Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cell ; 168(5): 867-877.e13, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28235198

RESUMO

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.


Assuntos
Receptor A1 de Adenosina/química , Agonistas do Receptor A1 de Adenosina/química , Antagonistas do Receptor A1 de Adenosina/química , Sítio Alostérico , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/química
2.
Nature ; 597(7877): 571-576, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34497422

RESUMO

The adenosine A1 receptor (A1R) is a promising therapeutic target for non-opioid analgesic agents to treat neuropathic pain1,2. However, development of analgesic orthosteric A1R agonists has failed because of a lack of sufficient on-target selectivity as well as off-tissue adverse effects3. Here we show that [2-amino-4-(3,5-bis(trifluoromethyl)phenyl)thiophen-3-yl)(4-chlorophenyl)methanone] (MIPS521), a positive allosteric modulator of the A1R, exhibits analgesic efficacy in rats in vivo through modulation of the increased levels of endogenous adenosine that occur in the spinal cord of rats with neuropathic pain. We also report the structure of the A1R co-bound to adenosine, MIPS521 and a Gi2 heterotrimer, revealing an extrahelical lipid-detergent-facing allosteric binding pocket that involves transmembrane helixes 1, 6 and 7. Molecular dynamics simulations and ligand kinetic binding experiments support a mechanism whereby MIPS521 stabilizes the adenosine-receptor-G protein complex. This study provides proof of concept for structure-based allosteric drug design of non-opioid analgesic agents that are specific to disease contexts.


Assuntos
Analgesia , Receptor A1 de Adenosina/metabolismo , Adenosina/química , Adenosina/metabolismo , Regulação Alostérica/efeitos dos fármacos , Analgesia/métodos , Animais , Sítios de Ligação , Modelos Animais de Doenças , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Hiperalgesia/tratamento farmacológico , Lipídeos , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/química , Transdução de Sinais/efeitos dos fármacos
3.
Chembiochem ; : e202400242, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38777792

RESUMO

Accumulating evidence suggests that G protein-coupled receptors (GPCRs) can exist and function in homodimer and heterodimer forms. The adenosine A1 receptor (A1R) has been shown to form both homodimers and heterodimers, but there is a lack of chemical tools to study these dimeric receptor populations. This work describes the synthesis and pharmacological evaluation of a novel class of bivalent GPCR chemical tools, where each ligand moiety of the bivalent compound contains a sulfonyl fluoride covalent warhead designed to be capable of simultaneously reacting with each A1R of an A1R homodimer. The novel compounds were characterised using radioligand binding assays, including washout assays, and functionally in cAMP assays. The bivalent dicovalent compounds were competitive A1R antagonists and showed evidence of covalent binding and simultaneous binding across an A1R homodimer. Greater selectivity for A1R over the adenosine A3 receptor was observed for bivalent dicovalent over the equivalent monovalent compounds, indicating subtype selectivity can be achieved with dual occupation by a bivalent dicovalent ligand.

4.
Nature ; 558(7711): 559-563, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29925945

RESUMO

The class A adenosine A1 receptor (A1R) is a G-protein-coupled receptor that preferentially couples to inhibitory Gi/o heterotrimeric G proteins, has been implicated in numerous diseases, yet remains poorly targeted. Here we report the 3.6 Å structure of the human A1R in complex with adenosine and heterotrimeric Gi2 protein determined by Volta phase plate cryo-electron microscopy. Compared to inactive A1R, there is contraction at the extracellular surface in the orthosteric binding site mediated via movement of transmembrane domains 1 and 2. At the intracellular surface, the G protein engages the A1R primarily via amino acids in the C terminus of the Gαi α5-helix, concomitant with a 10.5 Å outward movement of the A1R transmembrane domain 6. Comparison with the agonist-bound ß2 adrenergic receptor-Gs-protein complex reveals distinct orientations for each G-protein subtype upon engagement with its receptor. This active A1R structure provides molecular insights into receptor and G-protein selectivity.


Assuntos
Adenosina/química , Adenosina/metabolismo , Microscopia Crioeletrônica , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/ultraestrutura , Receptor A1 de Adenosina/química , Receptor A1 de Adenosina/ultraestrutura , Sítios de Ligação , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Modelos Moleculares , Receptor A1 de Adenosina/metabolismo , Rotação , Especificidade por Substrato
5.
Clin Sci (Lond) ; 136(23): 1731-1758, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36459456

RESUMO

Cardiovascular disease (CVD) remains the leading cause of death worldwide. An ongoing challenge remains the development of novel pharmacotherapies to treat CVD, particularly atherosclerosis. Effective mechanism-informed development and translation of new drugs requires a deep understanding of the known and currently unknown biological mechanisms underpinning atherosclerosis, accompanied by optimization of traditional drug discovery approaches. Current animal models do not precisely recapitulate the pathobiology underpinning human CVD. Accordingly, a fundamental limitation in early-stage drug discovery has been the lack of consensus regarding an appropriate experimental in vivo model that can mimic human atherosclerosis. However, when coupled with a clear understanding of the specific advantages and limitations of the model employed, preclinical animal models remain a crucial component for evaluating pharmacological interventions. Within this perspective, we will provide an overview of the mechanisms and modalities of atherosclerotic drugs, including those in the preclinical and early clinical development stage. Additionally, we highlight recent preclinical models that have improved our understanding of atherosclerosis and associated clinical consequences and propose model adaptations to facilitate the development of new and effective treatments.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Animais , Humanos , Aterosclerose/tratamento farmacológico , Descoberta de Drogas , Modelos Animais
6.
Purinergic Signal ; 18(3): 359-381, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35870032

RESUMO

Alzheimer's disease (AD) is the most common dementia in the elderly and its increasing prevalence presents treatment challenges. Despite a better understanding of the disease, the current mainstay of treatment cannot modify pathogenesis or effectively address the associated cognitive and memory deficits. Emerging evidence suggests adenosine G protein-coupled receptors (GPCRs) are promising therapeutic targets for Alzheimer's disease. The adenosine A1 and A2A receptors are expressed in the human brain and have a proposed involvement in the pathogenesis of dementia. Targeting these receptors preclinically can mitigate pathogenic ß-amyloid and tau neurotoxicity whilst improving cognition and memory. In this review, we provide an accessible summary of the literature on Alzheimer's disease and the therapeutic potential of A1 and A2A receptors. Although there are no available medicines targeting these receptors approved for treating dementia, we provide insights into some novel strategies, including allosterism and the targeting of oligomers, which may increase drug discovery success and enhance the therapeutic response.


Assuntos
Doença de Alzheimer , Adenosina/metabolismo , Idoso , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Humanos , Receptores Purinérgicos P1/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(10): E2419-E2428, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29453275

RESUMO

Subtype-selective antagonists for muscarinic acetylcholine receptors (mAChRs) have long been elusive, owing to the highly conserved orthosteric binding site. However, allosteric sites of these receptors are less conserved, motivating the search for allosteric ligands that modulate agonists or antagonists to confer subtype selectivity. Accordingly, a 4.6 million-molecule library was docked against the structure of the prototypical M2 mAChR, seeking molecules that specifically stabilized antagonist binding. This led us to identify a positive allosteric modulator (PAM) that potentiated the antagonist N-methyl scopolamine (NMS). Structure-based optimization led to compound '628, which enhanced binding of NMS, and the drug scopolamine itself, with a cooperativity factor (α) of 5.5 and a KB of 1.1 µM, while sparing the endogenous agonist acetylcholine. NMR spectral changes determined for methionine residues reflected changes in the allosteric network. Moreover, '628 slowed the dissociation rate of NMS from the M2 mAChR by 50-fold, an effect not observed at the other four mAChR subtypes. The specific PAM effect of '628 on NMS antagonism was conserved in functional assays, including agonist stimulation of [35S]GTPγS binding and ERK 1/2 phosphorylation. Importantly, the selective allostery between '628 and NMS was retained in membranes from adult rat hypothalamus and in neonatal rat cardiomyocytes, supporting the physiological relevance of this PAM/antagonist approach. This study supports the feasibility of discovering PAMs that confer subtype selectivity to antagonists; molecules like '628 can convert an armamentarium of potent but nonselective GPCR antagonist drugs into subtype-selective reagents, thus reducing their off-target effects.


Assuntos
Agonistas Muscarínicos/química , Receptor Muscarínico M2/química , Regulação Alostérica , Sítio Alostérico , Animais , Humanos , Cinética , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Simulação de Acoplamento Molecular , Agonistas Muscarínicos/metabolismo , Fosforilação , Ligação Proteica , Ratos , Receptor Muscarínico M2/metabolismo
8.
Nat Chem Biol ; 13(9): 929-937, 2017 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-28820879

RESUMO

G-protein-coupled receptors (GPCRs) are one of the most tractable classes of drug targets. These dynamic proteins can adopt multiple active states that are linked to distinct functional outcomes. Such states can be differentially stabilized by ligands interacting with the endogenous agonist-binding orthosteric site and/or by ligands acting via spatially distinct allosteric sites, leading to the phenomena of 'biased agonism' or 'biased modulation'. These paradigms are having a major impact on modern drug discovery, but it is becoming increasingly apparent that 'kinetic context', at the level of both ligand-receptor and receptor-signal pathway kinetics, can have a profound impact on the observation and quantification of these phenomena. The concept of kinetic context thus represents an important new consideration that should be routinely incorporated into contemporary chemical biology and drug discovery studies of GPCR bias and allostery.


Assuntos
Modelos Moleculares , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Regulação Alostérica , Descoberta de Drogas , Cinética , Ligantes , Transdução de Sinais
9.
Mol Pharmacol ; 93(5): 504-514, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29514854

RESUMO

Numerous positive and negative allosteric modulators (PAMs and NAMs) of class C G protein-coupled receptors (GPCRs) have been developed as valuable preclinical pharmacologic tools and therapeutic agents. Although many class C GPCR allosteric modulators have undergone subtype selectivity screening, most assay paradigms have failed to perform rigorous pharmacologic assessment. Using mGlu5 as a representative class C GPCR, we tested the hypothesis that allosteric modulator selectivity was based on cooperativity rather than affinity. Specifically, we aimed to identify ligands that bound to mGlu5 but exhibited neutral cooperativity with mGlu5 agonists. We additionally evaluated the potential for these ligands to exhibit biased pharmacology. Radioligand binding, intracellular calcium (iCa2+) mobilization, and inositol monophosphate (IP1) accumulation assays were undertaken in human embryonic kidney cells expressing low levels of rat mGlu5 (HEK293A-mGlu5-low) for diverse allosteric chemotypes. Numerous "non-mGlu5" class C GPCR allosteric modulators incompletely displaced allosteric mGlu5 radioligand [3H]methoxy-PEPy binding, consistent with a negative allosteric interaction. Affinity estimates for CPCCOEt (mGlu1 ligand), PHCCC (mGlu4 ligand), GS39783 (GABAB ligand), AZ12216052 (mGlu8 ligand), and CGP7930 (GABAB ligand) at mGlu5 were within 10-fold of their target receptor. Most class C GPCR allosteric modulators had neutral cooperativity with both orthosteric and allosteric mGlu5 agonists in functional assays; however, NPS2143 (calcium-sensing receptor (CaSR) NAM), cinacalcet (CaSR PAM), CGP7930, and AZ12216052 were partial mGlu5 agonists for IP1 accumulation, but not iCa2+ mobilization. By using mGlu5 as a model class C GPCR, we find that for many class C GPCR allosteric modulators, subtype selectivity is driven by cooperativity and misinterpreted owing to unappreciated bias.


Assuntos
Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulação Alostérica , Animais , Cálcio/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Feminino , Células HEK293 , Humanos , Fosfatos de Inositol/metabolismo , Ligantes , Camundongos , Neurônios/metabolismo , Ensaio Radioligante , Ratos , Trítio
10.
Clin Sci (Lond) ; 132(21): 2323-2338, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30389826

RESUMO

Metabotropic glutamate receptors belong to class C G-protein-coupled receptors and consist of eight subtypes that are ubiquitously expressed throughout the central nervous system. In recent years, the metabotropic glutamate receptor subtype 5 (mGlu5) has emerged as a promising target for a broad range of psychiatric and neurological disorders. Drug discovery programs targetting mGlu5 are primarily focused on development of allosteric modulators that interact with sites distinct from the endogenous agonist glutamate. Significant efforts have seen mGlu5 allosteric modulators progress into clinical trials; however, recent failures due to lack of efficacy or adverse effects indicate a need for a better understanding of the functional consequences of mGlu5 allosteric modulation. Biased agonism is an interrelated phenomenon to allosterism, describing how different ligands acting through the same receptor can differentially influence signaling to distinct transducers and pathways. Emerging evidence demonstrates that allosteric modulators can induce biased pharmacology at the level of intrinsic agonism as well as through differential modulation of orthosteric agonist-signaling pathways. Here, we present key considerations in the discovery and development of mGlu5 allosteric modulators and the opportunities and pitfalls offered by biased agonism and modulation.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Receptor de Glutamato Metabotrópico 5/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Sítios de Ligação , Sistema Nervoso Central/metabolismo , Fármacos do Sistema Nervoso Central/química , Fármacos do Sistema Nervoso Central/metabolismo , Agonistas de Aminoácidos Excitatórios/química , Agonistas de Aminoácidos Excitatórios/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Ligantes , Ligação Proteica , Conformação Proteica , Receptor de Glutamato Metabotrópico 5/química , Receptor de Glutamato Metabotrópico 5/metabolismo , Relação Estrutura-Atividade
12.
Proc Natl Acad Sci U S A ; 111(12): 4614-9, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24619092

RESUMO

The concepts of allosteric modulation and biased agonism are revolutionizing modern approaches to drug discovery, particularly in the field of G protein-coupled receptors (GPCRs). Both phenomena exploit topographically distinct binding sites to promote unique GPCR conformations that can lead to different patterns of cellular responsiveness. The adenosine A1 GPCR (A1AR) is a major therapeutic target for cardioprotection, but current agents acting on the receptor are clinically limited for this indication because of on-target bradycardia as a serious adverse effect. In the current study, we have rationally designed a novel A1AR ligand (VCP746)--a hybrid molecule comprising adenosine linked to a positive allosteric modulator--specifically to engender biased signaling at the A1AR. We validate that the interaction of VCP746 with the A1AR is consistent with a bitopic mode of receptor engagement (i.e., concomitant association with orthosteric and allosteric sites) and that the compound displays biased agonism relative to prototypical A1AR ligands. Importantly, we also show that the unique pharmacology of VCP746 is (patho)physiologically relevant, because the compound protects against ischemic insult in native A1AR-expressing cardiomyoblasts and cardiomyocytes but does not affect rat atrial heart rate. Thus, this study provides proof of concept that bitopic ligands can be designed as biased agonists to promote on-target efficacy without on-target side effects.


Assuntos
Adenosina/análogos & derivados , Desenho de Fármacos , Agonistas do Receptor Purinérgico P1/química , Tiofenos/química , Adenosina/efeitos adversos , Adenosina/química , Sítio Alostérico , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Ligantes , Agonistas do Receptor Purinérgico P1/efeitos adversos , Ratos , Tiofenos/efeitos adversos
13.
Mol Pharmacol ; 90(1): 12-22, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27136943

RESUMO

Biased agonism at G protein-coupled receptors (GPCRs) has significant implications for current drug discovery, but molecular determinants that govern ligand bias remain largely unknown. The adenosine A3 GPCR (A3AR) is a potential therapeutic target for various conditions, including cancer, inflammation, and ischemia, but for which biased agonism remains largely unexplored. We now report the generation of bias "fingerprints" for prototypical ribose containing A3AR agonists and rigidified (N)-methanocarba 5'-N-methyluronamide nucleoside derivatives with regard to their ability to mediate different signaling pathways. Relative to the reference prototypical agonist IB-MECA, (N)-methanocarba 5'-N-methyluronamide nucleoside derivatives with significant N(6) or C2 modifications, including elongated aryl-ethynyl groups, exhibited biased agonism. Significant positive correlation was observed between the C2 substituent length (in Å) and bias toward cell survival. Molecular modeling suggests that extended C2 substituents on (N)-methanocarba 5'-N-methyluronamide nucleosides promote a progressive outward shift of the A3AR transmembrane domain 2, which may contribute to the subset of A3AR conformations stabilized on biased agonist binding.


Assuntos
Agonistas do Receptor A3 de Adenosina/química , Agonistas do Receptor A3 de Adenosina/farmacologia , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Ligantes , Simulação de Acoplamento Molecular , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
14.
Mol Pharmacol ; 90(6): 715-725, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27683013

RESUMO

Allosteric modulation of adenosine A1 receptors (A1ARs) offers a novel therapeutic approach for the treatment of numerous central and peripheral disorders; however, despite decades of research, there is a relative paucity of structural information regarding the A1AR allosteric site and mechanisms governing cooperativity with orthosteric ligands. We combined alanine-scanning mutagenesis of the A1AR second extracellular loop (ECL2) with radioligand binding and functional interaction assays to quantify effects on allosteric ligand affinity, cooperativity, and efficacy. Docking and molecular dynamics (MD) simulations were performed using an A1AR homology model based on an agonist-bound A2AAR structure. Substitution of E172ECL2 for alanine reduced the affinity of the allosteric modulators PD81723 and VCP171 for the unoccupied A1AR. Residues involved in cooperativity with the orthosteric agonist NECA were different in PD81723 and VCP171; positive cooperativity between PD81723 and NECA was reduced on alanine substitution of a number of ECL2 residues, including E170ECL2 and K173ECL2, whereas mutation of W146ECL2 and W156ECL2 decreased VCP171 cooperativity with NECA. Molecular modeling localized a likely allosteric pocket for both modulators to an extracellular vestibule that overlaps with a region used by orthosteric ligands as they transit into the canonical A1AR orthosteric site. MD simulations confirmed a key interaction between E172ECL2 and both modulators. Bound PD81723 is flanked by another residue, E170ECL2, which forms hydrogen bonds with adjacent K168ECL2 and K173ECL2. Collectively, our data suggest E172ECL2 is a key allosteric ligand-binding determinant, whereas hydrogen-bonding networks within the extracellular vestibule may facilitate the transmission of cooperativity between orthosteric and allosteric sites.


Assuntos
Sítio Alostérico , Receptor A1 de Adenosina/química , Receptor A1 de Adenosina/metabolismo , Transdução de Sinais , Adenosina/farmacologia , Alanina/genética , Regulação Alostérica/efeitos dos fármacos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Ligantes , Simulação de Dinâmica Molecular , Mutação/genética , Estrutura Secundária de Proteína , Transdução de Sinais/efeitos dos fármacos , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
15.
Mol Pharmacol ; 90(6): 703-714, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27683014

RESUMO

The adenosine A1 G protein-coupled receptor (A1AR) is an important therapeutic target implicated in a wide range of cardiovascular and neuronal disorders. Although it is well established that the A1AR orthosteric site is located within the receptor's transmembrane (TM) bundle, prior studies have implicated extracellular loop 2 (ECL2) as having a significant role in contributing to orthosteric ligand affinity and signaling for various G protein-coupled receptors (GPCRs). We thus performed extensive alanine scanning mutagenesis of A1AR-ECL2 to explore the role of this domain on A1AR orthosteric ligand pharmacology. Using quantitative analytical approaches and molecular modeling, we identified ECL2 residues that interact either directly or indirectly with orthosteric agonists and antagonists. Discrete mutations proximal to a conserved ECL2-TM3 disulfide bond selectively affected orthosteric ligand affinity, whereas a cluster of five residues near the TM4-ECL2 juncture influenced orthosteric agonist efficacy. A combination of ligand docking, molecular dynamics simulations, and mutagenesis results suggested that the orthosteric agonist 5'-N-ethylcarboxamidoadenosine binds transiently to an extracellular vestibule formed by ECL2 and the top of TM5 and TM7, prior to entry into the canonical TM bundle orthosteric site. Collectively, this study highlights a key role for ECL2 in A1AR orthosteric ligand binding and receptor activation.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Receptor A1 de Adenosina/química , Receptor A1 de Adenosina/metabolismo , Adenosina/farmacologia , Alanina/genética , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Ligantes , Simulação de Dinâmica Molecular , Proteínas Mutantes/metabolismo , Estrutura Secundária de Proteína , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Xantinas/farmacologia
16.
J Pharmacol Exp Ther ; 357(1): 36-44, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26791603

RESUMO

Aberrant ligand-independent G protein-coupled receptor constitutive activity has been implicated in the pathophysiology of a number of cancers. The adenosine A2B receptor (A2BAR) is dynamically upregulated under pathologic conditions associated with a hypoxic microenvironment, including solid tumors. This, in turn, may amplify ligand-independent A2BAR signal transduction. The contribution of A2BAR constitutive activity to disease progression is currently unknown yet of fundamental importance, as the preferred therapeutic modality for drugs designed to reduce A2BAR constitutive activity would be inverse agonism as opposed to neutral antagonism. The current study investigated A2BAR constitutive activity in a heterologous expression system and a native 22Rv1 human prostate cancer cell line exposed to hypoxic conditions (2% O2). The A2BAR inverse agonists, ZM241385 [4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol] or PSB-603 (8-(4-(4-(4-chlorophenyl)piperazide-1-sulfonyl)phenyl)-1-propylxanthine), mediated a concentration-dependent decrease in baseline cAMP levels in both cellular systems. Proliferation of multiple prostate cancer cell lines was also attenuated in the presence of PSB-603. Importantly, both the decrease in baseline cAMP accumulation and the reduction of proliferation were not influenced by the addition of adenosine deaminase, demonstrating that these effects are not dependent on stimulation of A2BARs by the endogenous agonist adenosine. Our study is the first to reveal that wild-type human A2BARs have high constitutive activity in both model and native cells. Furthermore, our findings demonstrate that this ligand-independent A2BAR constitutive activity is sufficient to promote prostate cancer cell proliferation in vitro. More broadly, A2BAR constitutive activity may have wider, currently unappreciated implications in pathologic conditions associated with a hypoxic microenvironment.


Assuntos
Neoplasias da Próstata/patologia , Receptor A2B de Adenosina/efeitos dos fármacos , Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Adenosina Desaminase/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Desenho de Fármacos , Humanos , Fosfatos de Inositol/metabolismo , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Triazinas/farmacologia , Triazóis/farmacologia , Xantinas/farmacologia
17.
FASEB J ; 29(7): 2859-71, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25837585

RESUMO

At the ß1-adrenoceptor, CGP 12177 potently antagonizes agonist responses at the primary high-affinity catecholamine conformation while also exerting agonist effects of its own through a secondary low-affinity conformation. A recent mutagenesis study identified transmembrane region (TM)4 of the ß1-adrenoceptor as key for this low-affinity conformation. Others suggested that TM4 has a role in ß1-adrenoceptor oligomerization. Here, assessment of the dissociation rate of a fluorescent analog of CGP 12177 [bordifluoropyrromethane-tetramethylrhodamine-(±)CGP 12177 (BODIPY-TMR-CGP)] at the human ß1-adrenoceptor expressed in Chinese hamster ovary cells revealed negative cooperative interactions between 2 distinct ß1-adrenoceptor conformations. The dissociation rate of 3 nM BODIPY-TMR-CGP was 0.09 ± 0.01 min(-1) in the absence of competitor ligands, and this was enhanced 2.2- and 2.1-fold in the presence of 1 µM CGP 12177 and 1 µM propranolol, respectively. These effects on the BODIPY-TMR-CGP dissociation rate were markedly enhanced in ß1-adrenoceptor homodimers constrained by bimolecular fluorescence complementation (9.8- and 9.9-fold for 1 µM CGP 12177 and 1 µM propranolol, respectively) and abolished in ß1-adrenoceptors containing TM4 mutations vital for the second conformation pharmacology. This study suggests that negative cooperativity across a ß1-adrenoceptor homodimer may be responsible for generating the low-affinity pharmacology of the secondary ß1-adrenoceptor conformation.


Assuntos
Antagonistas Adrenérgicos beta/metabolismo , Propanolaminas/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Animais , Sítios de Ligação , Compostos de Boro/metabolismo , Células CHO , Cricetinae , Cricetulus , Ciclopentanos/metabolismo , Humanos , Cinética , Ligantes , Mutagênese Sítio-Dirigida , Conformação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Pirróis/metabolismo , Receptores Adrenérgicos beta 1/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Clin Exp Pharmacol Physiol ; 43(10): 976-82, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27377874

RESUMO

VCP746 is a novel A1 adenosine receptor (A1 AR) biased agonist previously shown to be cytoprotective with no effect on heart rate. The aim of this study was to investigate the potential anti-hypertrophic effect of VCP746 in neonatal rat cardiac myocytes (NCM). NCM hypertrophy was stimulated with interleukin (IL)-1ß (10 ng/mL), tumour necrosis factor (TNF)-α (10 ng/mL) or Ang II (100 nmol/L) and was assessed by (3) H-leucine incorporation assay. VCP746 significantly inhibited IL-1ß-, TNF-α- and Ang II-stimulated NCM hypertrophy as determined by (3) H-leucine incorporation. The anti-hypertrophic effect of VCP746 was also more potent than that of the prototypical A1 AR agonist, N(6) -cyclopentyladenosine (CPA). Further investigation with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell viability assay showed that neither CPA nor VCP746 had any effect on cell viability, confirming that the reduction in (3) H-leucine incorporation mediated by CPA and VCP746 was not due to a reduction in cell viability. IL-1ß, TNF-α and Ang II were also shown to increase the mRNA expression of hypertrophy biomarkers, ANP, ß-MHC and α-SKA in NCM. Treatment with VCP746 at concentrations as low as 1 nmol/L suppressed mRNA expression of ANP, ß-MHC and α-SKA stimulated by IL-1ß, TNF-α or Ang II, demonstrating the broad mechanistic basis of the potent anti-hypertrophic effect of VCP746. This study has shown that the novel A1 AR agonist, VCP746, is able to attenuate cardiac myocyte hypertrophy. As such, VCP746 is potentially useful as a pharmacological agent in attenuating cardiac remodelling, especially in the post-myocardial infarction setting, given its previously established cytoprotective properties.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/análogos & derivados , Sobrevivência Celular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Tiofenos/farmacologia , Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley
19.
Mol Pharmacol ; 88(3): 460-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26104547

RESUMO

In the spinal cord and periphery, adenosine inhibits neuronal activity through activation of the adenosine A1 receptor (A1R), resulting in antinociception and highlighting the potential of therapeutically targeting the receptor in the treatment of neuropathic pain. This study investigated the changes in adenosine tone and A1R signaling, together with the actions of a novel A1R positive allosteric modulator (PAM), VCP171 [(2-amino-4-(3-(trifluoromethyl)phenyl)thiophen-3-yl)(phenyl)methanone], on excitatory and inhibitory neurotransmission at spinal cord superficial dorsal horn synapses in a rat partial nerve-injury model of neuropathic pain. In the absence of A1R agonists, superfusion of the A1R antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX; 1 µM), produced a significantly greater increase in electrically evoked α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-mediated synaptic current (eEPSC) amplitude in both lamina I and II neurons from nerve-injured animals than in controls, suggesting that endogenous adenosine tone is increased in the dorsal horn. Inhibitory GABAergic and glycinergic synaptic currents were also significantly increased by DPCPX in controls but there was no difference after nerve injury. The A1R agonist, N6-cyclopentyladenosine, produced greater inhibition of eEPSC amplitude in lamina II but not lamina I of the spinal cord dorsal horn in nerve-injured versus control animals, suggesting a functional increase in A1R sensitivity in lamina II neurons after nerve injury. The A1R PAM, VCP171, produced a greater inhibition of eEPSC amplitude of nerve-injury versus control animals in both lamina I and lamina II neurons. Enhanced adenosine tone and A1R sensitivity at excitatory synapses in the dorsal horn after nerve injury suggest that new generation PAMs of the A1R can be effective treatments for neuropathic pain.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Potenciais Pós-Sinápticos Excitadores , Neuralgia/tratamento farmacológico , Neurônios Aferentes/efeitos dos fármacos , Tiofenos/farmacologia , Agonistas do Receptor A1 de Adenosina/uso terapêutico , Regulação Alostérica , Animais , Hiperalgesia/tratamento farmacológico , Masculino , Neurônios Aferentes/metabolismo , Neurônios Aferentes/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Tiofenos/uso terapêutico , Xantinas/farmacologia
20.
Br J Pharmacol ; 181(14): 2091-2094, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38798136

RESUMO

LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.


Assuntos
Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA