Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; 170: 105745, 2021 08.
Article in English | MEDLINE | ID: mdl-34182128

ABSTRACT

Polymorphic alleles of the human dopamine D4 receptor gene (DRD4) have been consistently associated with individual differences in personality traits and neuropsychiatric disorders, particularly between the gene encoding dopamine D4.7 receptor variant and attention deficit hyperactivity disorder (ADHD). The α2A adrenoceptor gene has also been associated with ADHD. In fact, drugs targeting the α2A adrenoceptor (α2AR), such as guanfacine, are commonly used in ADHD treatment. In view of the involvement of dopamine D4 receptor (D4R) and α2AR in ADHD and impulsivity, their concurrent localization in cortical pyramidal neurons and the demonstrated ability of D4R to form functional heteromers with other G protein-coupled receptors, in this study we evaluate whether the α2AR forms functional heteromers with D4R and weather these heteromers show different properties depending on the D4R variant involved. Using cortical brain slices from hD4.7R knock-in and wild-type mice, here, we demonstrate that α2AR and D4R heteromerize and constitute a significant functional population of cortical α2AR and D4R. Moreover, in cortical slices from wild-type mice and in cells transfected with α2AR and D4.4R, we detect a negative crosstalk within the heteromer. This negative crosstalk is lost in cortex from hD4.7R knock-in mice and in cells expressing the D4.7R polymorphic variant. We also show a lack of efficacy of D4R ligands to promote G protein activation and signaling only within the α2AR-D4.7R heteromer. Taken together, our results suggest that α2AR-D4R heteromers play a pivotal role in catecholaminergic signaling in the brain cortex and are likely targets for ADHD pharmacotherapy.


Subject(s)
Attention Deficit Disorder with Hyperactivity/metabolism , Cerebral Cortex/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Dopamine D4/metabolism , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/psychology , Cerebral Cortex/drug effects , Dopamine Agonists/pharmacology , Female , HEK293 Cells , Humans , Impulsive Behavior , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymorphism, Genetic , Protein Binding , Receptors, Adrenergic, alpha-2/genetics , Receptors, Dopamine D4/agonists , Receptors, Dopamine D4/genetics , Sheep, Domestic , Signal Transduction
2.
Elife ; 92020 06 09.
Article in English | MEDLINE | ID: mdl-32513388

ABSTRACT

Early Huntington's disease (HD) include over-activation of dopamine D1 receptors (D1R), producing an imbalance in dopaminergic neurotransmission and cell death. To reduce D1R over-activation, we present a strategy based on targeting complexes of D1R and histamine H3 receptors (H3R). Using an HD mouse striatal cell model and HD mouse organotypic brain slices we found that D1R-induced cell death signaling and neuronal degeneration, are mitigated by an H3R antagonist. We demonstrate that the D1R-H3R heteromer is expressed in HD mice at early but not late stages of HD, correlating with HD progression. In accordance, we found this target expressed in human control subjects and low-grade HD patients. Finally, treatment of HD mice with an H3R antagonist prevented cognitive and motor learning deficits and the loss of heteromer expression. Taken together, our results indicate that D1R - H3R heteromers play a pivotal role in dopamine signaling and represent novel targets for treating HD.


Subject(s)
Drug Delivery Systems/methods , Huntington Disease/metabolism , Receptors, Dopamine D1 , Receptors, Histamine H3 , Animals , Cells, Cultured , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Piperidines/pharmacology , Receptors, Dopamine D1/chemistry , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Histamine H3/chemistry , Receptors, Histamine H3/genetics , Receptors, Histamine H3/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Visual Cortex/cytology
3.
Expert Opin Drug Discov ; 14(12): 1297-1312, 2019 12.
Article in English | MEDLINE | ID: mdl-31507210

ABSTRACT

Introduction: G protein-coupled receptors (GPCRs) are a superfamily of membrane proteins highly expressed in the brain that are involved in almost all functions of the CNS. During the last twenty years, a large number of GPCRs have been reported to form homodimers, heterodimers and higher order oligomers. Areas covered: This review summarizes the functional and pharmacological characteristics of the dopamine D1 receptor (D1R) interactome constituted by heteromers with GPCRs or non-GPCRs. The review also focuses on heteromer-selective ligands reported for GPCRs, including those for the receptor-based interactome of D1R. Expert opinion: Since the D1R plays a key role in basal ganglia motor control, where all the mentioned D1R heteromers are present, the study of allosteric interactions within the D1R interactome may be of high therapeutic interest for treating motor dysfunction. Moreover, several of these heteromers have also been detected in the prefrontal cortex and hippocampus, where they are involved in learning, memory and attention dysfunction. We propose that drugs targeting specific D1R heteromers in the CNS will be more effective and safer, resulting in a reduction of side effects compared with traditional drugs targeting monomeric receptors. Heteromer-selective ligands will have a big impact on drug development with many pharmacological and clinical implications.


Subject(s)
Drug Development/methods , Receptors, Dopamine D1/drug effects , Receptors, G-Protein-Coupled/drug effects , Animals , Brain/drug effects , Brain/metabolism , Drug Design , Humans , Ligands , Receptors, Dopamine D1/metabolism , Receptors, G-Protein-Coupled/metabolism
4.
Adv Pharmacol ; 84: 37-78, 2019.
Article in English | MEDLINE | ID: mdl-31229177

ABSTRACT

Dopaminergic and purinergic signaling play a pivotal role in neurological diseases associated with motor symptoms, including Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis, Huntington disease, Restless Legs Syndrome (RLS), spinal cord injury (SCI), and ataxias. Extracellular dopamine and adenosine exert their functions interacting with specific dopamine (DR) or adenosine (AR) receptors, respectively, expressed on the surface of target cells. These receptors are members of the family A of G protein-coupled receptors (GPCRs), which is the largest protein superfamily in mammalian genomes. GPCRs are target of about 40% of all current marketed drugs, highlighting their importance in clinical medicine. The striatum receives the densest dopamine innervations and contains the highest density of dopamine receptors. The modulatory role of adenosine on dopaminergic transmission depends largely on the existence of antagonistic interactions mediated by specific subtypes of DRs and ARs, the so-called A2AR-D2R and A1R-D1R interactions. Due to the dopamine/adenosine antagonism in the CNS, it was proposed that ARs and DRs could form heteromers in the neuronal cell surface. Therefore, adenosine can affect dopaminergic signaling through receptor-receptor interactions and by modulations in their shared intracellular pathways in the striatum and spinal cord. In this work we describe the allosteric modulations between GPCR protomers, focusing in those of adenosine and dopamine within the A1R-D1R heteromeric complex, which is involved in RLS. We also propose that the knowledge about the intricate allosteric interactions within the A1R-D1R heterotetramer, may facilitate the treatment of motor alterations, not only when the dopamine pathway is hyperactivated (RLS, chorea, etc.) but also when motor function is decreased (SCI, aging, PD, etc.).


Subject(s)
Protein Multimerization , Receptor, Adenosine A1/chemistry , Receptors, Dopamine/chemistry , Restless Legs Syndrome/drug therapy , Adenosine/metabolism , Animals , Dopamine/metabolism , Humans , Receptor, Adenosine A1/metabolism , Receptors, Dopamine/metabolism
5.
Mol Neurobiol ; 56(10): 6756-6769, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30919214

ABSTRACT

Several studies found in vitro evidence for heteromerization of dopamine D1 receptors (D1R) and D3 receptors (D3R), and it has been postulated that functional D1R-D3R heteromers that are normally present in the ventral striatum mediate synergistic locomotor-activating effects of D1R and D3R agonists in rodents. Based also on results obtained in vitro, with mammalian transfected cells, it has been hypothesized that those behavioral effects depend on a D1R-D3R heteromer-mediated G protein-independent signaling. Here, we demonstrate the presence on D1R-D3R heteromers in the mouse ventral striatum by using a synthetic peptide that selectively destabilizes D1R-D3R heteromers. Parallel locomotor activity and ex vivo experiments in reserpinized mice and in vitro experiments in D1R-D3R mammalian transfected cells were performed to dissect the signaling mechanisms of D1R-D3R heteromers. Co-administration of D1R and D3R agonists in reserpinized mice produced synergistic locomotor activation and a selective synergistic AKT phosphorylation in the most ventromedial region of the striatum in the shell of the nucleus accumbens. Application of the destabilizing peptide in transfected cells and in the shell of the nucleus accumbens allowed demonstrating that both in vitro and in vivo co-activation of D3R induces a switch from G protein-dependent to G protein-independent D1R-mediated signaling determined by D1R-D3R heteromerization. The results therefore demonstrate that a biased G protein-independent signaling of D1R-D3R heteromers localized in the shell of the nucleus accumbens mediate the locomotor synergistic effects of D1R and D3R agonists in reserpinized mice.


Subject(s)
GTP-Binding Proteins/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Synergism , HEK293 Cells , Humans , Isoquinolines/pharmacology , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Peptides/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Dopamine D3/antagonists & inhibitors , Salicylamides/pharmacology , Sulfonamides/pharmacology
6.
Pharmacol Res ; 139: 337-347, 2019 01.
Article in English | MEDLINE | ID: mdl-30472462

ABSTRACT

An increasing number of G protein-coupled receptors (GPCRs) have been reported to be expressed in the plasma membrane as dimers. Since most ligand binding data are currently fitted by classical equations developed only for monomeric receptors, the interpretation of data could be misleading in the presence of GPCR dimers. On the other hand, the equations developed from dimer receptor models assuming the existence of two orthosteric binding sites within the dimeric molecule offer the possibility to directly calculate macroscopic equilibrium dissociation constants for the two sites, an index of cooperativity (DC) that reflects the molecular communication within the dimer and, importantly, a constant of radioligand-competitor allosteric interaction (KDAB) in competitive assays. Here, we provide a practical way to fit competitive binding data that allows the interpretation of apparently anomalous results, such as competition curves that could be either bell-shaped, monophasic or biphasic depending on the assay conditions. The consideration of a radioligand-competitor allosteric interaction allows fitting these curve patterns both under simulation conditions and in real radioligand binding experiments, obtaining competitor affinity parameters closer to the actual values. Our approach is the first that, assuming the formation of receptor homodimers, is able to explain several experimental results previously considered erroneous due to their impossibility to be fitted. We also deduce the radioligand concentration responsible for the conversion of biphasic to monophasic or to bell-shaped curves in competitive radioligand binding assays. In conclusion, bell-shaped curves in competitive binding experiments constitute evidence for GPCR homodimerization.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Animals , Binding, Competitive , Brain , Cell Membrane , Protein Multimerization , Radioligand Assay , Sheep
7.
J Med Chem ; 61(20): 9335-9346, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30257092

ABSTRACT

Bivalent ligands have emerged as chemical tools to study G protein-coupled receptor dimers. Using a combination of computational, chemical, and biochemical tools, here we describe the design of bivalent ligand 13 with high affinity ( KDB1 = 21 pM) for the dopamine D2 receptor (D2R) homodimer. Bivalent ligand 13 enhances the binding affinity relative to monovalent compound 15 by 37-fold, indicating simultaneous binding at both protomers. Using synthetic peptides with amino acid sequences of transmembrane (TM) domains of D2R, we provide evidence that TM6 forms the interface of the homodimer. Notably, the disturber peptide TAT-TM6 decreased the binding of bivalent ligand 13 by 52-fold and had no effect on monovalent compound 15, confirming the D2R homodimer through TM6 ex vivo. In conclusion, by using a versatile multivalent chemical platform, we have developed a precise strategy to generate a true bivalent ligand that simultaneously targets both orthosteric sites of the D2R homodimer.


Subject(s)
Drug Design , Protein Multimerization , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Animals , CHO Cells , Cricetulus , Female , Ligands , Male , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Sheep
8.
Mol Neurobiol ; 55(11): 8438-8454, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29552726

ABSTRACT

The poor norepinephrine innervation and high density of Gi/o-coupled α2A- and α2C-adrenoceptors in the striatum and the dense striatal dopamine innervation have prompted the possibility that dopamine could be an effective adrenoceptor ligand. Nevertheless, the reported adrenoceptor agonistic properties of dopamine are still inconclusive. In this study, we analyzed the binding of norepinephrine, dopamine, and several compounds reported as selective dopamine D2-like receptor ligands, such as the D3 receptor agonist 7-OH-PIPAT and the D4 receptor agonist RO-105824, to α2-adrenoceptors in cortical and striatal tissue, which express α2A-adrenoceptors and both α2A- and α2C-adrenoceptors, respectively. The affinity of dopamine for α2-adrenoceptors was found to be similar to that for D1-like and D2-like receptors. Moreover, the exogenous dopamine receptor ligands also showed high affinity for α2A- and α2C-adrenoceptors. Their ability to activate Gi/o proteins through α2A- and α2C-adrenoceptors was also analyzed in transfected cells with bioluminescent resonance energy transfer techniques. The relative ligand potencies and efficacies were dependent on the Gi/o protein subtype. Furthermore, dopamine binding to α2-adrenoceptors was functional, inducing changes in dynamic mass redistribution, adenylyl cyclase activity, and ERK1/2 phosphorylation. Binding events were further studied with computer modeling of ligand docking. Docking of dopamine at α2A- and α2C-adrenoceptors was nearly identical to its binding to the crystallized D3 receptor. Therefore, we provide conclusive evidence that α2A- and α2C-adrenoceptors are functional receptors for norepinephrine, dopamine, and other previously assumed selective D2-like receptor ligands, which calls for revisiting previous studies with those ligands.


Subject(s)
Dopamine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Dopamine/metabolism , Adenylyl Cyclases/metabolism , Animals , Cerebral Cortex/metabolism , Clonidine/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Idazoxan/analogs & derivatives , Idazoxan/pharmacology , Ligands , Neostriatum/metabolism , Norepinephrine/metabolism , Phosphorylation/drug effects , Quinpirole/pharmacology , Sheep , Tetrahydronaphthalenes/pharmacology
9.
Front Pharmacol ; 9: 106, 2018.
Article in English | MEDLINE | ID: mdl-29497379

ABSTRACT

Adenosine is an endogenous purine nucleoside that acts in all living systems as a homeostatic network regulator through many pathways, which are adenosine receptor (AR)-dependent and -independent. From a metabolic point of view, adenosine deaminase (ADA) is an essential protein in the regulation of the total intracellular and extracellular adenosine in a tissue. In addition to its cytosolic localization, ADA is also expressed as an ecto-enzyme on the surface of different cells. Dipeptidyl peptidase IV (CD26) and some ARs act as binding proteins for extracellular ADA in humans. Since CD26 and ARs interact with ADA at opposite sites, we have investigated if ADA can function as a cell-to-cell communication molecule by bridging the anchoring molecules CD26 and A2AR present on the surfaces of the interacting cells. By combining site-directed mutagenesis of ADA amino acids involved in binding to A2AR and a modification of the bioluminescence resonance energy transfer (BRET) technique that allows detection of interactions between two proteins expressed in different cell populations with low steric hindrance (NanoBRET), we show direct evidence of the specific formation of trimeric complexes CD26-ADA-A2AR involving two cells. By dynamic mass redistribution assays and ligand binding experiments, we also demonstrate that A2AR-NanoLuc fusion proteins are functional. The existence of this ternary complex is in good agreement with the hypothesis that ADA could bridge T-cells (expressing CD26) and dendritic cells (expressing A2AR). This is a new metabolic function for ecto-ADA that, being a single chain protein, it has been considered as an example of moonlighting protein, because it performs more than one functional role (as a catalyst, a costimulator, an allosteric modulator and a cell-to-cell connector) without partitioning these functions in different subunits.

10.
BMC Biol ; 16(1): 24, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29486745

ABSTRACT

BACKGROUND: G-protein-coupled receptor (GPCR) heteromeric complexes have distinct properties from homomeric GPCRs, giving rise to new receptor functionalities. Adenosine receptors (A1R or A2AR) can form A1R-A2AR heteromers (A1-A2AHet), and their activation leads to canonical G-protein-dependent (adenylate cyclase mediated) and -independent (ß-arrestin mediated) signaling. Adenosine has different affinities for A1R and A2AR, allowing the heteromeric receptor to detect its concentration by integrating the downstream Gi- and Gs-dependent signals. cAMP accumulation and ß-arrestin recruitment assays have shown that, within the complex, activation of A2AR impedes signaling via A1R. RESULTS: We examined the mechanism by which A1-A2AHet integrates Gi- and Gs-dependent signals. A1R blockade by A2AR in the A1-A2AHet is not observed in the absence of A2AR activation by agonists, in the absence of the C-terminal domain of A2AR, or in the presence of synthetic peptides that disrupt the heteromer interface of A1-A2AHet, indicating that signaling mediated by A1R and A2AR is controlled by both Gi and Gs proteins. CONCLUSIONS: We identified a new mechanism of signal transduction that implies a cross-communication between Gi and Gs proteins guided by the C-terminal tail of the A2AR. This mechanism provides the molecular basis for the operation of the A1-A2AHet as an adenosine concentration-sensing device that modulates the signals originating at both A1R and A2AR.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P1/metabolism , Signal Transduction/physiology , Amino Acid Sequence , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/genetics , HEK293 Cells , Humans , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/genetics
11.
Nat Commun ; 9(1): 1242, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593213

ABSTRACT

G protein-coupled receptors (GPCRs), G proteins and adenylyl cyclase (AC) comprise one of the most studied transmembrane cell signaling pathways. However, it is unknown whether the ligand-dependent interactions between these signaling molecules are based on random collisions or the rearrangement of pre-coupled elements in a macromolecular complex. Furthermore, it remains controversial whether a GPCR homodimer coupled to a single heterotrimeric G protein constitutes a common functional unit. Using a peptide-based approach, we here report evidence for the existence of functional pre-coupled complexes of heteromers of adenosine A2A receptor and dopamine D2 receptor homodimers coupled to their cognate Gs and Gi proteins and to subtype 5 AC. We also demonstrate that this macromolecular complex provides the necessary frame for the canonical Gs-Gi interactions at the AC level, sustaining the ability of a Gi-coupled GPCR to counteract AC activation mediated by a Gs-coupled GPCR.


Subject(s)
Adenylyl Cyclases/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Animals , Bacterial Proteins/metabolism , Computer Simulation , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Luminescent Proteins/metabolism , Macromolecular Substances , Neurons/metabolism , Peptides/chemistry , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Rats , Rats, Sprague-Dawley , Signal Transduction
12.
J Neurosci ; 37(5): 1176-1186, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28007761

ABSTRACT

The neuropeptide galanin has been shown to interact with the opioid system. More specifically, galanin counteracts the behavioral effects of the systemic administration of µ-opioid receptor (MOR) agonists. Yet the mechanism responsible for this galanin-opioid interaction has remained elusive. Using biophysical techniques in mammalian transfected cells, we found evidence for selective heteromerization of MOR and the galanin receptor subtype Gal1 (Gal1R). Also in transfected cells, a synthetic peptide selectively disrupted MOR-Gal1R heteromerization as well as specific interactions between MOR and Gal1R ligands: a negative cross talk, by which galanin counteracted MAPK activation induced by the endogenous MOR agonist endomorphin-1, and a cross-antagonism, by which a MOR antagonist counteracted MAPK activation induced by galanin. These specific interactions, which represented biochemical properties of the MOR-Gal1R heteromer, could then be identified in situ in slices of rat ventral tegmental area (VTA) with MAPK activation and two additional cell signaling pathways, AKT and CREB phosphorylation. Furthermore, in vivo microdialysis experiments showed that the disruptive peptide selectively counteracted the ability of galanin to block the dendritic dopamine release in the rat VTA induced by local infusion of endomorphin-1, demonstrating a key role of MOR-Gal1R heteromers localized in the VTA in the direct control of dopamine cell function and their ability to mediate antagonistic interactions between MOR and Gal1R ligands. The results also indicate that MOR-Gal1R heteromers should be viewed as targets for the treatment of opioid use disorders. SIGNIFICANCE STATEMENT: The µ-opioid receptor (MOR) localized in the ventral tegmental area (VTA) plays a key role in the reinforcing and addictive properties of opioids. With parallel in vitro experiments in mammalian transfected cells and in situ and in vivo experiments in rat VTA, we demonstrate that a significant population of these MORs form functional heteromers with the galanin receptor subtype Gal1 (Gal1R), which modulate the activity of the VTA dopaminergic neurons. The MOR-Gal1R heteromer can explain previous results showing antagonistic galanin-opioid interactions and offers a new therapeutic target for the treatment of opioid use disorder.


Subject(s)
Receptors, Galanin/metabolism , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein , Dopaminergic Neurons/drug effects , Galanin/pharmacology , HEK293 Cells , Humans , Ligands , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein v-akt/physiology , Phosphorylation , Rats , Receptor Cross-Talk , Receptor, Galanin, Type 1/genetics , Receptor, Galanin, Type 1/metabolism , Receptor, Galanin, Type 2/genetics , Receptor, Galanin, Type 2/metabolism , Receptors, Galanin/genetics , Receptors, Opioid, mu/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
13.
Expert Opin Drug Discov ; 11(7): 641-64, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27135354

ABSTRACT

INTRODUCTION: Dopamine is a neurotransmitter widely distributed in both the periphery and the central nervous system (CNS). Its physiological effects are mediated by five closely related G protein-coupled receptors (GPCRs) that are divided into two major subclasses: the D1-like (D1, D5) and the D2-like (D2, D3, D4) receptors. D3 receptors (D3Rs) have the highest density in the limbic areas of the brain, which are associated with cognitive and emotional functions. These receptors are therefore attractive targets for therapeutic management. AREAS COVERED: This review summarizes the functional and pharmacological characteristics of D3Rs, including the design and clinical relevance of full agonists, partial agonists and antagonists, as well as the capacity of these receptors to form active homodimers, heterodimers or higher order receptor complexes as pharmacological targets in several neurological and neurodegenerative disorders. EXPERT OPINION: The high sequence homology between D3R and the D2-type challenges the development of D3R-selective compounds. The design of new D3R-preferential ligands with improved physicochemical properties should provide a better pharmacokinetic/bioavailability profile and lesser toxicity than is found with existing D3R ligands. It is also essential to optimize D3R affinity and, especially, D3R vs. D2-type binding and functional selectivity ratios. Developing allosteric and bitopic ligands should help to improve the D3R selectivity of these drugs. As most evidence points to the ability of GPCRs to form homomers and heteromers, the most promising therapeutic strategy in the future is likely to involve the application of heteromer-selective drugs. These selective ligands would display different affinities for a given receptor depending on the receptor partners within the heteromer. Therefore, designing novel compounds that specifically target and modulate D1R-D3R heteromers would be an interesting approach for the treatment of levodopa (L-DOPA)-induced dyskinesias.


Subject(s)
Drug Design , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/antagonists & inhibitors , Animals , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Drug Partial Agonism , Humans , Levodopa/adverse effects , Ligands , Molecular Targeted Therapy , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Receptors, Dopamine D3/metabolism
14.
BMC Biol ; 14: 26, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27048449

ABSTRACT

BACKGROUND: G-protein-coupled receptors (GPCRs), in the form of monomers or homodimers that bind heterotrimeric G proteins, are fundamental in the transfer of extracellular stimuli to intracellular signaling pathways. Different GPCRs may also interact to form heteromers that are novel signaling units. Despite the exponential growth in the number of solved GPCR crystal structures, the structural properties of heteromers remain unknown. RESULTS: We used single-particle tracking experiments in cells expressing functional adenosine A1-A2A receptors fused to fluorescent proteins to show the loss of Brownian movement of the A1 receptor in the presence of the A2A receptor, and a preponderance of cell surface 2:2 receptor heteromers (dimer of dimers). Using computer modeling, aided by bioluminescence resonance energy transfer assays to monitor receptor homomerization and heteromerization and G-protein coupling, we predict the interacting interfaces and propose a quaternary structure of the GPCR tetramer in complex with two G proteins. CONCLUSIONS: The combination of results points to a molecular architecture formed by a rhombus-shaped heterotetramer, which is bound to two different interacting heterotrimeric G proteins (Gi and Gs). These novel results constitute an important advance in understanding the molecular intricacies involved in GPCR function.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Purinergic P1/chemistry , Receptors, Purinergic P1/metabolism , Animals , HEK293 Cells , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary
15.
Biochem Soc Trans ; 44(2): 595-600, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27068975

ABSTRACT

Heteromers of G-protein-coupled receptors (GPCRs) have emerged as potential novel targets for drug development. Accumulating evidence indicates that GPCRs can form homodimers and heteromers, with homodimers being the predominant species and oligomeric receptors being formed as multiples of dimers. Recently, heterotetrameric structures have been proposed for dopamine D1receptor (D1R)-dopamine D3receptor (D3R) and adenosine A2Areceptor (A2AR)-dopamine D2receptor (D2R) heteromers. The structural model proposed for these complexes is a heteromer constituted by two receptor homodimers. The existence of GPCR homodimers and heteromers provides a structural basis for inter-protomer allosteric mechanisms that might account for a multiplicity of unique pharmacological properties. In this review, we focus on the A2AR-D2R heterotetramer as an example of an oligomeric structure that is key in the modulation of striatal neuronal function. We also review the interfaces involved in this and other recently reported heteromers of GPCRs. Furthermore, we discuss several published studies showing theex vivoexpression of A2AR-D2R heteromers. The ability of A2AR agonists to decrease the affinity of D2R agonists has been reported and, on the basis of this interaction, A2AR antagonists have been proposed as potential drugs for the treatment of Parkinson's disease. The heterotetrameric structure of the A2AR-D2R complex offers a novel model that can provide new clues about how to adjust the drug dosage to the expected levels of endogenous adenosine.


Subject(s)
Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Allosteric Regulation , Dimerization , Humans , Ligands , Molecular Structure , Receptor, Adenosine A2A/chemistry , Receptors, Dopamine D2/chemistry
16.
Mol Neurobiol ; 53(8): 5436-45, 2016 10.
Article in English | MEDLINE | ID: mdl-26452359

ABSTRACT

Dopamine receptors in striatum are important for healthy brain functioning and are the target of levodopa-based therapy in Parkinson's disease. Lateralization of dopaminergic neurotransmission in striata from different hemispheres occurs in patients, but also in healthy individuals. Our data show that the affinity of dopamine binding to dopamine D1 receptors is significantly higher in left than in right striatum. Analysis of data from radioligand binding to striatal samples from naïve, 6-hydroxydopamine lesioned, levodopa-treated and levodopa-induced dyskinetic rats shows differential receptor structure and gives hints on the causes of right/left lateralization of dopamine binding to striatal D1 receptors. Moreover, binding data showed loss of lateralization in levodopa (L-DOPA)-induced dyskinetic rats.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Functional Laterality , Receptors, Dopamine D1/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/pharmacology , Dyskinesia, Drug-Induced/metabolism , Functional Laterality/drug effects , Male , Rats, Wistar , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/metabolism
17.
Proc Natl Acad Sci U S A ; 112(27): E3609-18, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26100888

ABSTRACT

Adenosine A2A receptor (A2AR)-dopamine D2 receptor (D2R) heteromers are key modulators of striatal neuronal function. It has been suggested that the psychostimulant effects of caffeine depend on its ability to block an allosteric modulation within the A2AR-D2R heteromer, by which adenosine decreases the affinity and intrinsic efficacy of dopamine at the D2R. We describe novel unsuspected allosteric mechanisms within the heteromer by which not only A2AR agonists, but also A2AR antagonists, decrease the affinity and intrinsic efficacy of D2R agonists and the affinity of D2R antagonists. Strikingly, these allosteric modulations disappear on agonist and antagonist coadministration. This can be explained by a model that considers A2AR-D2R heteromers as heterotetramers, constituted by A2AR and D2R homodimers, as demonstrated by experiments with bioluminescence resonance energy transfer and bimolecular fluorescence and bioluminescence complementation. As predicted by the model, high concentrations of A2AR antagonists behaved as A2AR agonists and decreased D2R function in the brain.


Subject(s)
Corpus Striatum/metabolism , Protein Multimerization , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Adenosine A2 Receptor Agonists/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Binding, Competitive/drug effects , Bioluminescence Resonance Energy Transfer Techniques , CHO Cells , Cricetinae , Cricetulus , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Dopamine D2 Receptor Antagonists/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kinetics , Male , Microscopy, Confocal , Protein Binding/drug effects , Rats, Sprague-Dawley , Receptor, Adenosine A2A/chemistry , Receptors, Dopamine D2/chemistry , Sheep , Time Factors
18.
Eur J Med Chem ; 97: 173-80, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25969169

ABSTRACT

Ligands acting at multiple dopamine receptors hold potential as therapeutic agents for a number of neurodegenerative disorders. Specifically, compounds able to bind at D1R and D2R with high affinity could restore the effects of dopamine depletion and enhance motor activation on degenerated nigrostriatal dopaminergic systems. We have directed our research towards the synthesis and characterisation of heterocycle-peptide hybrids based on the indolo[2,3-a]quinolizidine core. This privileged structure is a water-soluble and synthetically accessible scaffold with affinity for diverse GPCRs. Herein we have prepared a solid-phase combinatorial library of 80 indoloquinolizidine-peptides to identify compounds with enhanced binding affinity at D2R, a receptor that is crucial to re-establish activity on dopamine-depleted degenerated GABAergic neurons. We applied computational tools and high-throughput screening assays to identify 9a{1,3,3} as a ligand for dopamine receptors with nanomolar affinity and agonist activity at D2R. Our results validate the application of indoloquinolizidine-peptide combinatorial libraries to fine-tune the pharmacological profiles of multiple ligands at D1 and D2 dopamine receptors.


Subject(s)
Drug Design , Peptides/chemical synthesis , Quinolizidines/chemical synthesis , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Small Molecule Libraries/pharmacology , Animals , Binding Sites , CHO Cells , Combinatorial Chemistry Techniques , Cricetulus , Indoles , Models, Molecular , Molecular Dynamics Simulation , Peptides/chemistry , Peptides/pharmacology , Quinolizidines/chemistry , Quinolizidines/pharmacology , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
19.
Med Res Rev ; 35(1): 85-125, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24933472

ABSTRACT

Interest in adenosine deaminase (ADA) in the context of medicine has mainly focused on its enzymatic activity. This is justified by the importance of the reaction catalyzed by ADA not only for the intracellular purine metabolism, but also for the extracellular purine metabolism as well, because of its capacity as a regulator of the concentration of extracellular adenosine that is able to activate adenosine receptors (ARs). In recent years, other important roles have been described for ADA. One of these, with special relevance in immunology, is the capacity of ADA to act as a costimulator, promoting T-cell proliferation and differentiation mainly by interacting with the differentiation cluster CD26. Another role is the ability of ADA to act as an allosteric modulator of ARs. These receptors have very general physiological implications, particularly in the neurological system where they play an important role. Thus, ADA, being a single chain protein, performs more than one function, consistent with the definition of a moonlighting protein. Although ADA has never been associated with moonlighting proteins, here we consider ADA as an example of this family of multifunctional proteins. In this review, we discuss the different roles of ADA and their pathological implications. We propose a mechanism by which some of their moonlighting functions can be coordinated. We also suggest that drugs modulating ADA properties may act as modulators of the moonlighting functions of ADA, giving them additional potential medical interest.


Subject(s)
Adenosine Deaminase/drug effects , Drug Design , Animals , Humans
20.
Chem Biol ; 21(11): 1546-56, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25457181

ABSTRACT

The pharmacological significance of the adenosine A2A receptor (A2AR)-dopamine D2 receptor (D2R) heteromer is well established and it is being considered as an important target for the treatment of Parkinson's disease and other neuropsychiatric disorders. However, the physiological factors that control its distinctive biochemical properties are still unknown. We demonstrate that different intracellular Ca2+ levels exert a differential modulation of A2AR-D2R heteromer-mediated adenylyl-cyclase and MAPK signaling in striatal cells. This depends on the ability of low and high Ca2+ levels to promote a selective interaction of the heteromer with the neuronal Ca2+-binding proteins NCS-1 and calneuron-1, respectively. These Ca2+-binding proteins differentially modulate allosteric interactions within the A2AR-D2R heteromer, which constitutes a unique cellular device that integrates extracellular (adenosine and dopamine) and intracellular (Ca+2) signals to produce a specific functional response.


Subject(s)
Calcium/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenylyl Cyclases/metabolism , Animals , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Calmodulin/metabolism , Cells, Cultured , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuronal Calcium-Sensor Proteins/antagonists & inhibitors , Neuronal Calcium-Sensor Proteins/genetics , Neuronal Calcium-Sensor Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...