1
|
Kumar SS, Mountjoy KG. Measuring GPCR-Induced Intracellular Calcium Signaling Using a Quantitative High-Throughput Assay. Methods Mol Biol 2025; 2861:3-22. [PMID: 39395093 DOI: 10.1007/978-1-0716-4164-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Alterations in intracellular calcium are integral to signal transduction pathways for many G-protein-coupled receptors, but this signaling is not well studied. This is mostly due to a lack of reliable, robust, high-throughput, quantitative methods to monitor intracellular calcium concentrations in live cells. Recently, we developed a reliable, robust, quantitative method to measure intracellular calcium levels in which HEK293 cell suspensions loaded with Fura-2/AM are placed in 96-well plates. Minimum and maximum intracellular calcium levels, which are required for converting fluorescence into calcium concentrations, are calibrated using EGTA to chelate calcium and ionomycin to load calcium into cells, respectively. Fluorescence is monitored with a PHERAstar FS plate reader. We provide a detailed method for this high-throughput assay that can be used to quantitate intracellular calcium in endogenous and exogenously (stable or transient) expressed GPCRs in HEK293 cells.
Collapse
Affiliation(s)
- Shree S Kumar
- Faculty of Medical and Health Sciences, Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Kathleen G Mountjoy
- Faculty of Medical and Health Sciences, Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
2
|
Lehtimäki J, Jalava N, Unkila K, Aspegren J, Haapalinna A, Pesonen U. Tasipimidine-the pharmacological profile of a novel orally active selective α 2A-adrenoceptor agonist. Eur J Pharmacol 2022; 923:174949. [PMID: 35405115 DOI: 10.1016/j.ejphar.2022.174949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/05/2022] [Indexed: 11/03/2022]
Abstract
The pharmacological profile of tasipimidine, a novel orally active α2-adrenoceptor agonist developed for situational anxiety and fear in dogs, was studied in various in vitro and in vivo models. In the cell assays, tasipimidine demonstrated binding affinity and full agonism on the human α2A-adrenoceptors with a pEC50 of 7.57, while agonism on the α2B-and α2C-adrenoceptors and the rodent α2D-adrenoceptor was weaker, resulting in pEC50 values of 6.00, 6.29 and 6.56, respectively. Tasipimidine had a low binding affinity on the human α1-adrenoceptors. It had no functional effects in the LNCaP cells expressing endogenously the human α1A-adrenoceptors but was a weak agonist in the Chem-1 cells coexpressing Gα15 protein and α1A-adrenoceptors. In the recombinant CHO cells, although tasipimidine was a weak partial agonist in the inositol monophosphate accumulation assay, it was a full agonist in the intracellular [Ca2+] assay. No functional effects were observed on the human α1B-adrenoceptor, whereas in the rat α1A and α1B-adrenoceptors, tasipimidine was a weak partial agonist. In the rat vas deferens preparations, tasipimidine was a full agonist on the α2D-adrenoceptor but weak partial agonist on the α1-adrenoceptor. The receptor profile of tasipimidine indicated few secondary targets, and no functional effects were observed. Sedative effects of tasipimidine were demonstrated in vivo by the reduced acoustic startle reflex in rats with subcutaneous doses and decreased spontaneous locomotor activity in mice with subcutaneous and higher oral doses. It may be concluded that tasipimidine is an orally active and selective α2A-adrenoceptor agonist.
Collapse
Affiliation(s)
- Jyrki Lehtimäki
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland.
| | - Niina Jalava
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Kaisa Unkila
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - John Aspegren
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Antti Haapalinna
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Ullamari Pesonen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| |
Collapse
|
3
|
Singh K, Senatorov IS, Cheshmehkani A, Karmokar PF, Moniri NH. The Skeletal Muscle Relaxer Cyclobenzaprine Is a Potent Non-Competitive Antagonist of Histamine H1 Receptors. J Pharmacol Exp Ther 2022; 380:202-209. [PMID: 34992159 DOI: 10.1124/jpet.121.000998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/02/2021] [Indexed: 11/22/2022] Open
Abstract
Cyclobenzaprine is a tricyclic dimethylpropanamine skeletal muscle relaxant, which is used clinically to decrease muscle spasm and hypercontractility, as well as acute musculoskeletal pain. Although the absolute mechanism of action of cyclobenzaprine remains elusive, it is known to mediate its effects centrally via inhibition of tonic somatic motor function, likely through modulation of noradrenergic and serotonergic systems. While cyclobenzaprine is effective as a muscle relaxant, greater than 30% of patients experience drowsiness and sedative-hypnotic effects, yet the mechanisms that cause this adverse effect are also undescribed. Based on this common adverse effect profile and the structural similarity of cyclobenzaprine to tricyclic antidepressants, as well as ethanolamine first-generation antihistamines, we hypothesized that cyclobenzaprine facilitates sedative effects via off-target antagonism of central histamine H1 receptors (H1Rs). Here, for the first time, we present data that demonstrate that cyclobenzaprine exhibits low nanomolar affinity for the cloned human H1R, as well as that expressed in both rat and mouse brain. Using saturation radioligand binding, we also demonstrate that cyclobenzaprine binds to the H1R in a noncompetitive manner. Similarly, functional assays measuring both Ca+2 influx and novel TRUPATH G-protein subunit bioluminescence resonance energy transfer biosensors reveal that cyclobenzaprine also blocks histamine-mediated H1R functional activity in a noncompetitive manner, whereas the classical H1R antagonist diphenhydramine does so competitively. Given that cyclobenzaprine readily crosses the blood-brain barrier and its muscle relaxant effects occur centrally, our data suggest that off-target central antagonism of H1R by cyclobenzaprine facilitates the significant sedative effect of this agent seen in patients. SIGNIFICANCE STATEMENT: Cyclobenzaprine, a clinically used muscle relaxant that is strongly linked to sedation, demonstrates high-affinity noncompetitive antagonism at the histamine H1 receptor. This effect likely modulates the high degree of sedation that patients experience.
Collapse
Affiliation(s)
- Kirti Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia (K.S., I.S.S., A.C., P.F.K., N.H.M.) and Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, Georgia (N.H.M.)
| | - Ilya S Senatorov
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia (K.S., I.S.S., A.C., P.F.K., N.H.M.) and Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, Georgia (N.H.M.)
| | - Ameneh Cheshmehkani
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia (K.S., I.S.S., A.C., P.F.K., N.H.M.) and Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, Georgia (N.H.M.)
| | - Priyanka F Karmokar
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia (K.S., I.S.S., A.C., P.F.K., N.H.M.) and Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, Georgia (N.H.M.)
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, Georgia (K.S., I.S.S., A.C., P.F.K., N.H.M.) and Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, Georgia (N.H.M.)
| |
Collapse
|
4
|
Kumar SS, Ward ML, Mountjoy KG. Quantitative high-throughput assay to measure MC4R-induced intracellular calcium. J Mol Endocrinol 2021; 66:285-297. [PMID: 33739935 PMCID: PMC8111326 DOI: 10.1530/jme-20-0285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022]
Abstract
The melanocortin-4 receptor (MC4R), a critical G-protein-coupled receptor (GPCR) regulating energy homeostasis, activates multiple signalling pathways, including mobilisation of intracellular calcium ([Ca2+]i). However, very little is known about the physiological significance of MC4R-induced [Ca2+]i since few studies measure MC4R-induced [Ca2+]i. High-throughput, read-out assays for [Ca2+]i have proven unreliable for overexpressed GPCRs like MC4R, which exhibit low sensitivity mobilising [Ca2+]i. Therefore, we developed, optimised, and validated a robust quantitative high-throughput assay using Fura-2 ratio-metric calcium dye and HEK293 cells stably transfected with MC4R. The quantitation enables direct comparisons between assays and even between different research laboratories. Assay conditions were optimised step-by-step to eliminate interference from stretch-activated receptor increases in [Ca2+]i and to maximise ligand-activated MC4R-induced [Ca2+]i. Calcium imaging was performed using a PheraStar FS multi-well plate reader. Probenecid, included in the buffers to prevent extrusion of Fura-2 dye from cells, was found to interfere with the EGTA-chelation of calcium, required to determine Rmin for quantitation of [Ca2+]i. Therefore, we developed a method to determine Rmin in specific wells without probenecid, which was run in parallel with each assay. The validation of the assay was shown by reproducible α-melanocyte-stimulating hormone (α-MSH) concentration-dependent activation of the stably expressed human MC4R (hMC4R) and mouse MC4R (mMC4R), inducing increases in [Ca2+]i, for three independent experiments. This robust, reproducible, high-throughput assay that quantitatively measures MC4R-induced mobilisation of [Ca2+]i in vitro has potential to advance the development of therapeutic drugs and understanding of MC4R signalling associated with human obesity.
Collapse
Affiliation(s)
- Shree Senthil Kumar
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Marie-Louise Ward
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathleen Grace Mountjoy
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- Correspondence should be addressed to K G Mountjoy:
| |
Collapse
|
5
|
Kiss B, Némethy Z, Fazekas K, Kurkó D, Gyertyán I, Sághy K, Laszlovszky I, Farkas B, Kirschner N, Bolf-Terjéki E, Balázs O, Lendvai B. Preclinical pharmacodynamic and pharmacokinetic characterization of the major metabolites of cariprazine. Drug Des Devel Ther 2019; 13:3229-3248. [PMID: 31571826 PMCID: PMC6754336 DOI: 10.2147/dddt.s188760] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/01/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Cariprazine, a dopamine D3-preferring D3/D2 receptor partial agonist and serotonin 5-HT1A receptor partial agonist, has two major human metabolites, desmethyl-cariprazine (DCAR) and didesmethyl-cariprazine (DDCAR). The metabolite pharmacology was profiled to understand the contribution to cariprazine efficacy. METHODS In vitro receptor binding and functional assays, electrophysiology, animal models, microdialysis, and kinetic-metabolism approaches were used to characterize the pharmacology of DCAR and DDCAR. RESULTS Similar to cariprazine, both metabolites showed high affinity for human D3, D2L, 5-HT1A, 5-HT2A, and 5-HT2B receptors, albeit with higher selectivity than cariprazine for D3 versus D2 receptors. In [35S]GTPγS binding assays, cariprazine and DDCAR were antagonists in membranes from rat striatum and from cells expressing human D2 and D3 receptors, and were partial agonists in membranes from rat hippocampus. In cAMP signaling assays, cariprazine, DCAR, and DDCAR acted as partial agonists at D2 and D3 receptors; cariprazine and DDCAR were full agonists, whereas DCAR was a partial agonist at 5-HT1A receptors. Cariprazine, DCAR, and DDCAR were pure antagonists at human 5-HT2B receptors. Cariprazine and DDCAR increased rat striatal dopamine and reduced cortical serotonin turnover. Cariprazine and DDCAR showed similar in vivo D3 receptor occupancy in rat brain; however, cariprazine was more potent for D2 receptor occupancy. Both cariprazine and DDCAR dose-dependently but partially suppressed the spontaneous activity of midbrain dopaminergic neurons in rats, with the parent compound being more potent but shorter acting than its metabolite. Consistent with the D2 receptor occupancy profile, DDCAR was 3- to 10-fold less potent than cariprazine in rodent models of antipsychotic-like activity. Following acute cariprazine administration, DDCAR was detected in the rodent brain but at much lower levels than cariprazine. CONCLUSION Overall, in vitro and in vivo pharmacological profiles of DCAR and DDCAR demonstrated high similarity with cariprazine, suggesting that the major metabolites of cariprazine contribute significantly to its clinical efficacy.
Collapse
Affiliation(s)
- Béla Kiss
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Zsolt Némethy
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Károly Fazekas
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Dalma Kurkó
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - István Gyertyán
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Katalin Sághy
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - István Laszlovszky
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Bence Farkas
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Norbert Kirschner
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Etelka Bolf-Terjéki
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Ottilia Balázs
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacological and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| |
Collapse
|
6
|
Szőllősi E, Bobok A, Kiss L, Vass M, Kurkó D, Kolok S, Visegrády A, Keserű GM. Cell-based and virtual fragment screening for adrenergic α2C receptor agonists. Bioorg Med Chem 2015; 23:3991-9. [PMID: 25648685 DOI: 10.1016/j.bmc.2015.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/21/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022]
Abstract
Fragment-based drug discovery has emerged as an alternative to conventional lead identification and optimization strategies generally supported by biophysical detection techniques. Membrane targets like G protein-coupled receptors (GPCRs), however, offer challenges in lack of generic immobilization or stabilization methods for the dynamic, membrane-bound supramolecular complexes. Also modeling of different functional states of GPCRs proved to be a challenging task. Here we report a functional cell-based high concentration screening campaign for the identification of adrenergic α2C receptor agonists compared with the virtual screening of the same ligand set against an active-like homology model of the α2C receptor. The conventional calcium mobilization-based assay identified active fragments with a similar incidence to several other reported fragment screens on GPCRs. 16 out of 3071 screened fragments turned out as specific ligands of α2C, two of which were identified by virtual screening as well and several of the hits possessed surprisingly high affinity and ligand efficiency. Our results indicate that in vitro biological assays can be utilized in the fragment hit identification process for GPCR targets.
Collapse
Affiliation(s)
- Edit Szőllősi
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | - Amrita Bobok
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | - László Kiss
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | - Márton Vass
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | - Dalma Kurkó
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | - Sándor Kolok
- Gedeon Richter Plc., Gyömrői út 19-21, Budapest H-1103, Hungary
| | | | - György M Keserű
- Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Magyar tudósok körútja 2, Budapest H-1117, Hungary
| |
Collapse
|
7
|
Kurko D, Kapui Z, Nagy J, Lendvai B, Kolok S. Analysis of functional selectivity through G protein-dependent and -independent signaling pathways at the adrenergic α(2C) receptor. Brain Res Bull 2014; 107:89-101. [PMID: 25080296 DOI: 10.1016/j.brainresbull.2014.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/01/2023]
Abstract
Although G protein-coupled receptors (GPCRs) are traditionally categorized as Gs-, Gq-, or Gi/o-coupled, their signaling is regulated by multiple mechanisms. GPCRs can couple to several effector pathways, having the capacity to interact not only with more than one G protein subtype but also with alternative signaling or effector proteins such as arrestins. Moreover, GPCR ligands can have different efficacies for activating these signaling pathways, a characteristic referred to as biased agonism or functional selectivity. In this work our aim was to detect differences in the ability of various agonists acting at the α2C type of adrenergic receptors (α2C-ARs) to modulate cAMP accumulation, cytoplasmic Ca(2+) release, β-arrestin recruitment and receptor internalization. A detailed comparative pharmacological characterization of G protein-dependent and -independent signaling pathways was carried out using adrenergic agonists (norepinephrine, phenylephrine, brimonidine, BHT-920, oxymetazoline, clonidine, moxonidine, guanabenz) and antagonists (MK912, yohimbine). As initial analysis of agonist Emax and EC50 values suggested possible functional selectivity, ligand bias was quantified by applying the relative activity scale and was compared to that of the endogenous agonist norepinephrine. Values significantly different from 0 between pathways indicated an agonist that promoted different level of activation of diverse effector pathways most likely due to the stabilization of a subtly different receptor conformation from that induced by norepinephrine. Our results showed that a series of agonists acting at the α2C-AR displayed different degree of functional selectivity (bias factors ranging from 1.6 to 36.7) through four signaling pathways. As signaling via these pathways seems to have distinct functional and physiological outcomes, studying all these stages of receptor activation could have further implications for the development of more selective therapeutics with improved efficacy and/or fewer side effects.
Collapse
Affiliation(s)
- Dalma Kurko
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary.
| | - Zoltán Kapui
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - József Nagy
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Balázs Lendvai
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Sándor Kolok
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| |
Collapse
|
8
|
Levay M, Krobert KA, Wittig K, Voigt N, Bermudez M, Wolber G, Dobrev D, Levy FO, Wieland T. NSC23766, a Widely Used Inhibitor of Rac1 Activation, Additionally Acts as a Competitive Antagonist at Muscarinic Acetylcholine Receptors. J Pharmacol Exp Ther 2013; 347:69-79. [DOI: 10.1124/jpet.113.207266] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
9
|
Camarda V, Calo' G. Chimeric G proteins in fluorimetric calcium assays: experience with opioid receptors. Methods Mol Biol 2013; 937:293-306. [PMID: 23007594 DOI: 10.1007/978-1-62703-086-1_18] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High throughput calcium mobilization assays are extensively used for pharmacological characterization of GPCR ligands. These approaches, initially developed for G(q)-coupled receptors, can be extended to G(i) coupled GPCRs using chimeric G proteins. Here we used the Gα(qi5) protein to force the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor, as well as the classical opioid receptors to signal through the PLC-IP(3)-Ca(2+) pathway in CHO cells. Calcium levels were monitored using the fluorometric imaging plate reader FlexStation II and the Ca(2+) dye Fluo 4 AM. For investigating the pharmacology of the NOP receptor a panel of full and partial agonists and antagonists were assessed, while a small panel of agonists and antagonists was used for evaluating the pharmacological profile of opioid receptors. Some limitations of this assay and differences in the results obtained in comparison with those with G(i) based biochemical assays are described. Overall, the present results confirm that the chimeric G protein strategy is useful for studying the pharmacological activity of G(i) coupled receptor ligands and that the aberrant signaling does not produce any measurable change in the pharmacological profile of the receptor under study. Thus, this G protein strategy is extremely useful for setting up primary screening assays for NOP and classical opioid receptors and likely for other members of the GPCR family.
Collapse
Affiliation(s)
- Valeria Camarda
- Department of Experimental and Clinical Medicine, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|