconvolution study, their adiponectin-promoting activity was significantly correlated to their binding activity to both peroxisome proliferator activated receptor (PPAR) γ and PPARδ. They functioned as both PPARγ partial agonists and PPARδ antagonists. In the diabetic mouse model, 1a and its structural analogues A3 AR antagonists significantly decreased the serum levels of glucose and triglyceride, supporting their antidiabetic potential. These findings indicate that the polypharmacophore of these compounds may provide therapeutic insight into their multipotent efficacy against various human diseases.


Assuntos
Agonistas do Receptor A3 de Adenosina/uso terapêutico , Adenosina/análogos & derivados , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , PPAR delta/antagonistas & inibidores , PPAR gama/agonistas , Adenosina/química , Adenosina/farmacologia , Adenosina/uso terapêutico , Agonistas do Receptor A3 de Adenosina/química , Agonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/uso terapêutico , Adiponectina/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Experimental/metabolismo , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Resistência à Insulina , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR delta/metabolismo , PPAR gama/metabolismo , Polifarmacologia , Receptor A3 de Adenosina/metabolismo
10.
J Med Chem ; 60(17): 7555-7568, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28806076

RESUMO

We expanded on a series of pyrido[2,1-f]purine-2,4-dione derivatives as human adenosine A3 receptor (hA3R) antagonists to determine their kinetic profiles and affinities. Many compounds showed high affinities and a diverse range of kinetic profiles. We found hA3R antagonists with very short residence time (RT) at the receptor (2.2 min for 5) and much longer RTs (e.g., 376 min for 27 or 391 min for 31). Two representative antagonists (5 and 27) were tested in [35S]GTPγS binding assays, and their RTs appeared correlated to their (in)surmountable antagonism. From a kon-koff-KD kinetic map, we divided the antagonists into three subgroups, providing a possible direction for the further development of hA3R antagonists. Additionally, we performed a computational modeling study that sheds light on the crucial receptor interactions, dictating the compounds' binding kinetics. Knowledge of target binding kinetics appears useful for developing and triaging new hA3R antagonists in the early phase of drug discovery.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Purinas/química , Purinas/farmacologia , Receptor A3 de Adenosina/metabolismo , Animais , Células CHO , Cricetulus , Humanos , Cinética , Simulação de Acoplamento Molecular
11.
J Med Chem ; 60(10): 4327-4341, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28447789

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , Analgésicos/química , Analgésicos/farmacologia , Agonistas do Receptor Purinérgico P1/química , Agonistas do Receptor Purinérgico P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/química , Antagonistas de Receptores Purinérgicos P1/farmacologia , Dor Aguda/tratamento farmacológico , Adenosina/uso terapêutico , Agonistas do Receptor A1 de Adenosina/química , Agonistas do Receptor A1 de Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina/uso terapêutico , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/uso terapêutico , Analgésicos/uso terapêutico , Animais , Humanos , Camundongos , Modelos Moleculares , Agonistas do Receptor Purinérgico P1/uso terapêutico , Antagonistas de Receptores Purinérgicos P1/uso terapêutico , Receptor A1 de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Receptores Purinérgicos P1/metabolismo
12.
Int J Mol Sci ; 18(4)2017 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-28338619

RESUMO

Rheumatoid arthritis (RA), ankylosing spondylitis (AS) and psoriatic arthritis (PsA) are chronic inflammatory rheumatic diseases that affect joints, causing debilitating pain and disability. Adenosine receptors (ARs) play a key role in the mechanism of inflammation, and the activation of A2A and A3AR subtypes is often associated with a reduction of the inflammatory status. The aim of this study was to investigate the involvement of ARs in patients suffering from early-RA (ERA), RA, AS and PsA. Messenger RNA (mRNA) analysis and saturation binding experiments indicated an upregulation of A2A and A3ARs in lymphocytes obtained from patients when compared with healthy subjects. A2A and A3AR agonists inhibited nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activation and reduced inflammatory cytokines release, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß and IL-6. Moreover, A2A and A3AR activation mediated a reduction of metalloproteinases (MMP)-1 and MMP-3. The effect of the agonists was abrogated by selective antagonists demonstrating the direct involvement of these receptor subtypes. Taken together, these data confirmed the involvement of ARs in chronic autoimmune rheumatic diseases highlighting the possibility to exploit A2A and A3ARs as therapeutic targets, with the aim to limit the inflammatory responses usually associated with RA, AS and PsA.


Assuntos
Artrite Psoriásica/patologia , Artrite Reumatoide/patologia , Receptor A2A de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Espondilite Anquilosante/patologia , Adenosina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Agonistas do Receptor A2 de Adenosina/química , Agonistas do Receptor A2 de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina/química , Antagonistas do Receptor A2 de Adenosina/metabolismo , Agonistas do Receptor A3 de Adenosina/química , Agonistas do Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/metabolismo , Artrite Psoriásica/metabolismo , Artrite Reumatoide/metabolismo , Estudos de Casos e Controles , Citocinas/metabolismo , Feminino , Humanos , Cinética , Linfócitos/metabolismo , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Fenetilaminas/química , Fenetilaminas/metabolismo , Pirazóis/química , Pirazóis/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/genética , Receptor A3 de Adenosina/genética , Espondilite Anquilosante/metabolismo
13.
Eur J Med Chem ; 125: 611-628, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27721147

RESUMO

The imidazo[1,2-a]pyrazine ring system has been chosen as a new decorable core skeleton for the design of novel adenosine receptor (AR) antagonists targeting either the human (h) A3 or the hA2A receptor subtype. The N8-(hetero)arylcarboxyamido substituted compounds 4-14 and 21-30, bearing a 6-phenyl moiety or not, respectively, show good hA3 receptor affinity and selectivity versus the other ARs. In contrast, the 8-amino-6-(hetero)aryl substituted derivatives designed for targeting the hA2A receptor subtype (compounds 31-38) and also the 6-phenyl analogues 18-20 do not bind the hA2A AR, or show hA1 or balanced hA1/hA2A AR affinity in the micromolar range. Molecular docking of the new hA3 antagonists was carried out to depict their hypothetical binding mode to our refined model of the hA3 receptor. Some derivatives were evaluated for their fluorescent potentiality and showed some fluorescent emission properties. One of the most active hA3 antagonists herein reported, i.e. the 2,6-diphenyl-8-(3-pyridoylamino)imidazo[1,2-a]pyrazine 29, tested in a rat model of cerebral ischemia, delayed the occurrence of anoxic depolarization caused by oxygen and glucose deprivation in the hippocampus and allowed disrupted synaptic activity to recover.


Assuntos
Aminas/síntese química , Desenho de Fármacos , Imidazóis/síntese química , Antagonistas de Receptores Purinérgicos P1/síntese química , Pirazinas/síntese química , Receptor A2A de Adenosina/química , Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Aminas/química , Aminas/farmacologia , Animais , Isquemia Encefálica/tratamento farmacológico , Pareamento Cromossômico/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Humanos , Imidazóis/química , Imidazóis/farmacologia , Simulação de Acoplamento Molecular , Antagonistas de Receptores Purinérgicos P1/química , Antagonistas de Receptores Purinérgicos P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/uso terapêutico , Pirazinas/química , Pirazinas/farmacologia , Ratos
14.
Chirality ; 28(5): 434-40, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27095007

RESUMO

The chiral separation of enantiomeric couples of three potential A3 adenosine receptor antagonists: (R/S)-N-(6-(1-phenylethoxy)-2-(propylthio)pyrimidin-4-yl)acetamide (), (R/S)-N-(2-(1-phenylethylthio)-6-propoxypyrimidin-4-yl)acetamide (), and (R/S)-N-(2-(benzylthio)-6-sec-butoxypyrimidin-4-yl)acetamide () was achieved by high-performance liquid chromatography (HPLC). Three types of chiroptical spectroscopies, namely, optical rotatory dispersion (ORD), electronic circular dichroism (ECD), and vibrational circular dichroism (VCD), were applied to enantiomeric compounds. Through comparison with Density Functional Theory (DFT) calculations, encompassing extensive conformational analysis, full assignment of the absolute configuration (AC) for the three sets of compounds was obtained. Chirality 28:434-440, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Cromatografia Líquida de Alta Pressão/métodos , Dicroísmo Circular , Modelos Moleculares , Estrutura Molecular , Dispersão Óptica Rotatória , Pirimidinas/química , Estereoisomerismo
15.
Eur J Med Chem ; 108: 117-133, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26638043

RESUMO

In previous research, we identified some 7-oxo- and 7-acylamino-substituted pyrazolo[4,3-d]pyrimidine derivatives as potent and selective human (h) A3 adenosine receptor (AR) antagonists. Herein we report on the structural refinement of this class of antagonists aimed at achieving improved receptor-ligand recognition. Hence, substituents with different steric bulk, flexibility and lipophilicity (Me, Ar, heteroaryl, CH2Ph) were introduced at the 5- and 2-positions of the bicyclic scaffold of both the 7-oxo and 7-amino derivatives, and acyl residues were appended on the 7-amino group of the latter. All the 2-phenylpyrazolo[4,3-d]pyrimidin-7-amines and 7-acylamines bearing a 4-methoxyphenyl- or a 2-thienyl group at the 5-position showed high hA3 affinity and selectivity. In particular, the 2-phenyl-5-(2-thienyl)-pyrazolo[4,3-d]pyrimidin-7-(4-methoxybenzoyl)amine 25 (Ki = 0.027 nM) is one of the most potent and selective hA3 antagonists reported so far. By using an in silico receptor-driven approach the obtained binding data were rationalized and the molecular bases of the observed hA3 AR affinities were critically described.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Pirazóis/química , Pirazóis/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A3 de Adenosina/síntese química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Pirazóis/síntese química , Pirimidinas/síntese química , Relação Estrutura-Atividade
16.
Mol Divers ; 20(1): 55-76, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26205409

RESUMO

Adenosine regulates tissue function by activating four G-protein-coupled adenosine receptors (ARs). Selective agonists and antagonists for A3 ARs have been investigated for the treatment of a variety of immune disorders, cancer, brain, and heart ischemic conditions. We herein present a QSAR study based on a Topological sub-structural molecular design (TOPS-MODE) approach, intended to predict the A3 ARs of a diverse dataset of 124 (94 training set/ 30 prediction set) adenosine derivatives. The final model showed good fit and predictive capability, displaying 85.1 % of the experimental variance. The TOPS-MODE approach afforded a better understanding and interpretation of the developed model based on the useful information extracted from the analysis of the contribution of different molecular fragments to the affinity.


Assuntos
Agonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/química , Biologia Computacional/métodos , Receptor A3 de Adenosina/metabolismo , Agonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Descoberta de Drogas , Humanos , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Receptor A3 de Adenosina/química
17.
J Med Chem ; 58(24): 9578-90, 2015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26592528

RESUMO

Fragment-based lead discovery (FBLD) holds great promise for drug discovery, but applications to G protein-coupled receptors (GPCRs) have been limited by a lack of sensitive screening techniques and scarce structural information. If virtual screening against homology models of GPCRs could be used to identify fragment ligands, FBLD could be extended to numerous important drug targets and contribute to efficient lead generation. Access to models of multiple receptors may further enable the discovery of fragments that bind specifically to the desired target. To investigate these questions, we used molecular docking to screen >500 000 fragments against homology models of the A3 and A1 adenosine receptors (ARs) with the goal to discover A3AR-selective ligands. Twenty-one fragments with predicted A3AR-specific binding were evaluated in live-cell fluorescence-based assays; of eight verified ligands, six displayed A3/A1 selectivity, and three of these had high affinities ranging from 0.1 to 1.3 µM. Subsequently, structure-guided fragment-to-lead optimization led to the identification of a >100-fold-selective antagonist with nanomolar affinity from commercial libraries. These results highlight that molecular docking screening can guide fragment-based discovery of selective ligands even if the structures of both the target and antitarget receptors are unknown. The same approach can be readily extended to a large number of pharmaceutically important targets.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Pirimidinonas/química , Receptor A3 de Adenosina/química , Tiazóis/química , Tiofenos/química , Antagonistas do Receptor A1 de Adenosina/química , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Animais , Ligação Competitiva , Células CHO , Simulação por Computador , Cricetulus , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Simulação de Acoplamento Molecular , Pirimidinonas/farmacologia , Receptor A1 de Adenosina/química , Receptor A1 de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Homologia de Sequência de Aminoácidos , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade , Tiazóis/farmacologia , Tiofenos/farmacologia
18.
Future Med Chem ; 7(11): 1373-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26230877

RESUMO

BACKGROUND: A3AR antagonists are promising drug candidates as neuroprotective agents as well as for the treatment of inflammation or glaucoma. The most widely known A3AR antagonists are derived from polyheteroaromatic scaffolds, which usually show poor pharmacokinetic properties. Accordingly, the identification of structurally simple A3AR antagonists by the exploration of novel diversity spaces is a challenging goal. RESULTS: A convergent and efficient Ugi-based multicomponent approach enabled the discovery of pyrazin-2(1H)-ones as a novel class of A3AR antagonists. A combined experimental/computational strategy accelerated the establishment of the most salient features of the structure-activity and structure-selectivity relationships in this series. CONCLUSION: The optimization process provided pyrazin-2(1H)-ones with improved affinity and a plausible hypothesis regarding their binding modes was proposed.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Pirazinas/química , Pirazinas/farmacologia , Receptor A3 de Adenosina/metabolismo , Descoberta de Drogas , Humanos , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade
19.
Eur J Med Chem ; 96: 105-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25874336

RESUMO

A new series of 5-methyl-thiazolo[5,4-d]pyrimidine-7-ones bearing different substituents at position 2 (aryl, heteroaryl and arylamino groups) was synthesized and evaluated in radioligand binding assays to determine their affinities at the human (h) A1, A2A, and A3 adenosine receptors (ARs). Efficacy at the hA(2B) and antagonism of selected ligands at the hA3 were also assessed through cAMP experiments. Some of the new derivatives exhibited good to high hA3AR affinity and selectivity versus all the other AR subtypes. Compound 2-(4-chlorophenyl)-5-methyl-thiazolo[5,4-d]pyrimidine-7-one 4 was found to be the most potent and selective ligand of the series (K(I) hA3 = 18 nM). Molecular docking studies of the reported derivatives were carried out to depict their hypothetical binding mode in our hA3 receptor model.


Assuntos
Antagonistas do Receptor A3 de Adenosina/síntese química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Desenho de Fármacos , Pirimidinas/química , Pirimidinas/farmacologia , Receptor A3 de Adenosina/metabolismo , Tiazóis/química , Tiazóis/farmacologia , Antagonistas do Receptor A3 de Adenosina/química , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Pirimidinas/síntese química , Relação Estrutura-Atividade , Tiazóis/síntese química
20.
Eur J Med Chem ; 92: 784-98, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25633494

RESUMO

An efficient synthetic procedure was adopted to synthesize a series of new molecules containing the pyrazolo[3,4-d]pyrimidine (PP) scaffold, which have been evaluated as promising human adenosine receptor (AR) antagonists. The effect of substitutions at the N(2), C(4) and C(6) positions of PPs on the affinity and selectivity towards the adenosine receptors were explored. Most of the pyrazolo[3,4-d]pyrimidine-4-carboxylates displayed from moderate to good affinity at the human A3AR (hA3AR), as indicated by the low micromolar range of Ki values (Ki hA3AR = 0.7-34 µM). In particular, compounds 60 and 62 displayed good affinity at the hA3AR (60, Ki hA3AR = 2.2 µM and 62, Ki hA3AR = 2.9 µM) and selectivity towards the other AR subtypes (60, >46-fold selective and 62, >34-fold selective, respectively). In view of these results, these novel PP analogues were docked both in the crystallographic structure of the hA2AAR and in a homology model of the hA3AR in order to support the structure activity relationship (SAR) analysis. These preliminary results demonstrated that pyrazolo[3,4-d]pyrimidine can be considered a promising scaffold to obtain new molecules with potent hA3AR antagonist activity.


Assuntos
Antagonistas do Receptor A3 de Adenosina/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Receptor A3 de Adenosina/metabolismo , Antagonistas do Receptor A3 de Adenosina/síntese química , Antagonistas do Receptor A3 de Adenosina/química , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Pirazóis/síntese química , Pirazóis/química , Pirimidinas/síntese química , Pirimidinas/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA