1
|
Magnussen J. Advances in PET Imaging of α7 Nicotinic Receptors: From Radioligand Development to CNS Applications. Basic Clin Pharmacol Toxicol 2025; 136:e70025. [PMID: 40084546 PMCID: PMC11907392 DOI: 10.1111/bcpt.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/21/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Positron emission tomography (PET) has significantly advanced our understanding of the brain by enabling non-invasive imaging and quantification of molecular processes, including receptor binding. In this review, we explore the development and application of PET radioligands targeting the α7 nicotinic acetylcholine receptor (α7 nAChR), a receptor implicated in various central nervous system (CNS) diseases, such as Alzheimer's disease, schizophrenia and cognitive disorders. Despite challenges associated with the low density of α7 nAChRs and difficulties in achieving adequate brain penetration, several promising radioligands have been developed, including 11C-(R)-MeQAA, 11C-NS14492 and 18F-ASEM. These radioligands facilitate the evaluation of the 'three pillars of survival' in drug development: tissue accessibility, target engagement and downstream pharmacology. PET imaging offers critical insights into drug distribution across the blood-brain barrier, receptor occupancy and the pharmacodynamic effects of α7 nAChR-targeted therapies. By reviewing current radioligands and their applications, we highlight the potential of PET imaging to deepen our understanding of α7 nAChR-mediated signalling pathways and its implications for CNS drug discovery. Future innovations in radioligand development, including more selective and brain-penetrant compounds, will be key to fully realizing the potential of PET imaging in α7 nAChR-targeted research and treatment.
Collapse
|
2
|
Sparatore F, Sparatore A. 3,3-Disubstituted 3,4-Dihydro-1,2,4-benzotriazines: Chemistry, Biological Activity, and Affinity to Sigma Receptors. Molecules 2023; 29:132. [PMID: 38202715 PMCID: PMC10780181 DOI: 10.3390/molecules29010132] [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] [Received: 11/14/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
By reducing the 2-nitrophenylhydrazone of cyclohexanone with sodium dithionite, an unexpected yellow compound was obtained instead of the corresponding colorless amino derivative. Many years later, the structure of this compound, namely, cyclohexane-3-spiro-3,4-dihydro-1,2,4-benzotriazine, was demonstrated. From that time, the reduction of 2-nitrophenylhydrazones of different kinds of ketones, followed by air oxidation of the initially formed amino compounds, has represented a general way to synthesize a variety of 3,3-disubstituted 3,4-dihydro-1,2,4-benzotriazines. Many derivatives have been obtained so far by a single research group, and most of them have demonstrated interesting pharmacological activities, mainly antihypertensive, anti-inflammatory, and diuretic effects and other activities with lower diffusion. Moreover, 3,3-disubstituted 3,4-dihydro-1,2,4-benzotriazines represent a novel class of ligands for sigma receptors, with nanomolar affinity to the σ1 subtype. This property might promote the development of agents for cardiovascular, neurodegenerative, and proliferative pathologies. The present commentary, by collecting compounds and biological results obtained so far, intends to celebrate the centennial of the discovery of the first member of this class of compounds and to promote further investigation in the field.
Collapse
Affiliation(s)
- Fabio Sparatore
- Department of Pharmacy, University of Genova, 16132 Genova, Italy
| | - Anna Sparatore
- Department of Pharmaceutical Sciences (DISFARM), University of Milano, 20133 Milano, Italy;
| |
Collapse
|
3
|
Zhang JJ, Fu H, Lin R, Zhou J, Haider A, Fang W, Elghazawy NH, Rong J, Chen J, Li Y, Ran C, Collier TL, Chen Z, Liang SH. Imaging Cholinergic Receptors in the Brain by Positron Emission Tomography. J Med Chem 2023; 66:10889-10916. [PMID: 37583063 PMCID: PMC10461233 DOI: 10.1021/acs.jmedchem.3c00573] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 08/17/2023]
Abstract
Cholinergic receptors represent a promising class of diagnostic and therapeutic targets due to their significant involvement in cognitive decline associated with neurological disorders and neurodegenerative diseases as well as cardiovascular impairment. Positron emission tomography (PET) is a noninvasive molecular imaging tool that has helped to shed light on the roles these receptors play in disease development and their diverse functions throughout the central nervous system (CNS). In recent years, there has been a notable advancement in the development of PET probes targeting cholinergic receptors. The purpose of this review is to provide a comprehensive overview of the recent progress in the development of these PET probes for cholinergic receptors with a specific focus on ligand structure, radiochemistry, and pharmacology as well as in vivo performance and applications in neuroimaging. The review covers the structural design, pharmacological properties, radiosynthesis approaches, and preclinical and clinical evaluations of current state-of-the-art PET probes for cholinergic receptors.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Hualong Fu
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ruofan Lin
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ahmed Haider
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Weiwei Fang
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Nehal H. Elghazawy
- Department
of Pharmaceutical, Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, 11835 Cairo, Egypt
| | - Jian Rong
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jiahui Chen
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Yinlong Li
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chongzhao Ran
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02114, United States
| | - Thomas L. Collier
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Zhen Chen
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Steven H. Liang
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
4
|
Ouach A, Vercouillie J, Bertrand E, Rodrigues N, Pin F, Serriere S, Boiaryna L, Chartier A, Percina N, Tangpong P, Gulhan Z, Mothes C, Deloye JB, Guilloteau D, Page G, Suzenet F, Buron F, Chalon S, Routier S. Bis(het)aryl-1,2,3-triazole quinuclidines as α7 nicotinic acetylcholine receptor ligands: Synthesis, structure affinity relationships, agonism activity, [18F]-radiolabeling and PET study in rats. Eur J Med Chem 2019; 179:449-469. [DOI: 10.1016/j.ejmech.2019.06.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
|
5
|
Synthesis, biological evaluation and structure-activity relationships of self-assembled and solubilization properties of amphiphilic quaternary ammonium derivatives of quinuclidine. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
6
|
Potential of α7 nicotinic acetylcholine receptor PET imaging in atherosclerosis. Methods 2017; 130:90-104. [PMID: 28602809 DOI: 10.1016/j.ymeth.2017.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic events are usually acute and often strike otherwise asymptomatic patients. Although multiple clinical risk factors have been associated with atherosclerosis, as of yet no further individual prediction can be made as to who will suffer from its consequences based on biomarker analysis or traditional imaging methods like CT, MRI or angiography. Previously, non-invasive imaging with 18F-fluorodeoxyglucose (18F-FDG) PET was shown to potentially fill this niche as it offers high sensitive detection of metabolic processes associated with inflammatory changes in atherosclerotic plaques. However, 18F-FDG PET imaging of arterial vessels suffers from non-specificity and has still to be proven to reliably identify vulnerable plaques, carrying a high risk of rupture. Therefore, it may be regarded only as a secondary marker for monitoring treatment effects and it does not offer alternative treatment options or direct insight in treatment mechanisms. In this review, an overview is given of the current status and the potential of PET imaging of inflammation and angiogenesis in atherosclerosis in general and special emphasis is given to imaging of α7 nicotinic acetylcholine receptors (α7 nAChRs). Due to the gaps that still exist in our understanding of atherogenesis and the limitations of the available PET tracers, the search continues for a more specific radioligand, able to differentiate between stable atherosclerosis and plaques prone to rupture. The potential role of the α7 nAChR as imaging marker for plaque vulnerability is explored. Today, strong evidence exists that nAChRs are involved in the atherosclerotic disease process. They are suggested to mediate the deleterious effects of the major tobacco component, nicotine, a nAChR agonist. Mainly based on in vitro data, α7 nAChR stimulation might increase plaque burden via increased neovascularization. However, in animal studies, α7 nAChR manipulation appears to reduce plaque size due to its inhibitory effects on inflammatory cells. Thus, reliable identification of α7 nAChRs by in vivo imaging is crucial to investigate the exact role of α7 nAChR in atherosclerosis before any therapeutic approach in the human setting can be justified. In this review, we discuss the first experience with α7 nAChR PET tracers and developmental considerations regarding the "optimal" PET tracer to image vascular nAChRs.
Collapse
|
7
|
Kassenbrock A, Vasdev N, Liang SH. Selected PET Radioligands for Ion Channel Linked Neuroreceptor Imaging: Focus on GABA, NMDA and nACh Receptors. Curr Top Med Chem 2017; 16:1830-42. [PMID: 26975506 DOI: 10.2174/1568026616666160315142457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) neuroimaging of ion channel linked receptors is a developing area of preclinical and clinical research. The present review focuses on recent advances with radiochemistry, preclinical and clinical PET imaging studies of three receptors that are actively pursued in neuropsychiatric drug discovery: namely the γ-aminobutyric acid-benzodiazapine (GABA) receptor, nicotinic acetylcholine receptor (nAChR), and N-methyl-D-aspartate (NMDA) receptor. Recent efforts to develop new PET radioligands for these targets with improved brain uptake, selectivity, stability and pharmacokinetics are highlighted.
Collapse
Affiliation(s)
| | | | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Sarasamkan J, Scheunemann M, Apaijai N, Palee S, Parichatikanond W, Arunrungvichian K, Fischer S, Chattipakorn S, Deuther-Conrad W, Schüürmann G, Brust P, Vajragupta O. Varying Chirality Across Nicotinic Acetylcholine Receptor Subtypes: Selective Binding of Quinuclidine Triazole Compounds. ACS Med Chem Lett 2016; 7:890-895. [PMID: 27774124 DOI: 10.1021/acsmedchemlett.6b00146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/09/2016] [Indexed: 12/16/2022] Open
Abstract
The novel quinuclidine anti-1,2,3-triazole derivatives T1-T6 were designed based on the structure of QND8. The binding studies revealed that the stereochemistry at the C3 position of the quinuclidine scaffold plays an important role in the nAChR subtype selectivity. Whereas the (R)-enantiomers are selective to α7 over α4β2 (by factors of 44-225) and to a smaller degree over α3β4 (3-33), their (S)-counterparts prefer α3β4 over α4β2 (62-237) as well as over α7 (5-294). The (R)-derivatives were highly selective to α7 over α3β4 subtypes compared to (RS)- and (R)-QND8. The (S)-enantiomers are 5-10 times more selective to α4β2 than their (R) forms. The overall strongest affinity is observed for the (S)-enantiomer binding to α3β4 (Ki, 2.25-19.5 nM) followed by their (R)-counterpart binding to α7 (Ki, 22.5-117 nM), with a significantly weaker (S)-enantiomer binding to α4β2 (Ki, 414-1980 nM) still above the very weak respective (R)-analogue affinity (Ki, 5059-10436 nM).
Collapse
Affiliation(s)
- Jiradanai Sarasamkan
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Road, Bangkok 10400, Thailand
- Department
of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical
Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße15, 04318 Leipzig, Germany
- National
Cyclotron and PET Centre, Chulabhorn Hospital, 54 Kamphaengphet 6 Road, Bangkok 10210, Thailand
| | - Matthias Scheunemann
- Department
of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical
Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße15, 04318 Leipzig, Germany
| | - Nattayaporn Apaijai
- Neurophysiology
Unit, Cardiac Electrophysiology Research and Training Center, Faculty
of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siripong Palee
- Neurophysiology
Unit, Cardiac Electrophysiology Research and Training Center, Faculty
of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Warisara Parichatikanond
- Department
of Pharmacology, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayutthaya
Road, Bangkok 10400, Thailand
| | - Kuntarat Arunrungvichian
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Road, Bangkok 10400, Thailand
| | - Steffen Fischer
- Department
of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical
Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße15, 04318 Leipzig, Germany
| | - Siriporn Chattipakorn
- Neurophysiology
Unit, Cardiac Electrophysiology Research and Training Center, Faculty
of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Winnie Deuther-Conrad
- Department
of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical
Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße15, 04318 Leipzig, Germany
| | - Gerrit Schüürmann
- Department
of Ecological Chemistry, Helmholtz Centre for Environmental Research−UFZ, Permoserstraße15, 04318 Leipzig, Germany
- Institute
for Organic Chemistry, Technical University Bergakademie Freiberg, Leipziger Straße29, 09596 Freiberg, Germany
| | - Peter Brust
- Department
of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical
Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Permoserstraße15, 04318 Leipzig, Germany
| | - Opa Vajragupta
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayutthaya Road, Bangkok 10400, Thailand
| |
Collapse
|
9
|
Ouach A, Pin F, Bertrand E, Vercouillie J, Gulhan Z, Mothes C, Deloye JB, Guilloteau D, Suzenet F, Chalon S, Routier S. Design of α7 nicotinic acetylcholine receptor ligands using the (het)Aryl-1,2,3-triazole core: Synthesis, in vitro evaluation and SAR studies. Eur J Med Chem 2016; 107:153-64. [DOI: 10.1016/j.ejmech.2015.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 10/22/2022]
|
10
|
López JJ, Pérez EG, García-Colunga J. Dual effects of a 2-benzylquinuclidinium derivative on α7-containing nicotinic acetylcholine receptors in rat hippocampal interneurons. Neurosci Lett 2015; 607:35-39. [PMID: 26384784 DOI: 10.1016/j.neulet.2015.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 11/18/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are widely distributed in the brain. Particularly α7-containing nAChRs, associated with several physiological roles and pathologies, are one of the most abundant. Here, we studied 2-(4-hexyloxybenzyl)-1-methylquinuclidin-1-ium iodide (designated as 8d), on ion currents elicited by choline, ICh, (Ch, a selective agonist for α7-containing nAChRs), recorded in interneurons from the stratum radiatum of the rat hippocampal CA1 region by using the whole-cell voltage-clamp technique. The 8d-concentration/Ch-response relationship exhibited high and low inhibitory affinities for α7-containing nAChRs, with IC50 values of 0.59 and 6.80 μM, respectively. Interestingly, 8d in a range of 3-10 μM exerted opposite effects: a short early potentiation and a long late inhibition of the ICh; and 8d alone elicited a non-decaying inward current. Furthermore, potentiation and inhibition of the ICh by 8d depended on the membrane potential, both being stronger at -20 than at -70 mV; indicating that 8d interacts with at least two sites into the ion channel/receptor complex: one for potentiating and another for inhibiting the α7-containing nAChRs. These results suggest that 8d may act as agonist, antagonist and positive modulator of α7-containing nAChRs in hippocampal interneurons.
Collapse
Affiliation(s)
- Jhon J López
- Department of Organic Chemistry, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Casilla 306, Correo 22, Santiago, Chile
| | - Edwin G Pérez
- Department of Organic Chemistry, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Casilla 306, Correo 22, Santiago, Chile.
| | - Jesús García-Colunga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Querétaro 76230, Mexico.
| |
Collapse
|
11
|
Bourdin CM, Lebreton J, Mathé-Allainmat M, Thany SH. Pharmacological profile of zacopride and new quaternarized fluorobenzamide analogues on mammalian α7 nicotinic acetylcholine receptor. Bioorg Med Chem Lett 2015; 25:3184-8. [PMID: 26087938 DOI: 10.1016/j.bmcl.2015.05.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/14/2015] [Accepted: 05/29/2015] [Indexed: 11/26/2022]
Abstract
From quaternarization of quinuclidine enantiomers of 2-fluoro benzamide LMA10203 in dichloromethane, the corresponding N-chloromethyl derivatives LMA10227 and LMA10228 were obtained. Here, we compared the agonist action of known zacopride and its 2-fluoro benzamide analogues, LMA10203, LMA10227 and LMA10228 against mammalian homomeric α7 nicotinic acetylcholine receptor expressed in Xenopus oocytes. We found that LMA10203 was a partial agonist of α7 receptor with a pEC50 value of 4.25 ± 0.06 μM whereas LMA10227 and LMA10228 were poorly active on α7 homomeric nicotinic receptor. LMA10227 and LMA10228 were identified as antagonists of acetylcholine-induced currents with IC50 values of 28.4 μM and 39.3 μM whereas LMA10203 and zacopride possessed IC50 values of 8.07 μM and 7.04 μM, respectively. Moreover, despite their IC50 values, LMA10227 was the most potent inhibitor of nicotine-induced current amplitudes (65.7 ± 2.1% inhibition). LMA10203 and LMA10228 had the same inhibitory effects (26.5 ± 7.5% and 33.2 ± 4.1%, respectively), whereas zacopride had no significant inhibitory effect (4.37 ± 4%) on nicotine-induced responses. Our results revealed different pharmacological properties between the four compounds on acetylcholine and nicotine currents. The mode of action of benzamide compounds may need to be reinterpreted with respect to the potential role of α7 receptor.
Collapse
Affiliation(s)
- Céline M Bourdin
- Université d'Orléans, Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC) UPRES EA 1207, Rue de Chartres, BP 6759, 45067 Orléans, France; Université d'Angers, RCIM UPRES EA 2647 US INRA 1330, 2 rue Lavoisier, 49045 Angers, France
| | - Jacques Lebreton
- Université de Nantes, CEISAM UMR CNRS 6230, UFR des Sciences et des Techniques, 2 rue de la Houssinière, BP 92208, Nantes 44322, France
| | - Monique Mathé-Allainmat
- Université de Nantes, CEISAM UMR CNRS 6230, UFR des Sciences et des Techniques, 2 rue de la Houssinière, BP 92208, Nantes 44322, France
| | - Steeve H Thany
- Université d'Orléans, Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC) UPRES EA 1207, Rue de Chartres, BP 6759, 45067 Orléans, France.
| |
Collapse
|
12
|
Arunrungvichian K, Boonyarat C, Fokin VV, Taylor P, Vajragupta O. Cognitive Improvements in a Mouse Model with Substituted 1,2,3-Triazole Agonists for Nicotinic Acetylcholine Receptors. ACS Chem Neurosci 2015; 6:1331-40. [DOI: 10.1021/acschemneuro.5b00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Kuntarat Arunrungvichian
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayudhya Road, Bangkok 10400, Thailand
| | - Chantana Boonyarat
- Department
of Pharmaceutical Chemistry, Faculty of Pharmaceutical Science, KhonKaen University, 123 Muang, KhonKaen 40002, Thailand
| | - Valery V. Fokin
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Palmer Taylor
- Department
of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0650, United States
| | - Opa Vajragupta
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayudhya Road, Bangkok 10400, Thailand
| |
Collapse
|
13
|
Arunrungvichian K, Fokin VV, Vajragupta O, Taylor P. Selectivity Optimization of Substituted 1,2,3-Triazoles as α7 Nicotinic Acetylcholine Receptor Agonists. ACS Chem Neurosci 2015; 6:1317-30. [DOI: 10.1021/acschemneuro.5b00058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Kuntarat Arunrungvichian
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayudhya Road, Bangkok 10400, Thailand
- Department
of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0650, United States
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Valery V. Fokin
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Opa Vajragupta
- Center
of Excellence for Innovation in Drug Design and Discovery, Faculty
of Pharmacy, Mahidol University, 447 Sri-Ayudhya Road, Bangkok 10400, Thailand
| | - Palmer Taylor
- Department
of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0650, United States
| |
Collapse
|