Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(1): 111915, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640316

ABSTRACT

Modes of somatodendritic transmission range from rapid synaptic signaling to protracted regulation over distance. Somatodendritic dopamine secretion in the midbrain leads to D2 receptor-induced modulation of dopamine neurons on the timescale of seconds. Temporally imprecise release mechanisms are often presumed to be at play, and previous work indeed suggested roles for slow Ca2+ sensors. We here use mouse genetics and whole-cell electrophysiology to establish that the fast Ca2+ sensor synaptotagmin-1 (Syt-1) is important for somatodendritic dopamine release. Syt-1 ablation from dopamine neurons strongly reduces stimulus-evoked D2 receptor-mediated inhibitory postsynaptic currents (D2-IPSCs) in the midbrain. D2-IPSCs evoked by paired stimuli exhibit less depression, and high-frequency trains restore dopamine release. Spontaneous somatodendritic dopamine secretion is independent of Syt-1, supporting that its exocytotic mechanisms differ from evoked release. We conclude that somatodendritic dopamine transmission relies on the fast Ca2+ sensor Syt-1, leading to synchronous release in response to the initial stimulus.


Subject(s)
Dopamine , Synaptic Transmission , Animals , Mice , Synaptic Transmission/physiology , Cell Communication , Exocytosis/physiology , Dopaminergic Neurons , Calcium
2.
Brain Struct Funct ; 227(3): 925-941, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34854963

ABSTRACT

G-protein-coupled D2 autoreceptors expressed on dopamine neurons (D2Rs) inhibit transmitter release and cell firing at axonal endings and somatodendritic compartments. Mechanistic details of somatodendritic dopamine release remain unresolved, partly due to insufficient information on the subcellular distribution of D2Rs. Previous studies localizing D2Rs have been hindered by a dearth of antibodies validated for specificity in D2R knockout animals and have been limited by the small sampling areas imaged by electron microscopy. This study utilized sub-diffraction fluorescence microscopy and electron microscopy to examine D2 receptors in a superecliptic pHlourin GFP (SEP) epitope-tagged D2 receptor knockin mouse. Incubating live slices with an anti-SEP antibody achieved the selective labeling of plasma membrane-associated receptors for immunofluorescent imaging over a large area of the substantia nigra pars compacta (SNc). SEP-D2Rs appeared as puncta-like structures along the surface of dendrites and soma of dopamine neurons visualized by antibodies to tyrosine hydroxylase (TH). TH-associated SEP-D2Rs displayed a cell surface density of 0.66 puncta/µm2, which corresponds to an average frequency of 1 punctum every 1.50 µm. Separate ultrastructural experiments using silver-enhanced immunogold revealed that membrane-bound particles represented 28% of total D2Rs in putative dopamine cells within the SNc. Structures immediately adjacent to dendritic membrane gold particles were unmyelinated axons or axon varicosities (40%), astrocytes (19%), other dendrites (7%), or profiles unidentified (34%) in single sections. Some apposed profiles also expressed D2Rs. Fluorescent and ultrastructural analyses also provided the first visualization of membrane D2Rs at the axon initial segment, a compartment critical for action potential generation. The punctate appearance of anti-SEP staining indicates there is a population of D2Rs organized in discrete signaling sites along the plasma membrane, and for the first time, a quantitative estimate of spatial frequency is provided.


Subject(s)
Receptors, Dopamine D2/metabolism , Substantia Nigra , Animals , Autoreceptors/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mice , Receptors, Dopamine D2/analysis , Substantia Nigra/metabolism
3.
Elife ; 82019 09 05.
Article in English | MEDLINE | ID: mdl-31486769

ABSTRACT

Action potentials trigger neurotransmitter release at active zones, specialized release sites in axons. Many neurons also secrete neurotransmitters or neuromodulators from their somata and dendrites. However, it is unclear whether somatodendritic release employs specialized sites for release, and the molecular machinery for somatodendritic release is not understood. Here, we identify an essential role for the active zone protein RIM in stimulated somatodendritic dopamine release in the midbrain. In mice in which RIMs are selectively removed from dopamine neurons, action potentials failed to evoke significant somatodendritic release detected via D2 receptor-mediated currents. Compellingly, spontaneous dopamine release was normal upon RIM knockout. Dopamine neuron morphology, excitability, and dopamine release evoked by amphetamine, which reverses dopamine transporters, were also unaffected. We conclude that somatodendritic release employs molecular scaffolds to establish secretory sites for rapid dopamine signaling during firing. In contrast, basal release that is independent of action potential firing does not require RIM.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Dendrites/metabolism , Dopamine/metabolism , Mesencephalon/metabolism , Action Potentials , Animals , Mice , Receptors, Dopamine D2/metabolism , Synaptic Transmission
4.
Elife ; 72018 03 28.
Article in English | MEDLINE | ID: mdl-29589831

ABSTRACT

Phosphorylation of the µ-opioid receptor (MOR) is known as a key step in desensitization and internalization but the role in the development of long-term tolerance at the cellular level is not known. Viral expression of wild type (exWT) and mutant MORs, where all phosphorylation sites on the C-terminus (Total Phosphorylation Deficient (TPD)) were mutated to alanine, were examined in locus coeruleus neurons in a MOR knockout rat. Both receptors activated potassium conductance similar to endogenous receptors in wild type animals. The exWT receptors, like endogenous receptors, acutely desensitized, internalized and, after chronic morphine treatment, displayed signs of tolerance. However, TPD receptors did not desensitize or internalize with agonist treatment. In addition the TPD receptors did not develop cellular tolerance following chronic morphine treatment. Thus C-terminal phosphorylation is necessary for the expression of acute desensitization, trafficking and one sign of long-term tolerance to morphine at the cellular level.


Subject(s)
Protein Processing, Post-Translational , Receptors, Opioid, mu/metabolism , Animals , Animals, Genetically Modified , Gene Knockout Techniques , Locus Coeruleus/cytology , Morphine/metabolism , Neurons/metabolism , Phosphorylation , Potassium/metabolism , Rats
5.
Sci Rep ; 7(1): 4379, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28663556

ABSTRACT

Dendritic release of dopamine activates dopamine D2 autoreceptors, which are inhibitory G protein-coupled receptors (GPCRs), to decrease the excitability of dopamine neurons. This study used tagged D2 receptors to identify the localization and distribution of these receptors in living midbrain dopamine neurons. GFP-tagged D2 receptors were found to be unevenly clustered on the soma and dendrites of dopamine neurons within the substantia nigra pars compacta (SNc). Physiological signaling and desensitization of the tagged receptors were not different from wild type receptors. Unexpectedly, upon desensitization the tagged D2 receptors were not internalized. When tagged D2 receptors were expressed in locus coeruleus neurons, a desensitizing protocol induced significant internalization. Likewise, when tagged µ-opioid receptors were expressed in dopamine neurons they too were internalized. The distribution and lack of agonist-induced internalization of D2 receptors on dopamine neurons indicate a purposefully regulated localization of these receptors.


Subject(s)
Autoreceptors/metabolism , Receptors, Dopamine D2/metabolism , Action Potentials , Animals , Autoreceptors/genetics , Dopamine , Dopaminergic Neurons/metabolism , Endocytosis , Female , Gene Expression , Gene Knock-In Techniques , Genes, Reporter , Male , Mice , Mice, Transgenic , Protein Transport , Receptors, Dopamine D2/genetics , Recombinant Fusion Proteins , Signal Transduction
6.
Elife ; 42015 Aug 26.
Article in English | MEDLINE | ID: mdl-26308580

ABSTRACT

D2 autoreceptors regulate dopamine release throughout the brain. Two isoforms of the D2 receptor, D2S and D2L, are expressed in midbrain dopamine neurons. Differential roles of these isoforms as autoreceptors are poorly understood. By virally expressing the isoforms in dopamine neurons of D2 receptor knockout mice, this study assessed the calcium-dependence and drug-induced plasticity of D2S and D2L receptor-dependent G protein-coupled inwardly rectifying potassium (GIRK) currents. The results reveal that D2S, but not D2L receptors, exhibited calcium-dependent desensitization similar to that exhibited by endogenous autoreceptors. Two pathways of calcium signaling that regulated D2 autoreceptor-dependent GIRK signaling were identified, which distinctly affected desensitization and the magnitude of D2S and D2L receptor-dependent GIRK currents. Previous in vivo cocaine exposure removed calcium-dependent D2 autoreceptor desensitization in wild type, but not D2S-only mice. Thus, expression of D2S as the exclusive autoreceptor was insufficient for cocaine-induced plasticity, implying a functional role for the co-expression of D2S and D2L autoreceptors.


Subject(s)
Autoreceptors/metabolism , Calcium/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction , Animals , Cocaine/metabolism , Mice, Knockout , Protein Isoforms/metabolism
7.
Mol Pharmacol ; 88(4): 816-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25934731

ABSTRACT

Sustained activation of G protein-coupled receptors can lead to a rapid decline in signaling through acute receptor desensitization. In the case of the µ-opioid receptor (MOPr), this desensitization may play a role in the development of analgesic tolerance. It is understood that phosphorylation of MOPr promotes association with ß-arrestin proteins, which then facilitates desensitization and receptor internalization. Agonists that induce acute desensitization have been shown to induce a noncanonical high-affinity agonist binding state in MOPr, conferring a persistent memory of prior receptor activation. In the current study, live-cell confocal imaging was used to investigate the role of receptor phosphorylation in agonist binding to MOPr. A phosphorylation cluster in the C-terminal tail of MOPr was identified as a mediator of agonist-induced affinity changes in MOPr. This site is unique from the primary phosphorylation cluster responsible for ß-arrestin binding and internalization. Electrophysiologic measurements of receptor function suggest that both phosphorylation clusters may play a parallel role during acute receptor desensitization. Desensitization was unaffected by alanine mutation of either phosphorylation cluster, but was largely eliminated when both clusters were mutated. Overall, this work suggests that there are multiple effects of MOPr phosphorylation that appear to regulate MOPr function: one affecting ß-arrestin binding and a second affecting agonist binding.


Subject(s)
Analgesics, Opioid/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Arrestins/chemistry , Arrestins/metabolism , Arrestins/pharmacology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Organ Culture Techniques , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Binding/drug effects , Protein Binding/physiology , beta-Arrestins
8.
J Med Chem ; 57(2): 378-90, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24354319

ABSTRACT

Amphetamines are widely abused drugs that interfere with dopamine transport and storage. Recently, however, another mechanism of action was identified: stereoselective activation of the GαS protein-coupled trace amine-associated receptor 1 (TAAR1). To identify structural determinants of this stereoselectivity, we functionally evaluated six mutant receptors in vitro and then used homology modeling and dynamic simulation to predict drug affinities. Converting Asp102 to Ala rendered mouse and rat TAAR1 (mTAAR1 and rTAAR1, respectively) insensitive to ß-phenylethylamine, amphetamine (AMPH), and methamphetamine (METH). Mutating Met268 in rTAAR1 to Thr shifted the concentration-response profiles for AMPH and METH isomers rightward an order of magnitude, whereas replacing Thr268 with Met in mTAAR1 resulted in profiles leftward shifted 10-30-fold. Replacing Asn287 with Tyr in rTAAR1 produced a mouselike receptor, while the reciprocal mTAAR1 mutant was rTAAR1-like. These results confirm TAAR1 is an AMPH/METH receptor in vitro and establish residues 102 (3.32) and 268 (6.55) as major contributors to AMPH/METH binding with residue 287 (7.39) determining species stereoselectivity.


Subject(s)
Amphetamine/metabolism , Central Nervous System Stimulants/metabolism , Methamphetamine/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Amphetamine/chemistry , Amphetamine/pharmacology , Animals , Binding Sites , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/pharmacology , Cyclic AMP/biosynthesis , HEK293 Cells , Humans , Methamphetamine/chemistry , Methamphetamine/pharmacology , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Rats , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Sequence Homology, Amino Acid , Stereoisomerism
9.
Neuron ; 78(5): 807-12, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23764286

ABSTRACT

G protein-coupled receptors (GPCRs) affect many physiological processes by modulating both intrinsic membrane conductances and synaptic transmission. This study describes spontaneous miniature inhibitory postsynaptic currents mediated by vesicular dopamine release acting locally on metabotropic D2 receptors leading to the activation of a G protein-coupled inwardly rectifying potassium conductance. Thus, individual exocytotic events result in spontaneous GPCR-mediated transmission, similar to synaptic activation of classical ligand-gated ion channels.


Subject(s)
Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Receptors, G-Protein-Coupled/metabolism , Analysis of Variance , Animals , Chelating Agents/pharmacology , Colforsin/pharmacology , Dopamine/metabolism , Dopamine Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , GABA Antagonists/pharmacology , Humans , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Levodopa/pharmacology , Male , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Neurons/drug effects , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques , Receptors, Dopamine D2/genetics , Serotonin Agents/pharmacology , Tyrosine 3-Monooxygenase/genetics
10.
ACS Chem Biol ; 4(3): 209-20, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19256523

ABSTRACT

The trace amine-associated receptor 1 (TAAR(1)) is an aminergic G protein-coupled receptor (GPCR) potently activated by 3-iodothyronamine (1), an endogenous derivative of thyroid hormone. Structure-activity relationship studies on 1 and related agonists showed that the rat and mouse species of TAAR(1) accommodated structural modifications and functional groups on the ethylamine portion and the biaryl ether moiety of the molecule. However, the two receptors clearly exhibited distinct, species-specific ligand preferences despite being remarkably similar with 93% sequence similarity. In this study, we generated single and double mutants of rat and mouse TAAR(1) to probe the molecular recognition of agonists and the underlying basis for the ligand selectivity of rat and mouse TAAR(1). Key, nonconserved specificity determinant residues in transmembranes helices 4 and 7 within the ligand binding site appear to be the primary source of a number of the observed ligand preferences. Residue 7.39 in transmembrane 7 dictated the preference for a beta-phenyl ring, while residue 4.56 in transmembrane 4 was partially responsible for the lower potency of 1 and tyramine for the mouse receptor. Additionally, 1 and tyramine were found to have the same binding mode in rat TAAR(1) despite structure-activity relationship data suggesting the possibility of each molecule having different binding orientations. These findings provide valuable insights into the critical binding site residues involved in the ligand-receptor interaction that can influence compound selectivity and functional activity of aminergic GPCRs.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Thyronines/pharmacology , Amino Acid Sequence , Animals , Humans , Ligands , Mice , Molecular Sequence Data , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Structure, Secondary , Rats , Receptors, G-Protein-Coupled/genetics , Sequence Alignment , Structure-Activity Relationship , Thyronines/chemistry , Tyramine/metabolism , Tyramine/pharmacology
11.
J Med Chem ; 50(12): 2787-98, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17497842

ABSTRACT

3-iodothyronamine (1, T1AM) is a naturally occurring derivative of thyroid hormone that can potently activate the orphan G protein-coupled receptor (GPCR) known as the trace amine-associated receptor 1 (TAAR1). We have previously found that modifying the outer ring of the phenoxyphenethylamine core scaffold of 1 can improve potency and provide potent agonists. In this study, we explored the tolerance of rat and mouse TAAR1 (rTAAR1 and mTAAR1) for structural modifications in the ethylamine portion of 1. We found that incorporating unsaturated hydrocarbon substituents and polar, hydrogen-bond-accepting groups were beneficial for rTAAR1 and mTAAR1, respectively, providing compounds that were equipotent or more potent than 1. Additionally, we have discovered that a naphthyl group is an excellent isosteric replacement for the iodophenyl ring of 1.


Subject(s)
Ethylamines/chemical synthesis , Receptors, G-Protein-Coupled/agonists , Thyronines/chemical synthesis , Animals , Cell Line , Cyclic AMP/biosynthesis , Ethylamines/chemistry , Ethylamines/pharmacology , Humans , Hydrogen Bonding , Mice , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Naphthalenes/pharmacology , Rats , Structure-Activity Relationship , Thyronines/chemistry , Thyronines/pharmacology
12.
J Med Chem ; 49(3): 1101-12, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16451074

ABSTRACT

We have previously shown that several thyronamines, decarboxylated and deiodinated metabolites of the thyroid hormone, potently activate an orphan G protein-coupled receptor in vitro (TAAR1) and induced hypothermia in vivo on a rapid time scale [Scanlan, T. S.; Suchland, K. L.; Hart, M. E.; Chiellini, G.; Huang, Y.; Kruzich, P. J.; Frascarelli, S.; Crossley, D. A.; Bunzow, J. R.; Ronca-Testoni, S.; Lin, E. T.; Hatton, D.; Zucchi, R.; Grandy, D. K. 3-Iodothyronamine is an endogenous and rapid-acting derivative of thyroid hormone. Nat. Med. 2004, 10 (6), 638-642]. Herein, we report the synthesis of these thyronamines. Additionally, a large number of thyroamine derivatives were synthesized in an effort to understand the molecular basis of TAAR1 activation and hypothermia induction. Several derivatives were found to potently activate both rTAAR1 and mTAAR1 in vitro (compounds 77, 85, 91, and 92). When administered to mice at a 50 mg/kg dose, these derivatives all induced significant hypothermia within 60 min and exhibited a hypothermic induction profile analogous to 3-iodothyronamine (1, T(1)AM) except 91, which proved to be more efficacious. On the basis of this result, a dose-dependent profile for 91 was generated and an ED(50) of 30 mumol/kg was calculated. Compound 91 proved to be more potent than T(1)AM for TAAR1 activation and exhibits increased potency and efficacy for hypothermia induction. These data further strengthen the pharmacological correlation linking TAAR1 activation by thyronamines and hypothermia induction in mice.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Thyronines/chemical synthesis , Animals , Body Temperature/drug effects , Cell Line , Cyclic AMP/biosynthesis , Humans , Hypothermia/chemically induced , Mice , Mice, Inbred C57BL , Rats , Receptors, G-Protein-Coupled/genetics , Structure-Activity Relationship , Thyronines/chemistry , Thyronines/pharmacology , Transfection
13.
Genomics ; 85(3): 372-85, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15718104

ABSTRACT

Trace amines are endogenous compounds structurally related to classical biogenic amines that have been studied for decades, triggered by their link to psychiatric conditions of high epidemiological and economical relevance. The understanding of their pharmacology on the molecular level was hampered until the recent discovery of trace-amine-specific receptors. We completed the identification of all members of this novel GPCR family in human, chimpanzee, rat, and mouse and observed remarkable interspecies differences, even between human and chimpanzee. The analysis of the chromosomal localizations, phylogenetic relationships, and ligand pocket vectors reveals three distinct receptor subfamilies. As most of these receptors do not respond to trace amines, each subfamily will presumably have a distinct pharmacological profile, which remains to be identified. We propose a uniform nomenclature describing this novel GPCR family in all mammalian species as trace-amine-associated receptors (TAARs), which resolves the ambiguities and contradictions of the previous naming.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Chromosome Mapping , Humans , Molecular Sequence Data , Phylogeny , Receptors, G-Protein-Coupled/classification , Sequence Homology, Amino Acid
14.
Nat Med ; 10(6): 638-42, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15146179

ABSTRACT

Thyroxine (T(4)) is the predominant form of thyroid hormone (TH). Hyperthyroidism, a condition associated with excess TH, is characterized by increases in metabolic rate, core body temperature and cardiac performance. In target tissues, T(4) is enzymatically deiodinated to 3,5,3'-triiodothyronine (T(3)), a high-affinity ligand for the nuclear TH receptors TR alpha and TR beta, whose activation controls normal vertebrate development and physiology. T(3)-modulated transcription of target genes via activation of TR alpha and TR beta is a slow process, the effects of which manifest over hours and days. Although rapidly occurring effects of TH have been documented, the molecules that mediate these non-genomic effects remain obscure. Here we report the discovery of 3-iodothyronamine (T(1)AM), a naturally occurring derivative of TH that in vitro is a potent agonist of the G protein-coupled trace amine receptor TAR1. Administering T(1)AM in vivo induces profound hypothermia and bradycardia within minutes. T(1)AM treatment also rapidly reduces cardiac output in an ex vivo working heart preparation. These results suggest the existence of a new signaling pathway, stimulation of which leads to rapid physiological and behavioral consequences that are opposite those associated with excess TH.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Thyroid Hormone/metabolism , Signal Transduction/physiology , Thyronines/analogs & derivatives , Thyronines/chemistry , Thyronines/metabolism , Thyroxine/metabolism , Animals , Body Temperature , Brain Chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Hypothermia , Ligands , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Wistar , Thyroxine/chemistry , Time Factors
15.
Biochem Pharmacol ; 63(5): 859-63, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11911837

ABSTRACT

Proteins that bind to G protein-coupled receptors have been identified as regulators of receptor localization and signaling. In our previous studies, a cytoskeletal protein, actin-binding protein 280 (ABP-280), was found to associate with the third cytoplasmic loop of dopamine D(2) receptors. In this study, we demonstrate that ABP-280 also interacts with dopamine D(3) receptors, but not with D(4) receptors. Similar to the dopamine D(2) receptor, the D(3)/ABP-280 association is of signaling importance. In human melanoma M2 cells lacking ABP-280, D(3) receptors were unable to inhibit forskolin-stimulated cyclic AMP (cAMP) production significantly. D(4) receptors, however, exhibited a similar degree of inhibition of forskolin-stimulated cAMP production in ABP-280-deficient M2 cells and ABP-280-replent M2 subclones (A7 cells). Further experiments revealed that the D(3)/ABP-280 interaction was critically dependent upon a 36 amino acid carboxyl domain of the D(3) receptor third loop, which is conserved in the D(2) receptor but not in the D(4) receptor. Our results demonstrate a subtype-specific regulation of dopamine D(2)-family receptor signaling by the cytoskeletal protein ABP-280.


Subject(s)
Adenylyl Cyclases/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, Dopamine D2/metabolism , Adenylyl Cyclase Inhibitors , Filamins , Humans , Radioligand Assay , Receptors, Dopamine D3 , Tumor Cells, Cultured , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...