1
|
Courault P, Mérida I, Costes N, Redoute J, Zimmer L, Lancelot S. PET imaging of functional 5-HT 1A receptors with [ 18F]F13640: From PET kinetics modeling to static Standardized Uptake Values Ratio. Neuroimage 2025; 310:121110. [PMID: 40064318 DOI: 10.1016/j.neuroimage.2025.121110] [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: 06/05/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE F13640 is a highly selective serotonin 5-HT1A receptor ligand with agonist properties identified as a PET radiopharmaceutical candidate. In previous work, we showed the possibility to use long dynamic PET acquisition (225 min) combined with simplified kinetic modelling for [18F]F13640 quantification. In this work, we assessed the feasibility of static acquisition and quantification using standardized uptake value ratio (SUVR) as an alternative. METHODS Test-retest PET-MRI scans of 225 min were conducted in eight healthy male volunteers. For 17 brain regions, distribution volume ratios (DVR) were calculated from the whole kinetics using Logan plot modelling method with the cerebellum white matter as reference region. For the same regions, SUVR were also calculated from static images, for four 20-minute and four 10-minute time-intervals at various time of uptake. Reliability between SUVR and DVR measures were studied, and test-retest parameters were assessed between PET sessions for each time-interval. RESULTS Reproducibility of measures of SUVR compared to DVR were excellent, whatever the time interval (p < 0.001). For the 20-min. time-intervals, SUVR150-170 showed the best reproducibility and correlation parameters (R2 = 0.95 ± 0.03, intercept = 0.06 ± 0.02, slope = 0.95 ± 0.01). As well, for the 10-min. time-intervals, SUVR150-160 showed the best correlation (R2 = 0.94 ± 0.03, intercept = 0.07 ± 0.02, slope = 0.94 ± 0.01). SUVR reproducibility between test-retest sessions was also excellent for each time-interval. These results were valid for pooled regions as well as at regional level. CONCLUSION This study confirms the feasibility of static acquisitions to facilitate clinical use of the [18F]F13640 radiopharmaceutical to image functional 5-HT1A receptors. This involves off-camera injection, 10 to 20 mins static acquisition duration, and quantification using SUVR, while improving patient comfort.
Collapse
Affiliation(s)
- Pierre Courault
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRS, Université Claude Bernard Lyon1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP, Lyon, France.
| | | | - Nicolas Costes
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRS, Université Claude Bernard Lyon1, Lyon, France; CERMEP, Lyon, France
| | | | - Luc Zimmer
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRS, Université Claude Bernard Lyon1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP, Lyon, France
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), INSERM, CNRS, Université Claude Bernard Lyon1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP, Lyon, France
| |
Collapse
|
2
|
Zimmer L, Newman-Tancredi A. Serotonin 5-HT 1A receptor biased agonists: The challenge of translating an innovative neuropharmacological concept into therapeutics. Neuropharmacology 2025; 265:110267. [PMID: 39681214 DOI: 10.1016/j.neuropharm.2024.110267] [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: 09/06/2024] [Revised: 10/24/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Serotonin 5-HT1A receptor agonists are prime candidates for CNS drug discovery due to their involvement physiological and pathological processes relevant to neurology and psychiatry. However, the lack of target specificity of many previously characterized agonists has long been a barrier to pharmacological and therapeutic progress. Some of the obstacles may be overcome through the recent concept of biased agonism, which has attracted considerable attention to the development of novel chemical entities at 5-HT, and particularly 5-HT1A receptors, by specifically targeting intracellular signalling pathways that may themselves be linked to specific brain regions and therapeutic indications. There is now abundant translational data demonstrating distinct molecular and functional pharmacological signatures between different 5-HT1A receptor agonists, opening new opportunities for research in neurology and psychiatry. Nevertheless, important limitations need to be overcome, including understanding the precise molecular basis for biased agonism, the need for improved translatable models, and the currently limited clinical data on biased agonists. Here, we review the current limits of our knowledge of 5-HT1A receptor biased agonists and the limitations of available pharmacological tools, counterbalanced by the translational possibilities and therapeutic perspectives opened by novel, highly selective 5-HT1A receptor drug-candidates. This article is part of the Special Issue on "Ligand Bias".
Collapse
Affiliation(s)
- Luc Zimmer
- Université Claude Bernard Lyon 1, Inserm, CNRS, Lyon Neuroscience Research Center, Lyon, France; Hospices Civils de Lyon, Lyon, France.
| | | |
Collapse
|
3
|
Soyer A, Leterrier S, Goutal S, Corvo C, Saba W, Caillé F, Bo GD, Winkeler A, Thibault K, Leroy C, Tournier N. Decreased opioid receptor availability and impaired neurometabolic coupling as signatures of morphine tolerance in male rats: A positron emission tomography study. Biomed Pharmacother 2025; 183:117848. [PMID: 39823723 DOI: 10.1016/j.biopha.2025.117848] [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: 11/06/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
Translational neuroimaging techniques are needed to address the impact of opioid tolerance on brain function and quantitatively monitor the impaired neuropharmacological response to opioids at the CNS level. A multiparametric PET study was conducted in rats. Rats received morphine daily to induce tolerance (15 mg/kg/day for 5 days), followed by 2-day withdrawal. Then, opioid effects were precipitated using a buprenorphine challenge (0.1 mg/kg, s.c, BUP-challenge), which safely enables full occupancy of available mu-opioid receptors (MOR). The impact of the BUP-challenge on the pain threshold was estimated using the hot-plate test. The corresponding availability of MOR was estimated using [11C]buprenorphine PET imaging (n = 4). The brain glucose metabolism was investigated using [18F]2-fluoro-D-deoxy-glucose ([18F]FDG) PET imaging after the BUP-challenge or saline (n = 5-6). Opioid tolerance was confirmed by the attenuated antinociceptive response to the BUP-challenge in morphine-treated rats compared to saline controls (p < 0.001). In tolerant rats, [11C]buprenorphine binding was decreased in MOR-rich regions (p < 0.01), and the baseline uptake of [18F]FDG was decreased (p < 0.05). The BUP-challenge decreased [18F]FDG uptake to a lower extent in tolerant rats compared with opioid-naive animals (p < 0.05), suggesting impaired neurometabolic coupling. Moreover, the impact of the BUP-challenge on the neurometabolic connectivity across brain regions was disrupted by opioid tolerance. PET imaging enables the study of the decreased availability of MOR and impaired neurometabolic coupling as molecular signatures of opioid tolerance in rats. Combining molecular neuroimaging with a suitable pharmacological challenge may provide a translational and quantitative paradigm to explore opioid tolerance at the CNS level in parallel to pharmacodynamics.
Collapse
Affiliation(s)
- Amélie Soyer
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sarah Leterrier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sébastien Goutal
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Cassandre Corvo
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Wadad Saba
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Fabien Caillé
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Gregory Dal Bo
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France; Department of Radiation Biological Effects, Armed Forces Biomedical Research Institute, Bretigny-sur-Orge 91220, France
| | - Alexandra Winkeler
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Karine Thibault
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France; Department of Radiation Biological Effects, Armed Forces Biomedical Research Institute, Bretigny-sur-Orge 91220, France
| | - Claire Leroy
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
4
|
Gündel D, Maqbool M, Teodoro R, Ludwig FA, Heerklotz A, Toussaint M, Deuther-Conrad W, Bormans G, Brust P, Kopka K, Moldovan RP. Development and evaluation of deuterated [ 18F]JHU94620 isotopologues for the non-invasive assessment of the cannabinoid type 2 receptor in brain. EJNMMI Radiopharm Chem 2024; 9:91. [PMID: 39714717 DOI: 10.1186/s41181-024-00319-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 11/29/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND The cannabinoid type 2 receptors (CB2R) represent a target of increasing importance in neuroimaging due to its upregulation under various neuropathological conditions. Previous evaluation of [18F]JHU94620 for the non-invasive assessment of the CB2R availability by positron emission tomography (PET) revealed favourable binding properties and brain uptake, however rapid metabolism, and generation of brain-penetrating radiometabolites have been its main limitations. To reduce the bias of CB2R quantification by blood-brain barrier (BBB)-penetrating radiometabolites, we aimed to improve the metabolic stability by developing -d4 and -d8 deuterated isotopologues of [18F]JHU94620. RESULTS The deuterated [18F]JHU94620 isotopologues showed improved metabolic stability avoiding the accumulation of BBB-penetrating radiometabolites in the brain over time. CB2R-specific binding with KD values in the low nanomolar range was determined across species. Dynamic PET studies revealed a CB2R-specific and reversible uptake of [18F]JHU94620-d8 in the spleen and to a local hCB2R(D80N) protein overexpression in the striatal region in rats. CONCLUSION These results support further investigations of [18F]JHU94620-d8 in pathological models and tissues with a CB2R overexpression as a prerequisite for clinical translation.
Collapse
Affiliation(s)
- Daniel Gündel
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany.
| | - Mudasir Maqbool
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892-1026, USA
| | - Rodrigo Teodoro
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
- Life Molecular Imaging GmbH, 13353, Berlin, Germany
| | - Friedrich-Alexander Ludwig
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
| | - Anne Heerklotz
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
| | - Magali Toussaint
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
| | - Winnie Deuther-Conrad
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, BE-3000, Leuven, Belgium
| | - Peter Brust
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
- The Lübeck Institute of Experimental Dermatology, University Medical Center Schleswig-Holstein, 23562, Lübeck, Germany
| | - Klaus Kopka
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, TU Dresden, 01069, Dresden, Germany
| | - Rareş-Petru Moldovan
- Department of Experimental Neurooncological Radiopharmacy, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Research Site Leipzig, Permoserstrasse 15, 04318, Leipzig, Germany.
| |
Collapse
|
5
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
6
|
Zimmer L. Recent applications of positron emission tomographic (PET) imaging in psychiatric drug discovery. Expert Opin Drug Discov 2024; 19:161-172. [PMID: 37948046 DOI: 10.1080/17460441.2023.2278635] [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: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Psychiatry is one of the medical disciplines that suffers most from a lack of innovation in its therapeutic arsenal. Many failures in drug candidate trials can be explained by pharmacological properties that have been poorly assessed upstream, in terms of brain passage, brain target binding and clinical outcomes. Positron emission tomography can provide pharmacokinetic and pharmacodynamic data to help select candidate-molecules for further clinical trials. AREAS COVERED This review aims to explain and discuss the various methods using positron-emitting radiolabeled molecules to trace the cerebral distribution of the drug-candidate or indirectly measure binding to its therapeutic target. More than an exhaustive review of PET studies in psychopharmacology, this article highlights the contributions this technology can make in drug discovery applied to psychiatry. EXPERT OPINION PET neuroimaging is the only technological approach that can, in vivo in humans, measure cerebral delivery of a drug candidate, percentage and duration of target binding, and even the pharmacological effects. PET studies in a small number of subjects in the early stages of the development of a psychotropic drug can therefore provide the pharmacokinetic/pharmacodynamic data required for subsequent clinical evaluation. While PET technology is demanding in terms of radiochemical, radiopharmacological and nuclear medicine expertise, its integration into the development process of new drugs for psychiatry has great added value.
Collapse
Affiliation(s)
- Luc Zimmer
- Lyon Neuroscience Research Center, Université Claude Bernard, Lyon, France
- CERMEP, Hospices Civils de Lyon, Lyon, France
- Institut National des Sciences et Technologies Nucléaire, Saclay, France
| |
Collapse
|
7
|
Millard M, Kilian J, Ozenil M, Mogeritsch M, Schwingenschlögl-Maisetschläger V, Holzer W, Hacker M, Langer T, Pichler V. Design, synthesis and preclinical evaluation of muscarine receptor antagonists via a scaffold-hopping approach. Eur J Med Chem 2023; 262:115891. [PMID: 37897926 DOI: 10.1016/j.ejmech.2023.115891] [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: 09/01/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
Our research group recently identified a rearrangement product of pirenzepine as starting point for a comprehensive rational drug design approach towards orthosteric muscarinic acetylcholine receptor ligands. Chemical reduction and bioscaffold hop lead to the development of sixteen promising compounds featuring either a benzimidazole or carbamate moiety, all exhibiting comparable pharmacophoric characteristics. The synthesized compounds were characterized by NMR, HR-MS, and RP-HPLC techniques. Subsequent evaluation encompassed binding affinity assessment on CHO-hM1-5 cells, mode of action determination, and analysis of physico-chemical parameters. The CNS MPO score indicated favorable drug-like attributes and potential CNS activity for the antagonistic ligands. The most promising compounds displayed Ki-values within a desirable low nanomolar range, and their structural features allow for potential carbon-11 radiolabeling. Our optimization efforts resulted in compounds with a remarkable 138-fold increase in binding affinity compared to the previously mentioned rearrangement product towards human M5, suggesting their prospective utility in positron emission tomography applications.
Collapse
Affiliation(s)
- Marlon Millard
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Jonas Kilian
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, Vienna, Austria
| | - Marius Ozenil
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Mariella Mogeritsch
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Verena Schwingenschlögl-Maisetschläger
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, Vienna, Austria
| | - Wolfgang Holzer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Verena Pichler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Arjmand S, Bender D, Jakobsen S, Wegener G, Landau AM. Peering into the Brain's Estrogen Receptors: PET Tracers for Visualization of Nuclear and Extranuclear Estrogen Receptors in Brain Disorders. Biomolecules 2023; 13:1405. [PMID: 37759805 PMCID: PMC10526964 DOI: 10.3390/biom13091405] [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: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogen receptors (ERs) play a multitude of roles in brain function and are implicated in various brain disorders. The use of positron emission tomography (PET) tracers for the visualization of ERs' intricate landscape has shown promise in oncology but remains limited in the context of brain disorders. Despite recent progress in the identification and development of more selective ligands for various ERs subtypes, further optimization is necessary to enable the reliable and efficient imaging of these receptors. In this perspective, we briefly touch upon the significance of estrogen signaling in the brain and raise the setbacks associated with the development of PET tracers for identification of specific ERs subtypes in the brain. We then propose avenues for developing efficient PET tracers to non-invasively study the dynamics of ERs in the brain, as well as neuropsychiatric diseases associated with their malfunction in a longitudinal manner. This perspective puts several potential candidates on the table and highlights the unmet needs and areas requiring further research to unlock the full potential of PET tracers for ERs imaging, ultimately aiding in deepening our understanding of ERs and forging new avenues for potential therapeutic strategies.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Dirk Bender
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
| | - Anne M. Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark;
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark; (D.B.); (S.J.)
| |
Collapse
|
9
|
Sadee W. Ligand-Free Signaling of G-Protein-Coupled Receptors: Physiology, Pharmacology, and Genetics. Molecules 2023; 28:6375. [PMID: 37687205 PMCID: PMC10489045 DOI: 10.3390/molecules28176375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are ubiquitous sensors and regulators of cellular functions. Each GPCR exists in complex aggregates with multiple resting and active conformations. Designed to detect weak stimuli, GPCRs can also activate spontaneously, resulting in basal ligand-free signaling. Agonists trigger a cascade of events leading to an activated agonist-receptor G-protein complex with high agonist affinity. However, the ensuing signaling process can further remodel the receptor complex to reduce agonist affinity, causing rapid ligand dissociation. The acutely activated ligand-free receptor can continue signaling, as proposed for rhodopsin and μ opioid receptors, resulting in robust receptor activation at low agonist occupancy with enhanced agonist potency. Continued receptor stimulation can further modify the receptor complex, regulating sustained ligand-free signaling-proposed to play a role in opioid dependence. Basal, acutely agonist-triggered, and sustained elevated ligand-free signaling could each have distinct functions, reflecting multi-state conformations of GPCRs. This review addresses basal and stimulus-activated ligand-free signaling, its regulation, genetic factors, and pharmacological implications, focusing on opioid and serotonin receptors, and the growth hormone secretagogue receptor (GHSR). The hypothesis is proposed that ligand-free signaling of 5-HT2A receptors mediate therapeutic effects of psychedelic drugs. Research avenues are suggested to close the gaps in our knowledge of ligand-free GPCR signaling.
Collapse
Affiliation(s)
- Wolfgang Sadee
- Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- Aether Therapeutics Inc., Austin, TX 78756, USA
| |
Collapse
|
10
|
Pakula RJ, Scott PJH. Applications of radiolabeled antibodies in neuroscience and neuro-oncology. J Labelled Comp Radiopharm 2023; 66:269-285. [PMID: 37322805 DOI: 10.1002/jlcr.4049] [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: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Positron emission tomography (PET) is a powerful tool in medicine and drug development, allowing for non-invasive imaging and quantitation of biological processes in live organisms. Targets are often probed with small molecules, but antibody-based PET is expanding because of many benefits, including ease of design of new antibodies toward targets, as well as the very strong affinities that can be expected. Application of antibodies to PET imaging of targets in the central nervous system (CNS) is a particularly nascent field, but one with tremendous potential. In this review, we discuss the growth of PET in imaging of CNS targets, present the promises and progress in antibody-based CNS PET, explore challenges faced by the field, and discuss questions that this promising approach will need to answer moving forward for imaging and perhaps even radiotherapy.
Collapse
Affiliation(s)
- Ryan J Pakula
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Saumell-Esnaola M, Elejaga-Jimeno A, Echeazarra L, Borrega-Román L, Barrondo S, López de Jesús M, González-Burguera I, Gómez-Caballero A, Goicolea MA, Sallés J, García del Caño G. Design and validation of recombinant protein standards for quantitative Western blot analysis of cannabinoid CB1 receptor density in cell membranes: an alternative to radioligand binding methods. Microb Cell Fact 2022; 21:192. [PMID: 36109736 PMCID: PMC9479267 DOI: 10.1186/s12934-022-01914-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Background Replacement of radioligand binding assays with antibody-antigen interaction-based approaches for quantitative analysis of G protein-coupled receptor (GPCR) levels requires the use of purified protein standards containing the antigen. GPCRs in general and cannabinoid CB1 receptor in particular show a progressive tendency to aggregate and precipitate in aqueous solution outside of their biological context due to the low solubility that the hydrophobic nature imprinted by their seven transmembrane domains. This renders full-length recombinant GPCRs useless for analytical purposes, a problem that can be overcome by engineering soluble recombinant fragments of the receptor containing the antigen. Results Here we generated highly soluble and stable recombinant protein constructs GST-CB1414–472 and GST-CB1414-442 containing much of the human CB1 receptor C-terminal tail for use as standard and negative control, respectively, in quantitative Western blot analysis of CB1 receptor expression on crude synaptosomes of the adult rat brain cortex. To this end we used three different antibodies, all raised against a peptide comprising the C-terminal residues 443–473 of the mouse CB1 receptor that corresponds to residues 442–472 in the human homolog. Estimated values of CB1 receptor density obtained by quantitative Western blot were of the same order of magnitude but slightly higher than values obtained by the radioligand saturation binding assay. Conclusions Collectively, here we provide a suitable Western blot-based design as a simple, cost-effective and radioactivity-free alternative for the quantitative analysis of CB1 receptor expression, and potentially of any GPCR, in a variety of biological samples. The discrepancies between the results obtained by quantitative Western blot and radioligand saturation binding techniques are discussed in the context of their particular theoretical bases and methodological constraints. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-022-01914-1.
Collapse
|
12
|
Nerella SG, Singh P, Sanam T, Digwal CS. PET Molecular Imaging in Drug Development: The Imaging and Chemistry Perspective. Front Med (Lausanne) 2022; 9:812270. [PMID: 35295604 PMCID: PMC8919964 DOI: 10.3389/fmed.2022.812270] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
Positron emission tomography with selective radioligands advances the drug discovery and development process by revealing information about target engagement, proof of mechanism, pharmacokinetic and pharmacodynamic profiles. Positron emission tomography (PET) is an essential and highly significant tool to study therapeutic drug development, dose regimen, and the drug plasma concentrations of new drug candidates. Selective radioligands bring up target-specific information in several disease states including cancer, cardiovascular, and neurological conditions by quantifying various rates of biological processes with PET, which are associated with its physiological changes in living subjects, thus it reveals disease progression and also advances the clinical investigation. This study explores the major roles, applications, and advances of PET molecular imaging in drug discovery and development process with a wide range of radiochemistry as well as clinical outcomes of positron-emitting carbon-11 and fluorine-18 radiotracers.
Collapse
Affiliation(s)
- Sridhar Goud Nerella
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Priti Singh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Tulja Sanam
- Department of Microbiology and Applied Sciences, University of Agricultural Sciences, Bangalore, India
| | - Chander Singh Digwal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
13
|
Kolaczynska KE, Luethi D, Trachsel D, Hoener MC, Liechti ME. Receptor Interaction Profiles of 4-Alkoxy-3,5-Dimethoxy-Phenethylamines (Mescaline Derivatives) and Related Amphetamines. Front Pharmacol 2022; 12:794254. [PMID: 35222010 PMCID: PMC8865417 DOI: 10.3389/fphar.2021.794254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
3,4,5-Trimethoxyphenethylamine (mescaline) is a psychedelic alkaloid found in peyote cactus. Related 4-alkoxy-3,5-dimethoxy-substituted phenethylamines (scalines) and amphetamines (3C-scalines) are reported to induce similarly potent psychedelic effects and are therefore potential novel therapeutics for psychedelic-assisted therapy. Herein, several pharmacologically uninvestigated scalines and 3C-scalines were examined at key monoamine targets in vitro. Binding affinity at human serotonergic 5-HT1A, 5-HT2A, and 5-HT2C, adrenergic α1A and α2A, and dopaminergic D2 receptors, rat and mouse trace amine-associated receptor 1 (TAAR1), and human monoamine transporters were assessed using target specific transfected cells. Furthermore, activation of human 5-HT2A and 5-HT2B receptors, and TAAR1 was examined. Generally, scalines and 3C-scalines bound with weak to moderately high affinity to the 5-HT2A receptor (Ki = 150–12,000 nM). 3C-scalines showed a marginal preference for the 5-HT2A vs the 5-HT2C and 5-HT1A receptors whereas no preference was observed for the scalines. Extending the 4-alkoxy substituent increased 5-HT2A and 5-HT2C receptors binding affinities, and enhanced activation potency and efficacy at the 5-HT2A but not at the 5-HT2B receptor. Introduction of fluorinated 4-alkoxy substituents generally increased 5-HT2A and 5-HT2C receptors binding affinities and increased the activation potency and efficacy at the 5-HT2A and 5-HT2B receptors. Overall, no potent affinity was observed at non-serotonergic targets. As observed for other psychedelics, scalines and 3C-scalines interacted with the 5-HT2A and 5-HT2C receptors and bound with higher affinities (up to 63-fold and 34-fold increase, respectively) when compared to mescaline.
Collapse
Affiliation(s)
- Karolina E Kolaczynska
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.,Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Marius C Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
Ribeiro D, Hallett W, Howes O, McCutcheon R, Nour MM, Tavares AAS. Assessing the impact of different penalty factors of the Bayesian reconstruction algorithm Q.Clear on in vivo low count kinetic analysis of [ 11C]PHNO brain PET-MR studies. EJNMMI Res 2022; 12:11. [PMID: 35184229 PMCID: PMC8859021 DOI: 10.1186/s13550-022-00883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/26/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Q.Clear is a Bayesian penalised likelihood (BPL) reconstruction algorithm available on General Electric (GE) Positron Emission Tomography (PET)-Computed Tomography (CT) and PET-Magnetic Resonance (MR) scanners. This algorithm is regulated by a β value which acts as a noise penalisation factor and yields improvements in signal to noise ratio (SNR) in clinical scans, and in contrast recovery and spatial resolution in phantom studies. However, its performance in human brain imaging studies remains to be evaluated in depth. This pilot study aims to investigate the impact of Q.Clear reconstruction methods using different β value versus ordered subset expectation maximization (OSEM) on brain kinetic modelling analysis of low count brain images acquired in the PET-MR. METHODS Six [11C]PHNO PET-MR brain datasets were reconstructed with Q.Clear with β100-1000 (in increments of 100) and OSEM. The binding potential relative to non-displaceable volume (BPND) were obtained for the Substantia Nigra (SN), Striatum (St), Globus Pallidus (GP), Thalamus (Th), Caudate (Cd) and Putamen (Pt), using the MIAKAT™ software. Intraclass correlation coefficients (ICC), repeatability coefficients (RC), coefficients of variation (CV) and bias from Bland-Altman plots were reported. Statistical analysis was conducted using a 2-way ANOVA model with correction for multiple comparisons. RESULTS When comparing a standard OSEM reconstruction of 6 iterations/16 subsets and 5 mm filter with Q.Clear with different β values under low counts, the bias and RC were lower for Q.Clear with β100 for the SN (RC = 2.17), Th (RC = 0.08) and GP (RC = 0.22) and with β200 for the St (RC = 0.14), Cd (RC = 0.18)and Pt (RC = 0.10). The p-values in the 2-way ANOVA model corroborate these findings. ICC values obtained for Th, St, GP, Pt and Cd demonstrate good reliability (0.87, 0.99, 0.96, 0.99 and 0.96, respectively). For the SN, ICC values demonstrate poor reliability (0.43). CONCLUSION BPND results obtained from quantitative low count brain PET studies using [11C]PHNO and reconstructed with Q.Clear with β < 400, which is the value used for clinical [18F]FDG whole-body studies, demonstrate the lowest bias versus the typical iterative reconstruction method OSEM.
Collapse
Affiliation(s)
- Daniela Ribeiro
- Invicro, Centre for Imaging Sciences, Hammersmith Hospital, Invicro, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK.
- Edinburgh Imaging, The University of Edinburgh, Edinburgh, UK.
| | - William Hallett
- Invicro, Centre for Imaging Sciences, Hammersmith Hospital, Invicro, Imperial College London, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Institute of Medical Sciences, Medical Research Council London, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Robert McCutcheon
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Institute of Medical Sciences, Medical Research Council London, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Matthew M Nour
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Max Planck Centre for Computational Psychiatry and Ageing Research, Institute of Neurology, University College London, London, UK
- Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, London, UK
| | | |
Collapse
|
15
|
Design, Synthesis, and Biological Evaluation of 4,4’-Difluorobenzhydrol Carbamates as Selective M1 Antagonists. Pharmaceuticals (Basel) 2022; 15:ph15020248. [PMID: 35215360 PMCID: PMC8879200 DOI: 10.3390/ph15020248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Due to their important role in mediating a broad range of physiological functions, muscarinic acetylcholine receptors (mAChRs) have been a promising target for therapeutic and diagnostic applications alike; however, the list of truly subtype-selective ligands is scarce. Within this work, we have identified a series of twelve 4,4’-difluorobenzhydrol carbamates through a rigorous docking campaign leveraging commercially available amine databases. After synthesis, these compounds have been evaluated for their physico–chemical property profiles, including characteristics such as HPLC-logD, tPSA, logBB, and logPS. For all the synthesized carbamates, these characteristics indicate the potential for BBB permeation. In competitive radioligand binding experiments using Chinese hamster ovary cell membranes expressing the individual human mAChR subtype hM1-hM5, the most promising compound 2 displayed a high binding affinitiy towards hM1R (1.2 nM) while exhibiting modest-to-excellent selectivity versus the hM2-5R (4–189-fold). All 12 compounds were shown to act in an antagonistic fashion towards hM1R using a dose-dependent calcium mobilization assay. The structural eligibility for radiolabeling and their pharmacological and physico–chemical property profiles render compounds 2, 5, and 7 promising candidates for future position emission tomography (PET) tracer development.
Collapse
|
16
|
Neural correlates of beneficial effects of young plasma treatment in aged mice: PET-SPM analyses and neuro-behavioural/molecular biological studies. Eur J Nucl Med Mol Imaging 2021; 49:1456-1469. [PMID: 34859282 DOI: 10.1007/s00259-021-05598-4] [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: 07/19/2021] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE To investigate the in vivo neurofunctional changes and therapeutic effects of young blood plasma (YBP) in aged mice, as well as the molecular mechanisms underlying the therapeutic effects of YBP ex vivo and in vitro. METHODS Aged C57/BL6 mice received systemic administrations of phosphate-buffered saline (PBS) or YBP twice a week, for 4 weeks. In vivo 2-[18F]-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) under conscious state and cognitive behavioural tests were performed after 4-week treatment. In addition, an in vitro senescent model was established, and the expressions of key cognition-associated proteins and/or the alterations of key neuronal pathways were analysed in both brain tissues and cultured cells. RESULTS Aged mice treated with YBP demonstrated higher glucose metabolism in the right hippocampus and bilateral somatosensory cortices, and lower glucose metabolism in the right bed nucleus of stria terminalis and left cerebellum. YBP treatment exerted beneficial effects on the spatial and long-term social recognition memory, and significantly increased the expressions of several cognition-related proteins and altered the key neuronal signalling pathways in the hippocampus and somatosensory cortex. Further in vitro studies suggested that YBP but not aged blood plasma significantly upregulated the expressions of several cognition-associated proteins. CONCLUSION Our results highlight the role of the hippocampus and somatosensory cortex in YBP-induced beneficial effects on recognition memory in aged mice. 18F-FDG PET imaging under conscious state provides a new avenue for exploring the mechanisms underlying YBP treatment against age-related cognitive decline.
Collapse
|
17
|
Placzek MS. Imaging Kappa Opioid Receptors in the Living Brain with Positron Emission Tomography. Handb Exp Pharmacol 2021; 271:547-577. [PMID: 34363128 DOI: 10.1007/164_2021_498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Kappa opioid receptor (KOR) neuroimaging using positron emission tomography (PET) has been immensely successful in all phases of discovery and validation in relation to radiotracer development from preclinical imaging to human imaging. There are now several KOR-specific PET radiotracers that can be utilized for neuroimaging, including agonist and antagonist ligands, as well as C-11 and F-18 variants. These technologies will increase KOR PET utilization by imaging centers around the world and have provided a foundation for future studies. In this chapter, I review the advances in KOR radiotracer discovery, focusing on ligands that have been translated into human imaging, and highlight key attributes unique to each KOR PET radiotracer. The utilization of these radiotracers in KOR PET neuroimaging can be subdivided into three major investigational classes: the first, measurement of KOR density; the second, measurement of KOR drug occupancy; the third, detecting changes in endogenous dynorphin following activation or deactivation. Given the involvement of the KOR/dynorphin system in a number of brain disorders including, but not limited to, pain, itch, mood disorders and addiction, measuring KOR density in the living brain will offer insight into the chronic effects of these disorders on KOR tone in humans. Notably, KOR PET has been successful at measuring drug occupancy in the human brain to guide dose selection for maximal therapeutic efficacy while avoiding harmful side effects. Lastly, we discuss the potential of KOR PET to detect changes in endogenous dynorphin in the human brain, to elucidate neural mechanisms and offer critical insight into disease-modifying therapeutics. We conclude with comments on other translational neuroimaging modalities such as MRI that could be used to study KOR-dynorphin tone in the living human brain.
Collapse
Affiliation(s)
- Michael S Placzek
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA. .,Department of Radiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Vidal B, Levigoureux E, Chaib S, Bouillot C, Billard T, Newman-Tancredi A, Zimmer L. Different Alterations of Agonist and Antagonist Binding to 5-HT1A Receptor in a Rat Model of Parkinson’s Disease and Levodopa-Induced Dyskinesia: A MicroPET Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:1257-1269. [DOI: 10.3233/jpd-212580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: The gold-standard treatment for Parkinson’s disease is L-DOPA, which in the long term often leads to levodopa-induced dyskinesia. Serotonergic neurons are partially responsible for this, by converting L-DOPA into dopamine leading to its uncontrolled release as a “false neurotransmitter”. The stimulation of 5-HT1A receptors can reduce involuntary movements but this mechanism is poorly understood. Objective: This study aimed to investigate the functionality of 5-HT1A receptors using positron emission tomography in hemiparkinsonian rats with or without dyskinesia induced by 3-weeks daily treatment with L-DOPA. Imaging sessions were performed “off” L-DOPA. Methods: Each rat underwent a positron emission tomography scan with [18F]F13640, a 5-HT1AR agonist which labels receptors in a high affinity state for agonists, or with [18F]MPPF, a 5-HT1AR antagonist which labels all the receptors. Results: There were decreases of [18F]MPPF binding in hemiparkinsonian rats in cortical areas. In dyskinetic animals, changes were slighter but also found in other regions. In hemiparkinsonian rats, [18F]F13640 uptake was decreased bilaterally in the globus pallidus and thalamus. On the non-lesioned side, binding was increased in the insula, the hippocampus and the amygdala. In dyskinetic animals, [18F]F13640 binding was strongly increased in cortical and limbic areas, especially in the non-lesioned side. Conclusion: These data suggest that agonist and antagonist 5-HT1A receptor-binding sites are differently modified in Parkinson’s disease and levodopa-induced dyskinesia. In particular, these observations suggest a substantial involvement of the functional state of 5-HT1AR in levodopa-induced dyskinesia and emphasize the need to characterize this state using agonist radiotracers in physiological and pathological conditions.
Collapse
Affiliation(s)
- Benjamin Vidal
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
| | - Elise Levigoureux
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | - Sarah Chaib
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
| | | | - Thierry Billard
- CERMEP-Imaging Platform, Bron, France
- Institute of Chemistry and Biochemistry, Université de Lyon, CNRS, Villeurbanne, France
| | | | - Luc Zimmer
- Lyon Neuroscience Research Center, Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Lyon, France
- Hospices Civils de Lyon, Lyon, France
- CERMEP-Imaging Platform, Bron, France
| |
Collapse
|
19
|
Amini M, Pedram MM, Moradi A, Jamshidi M, Ouchani M. Single and Combined Neuroimaging Techniques for Alzheimer's Disease Detection. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2021; 2021:9523039. [PMID: 34335726 PMCID: PMC8292054 DOI: 10.1155/2021/9523039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/04/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) consists of the gradual process of decreasing volume and quality of neuron connection in the brain, which consists of gradual synaptic integrity and loss of cognitive functions. In recent years, there has been significant attention in AD classification and early detection with machine learning algorithms. There are different neuroimaging techniques for capturing data and using it for the classification task. Input data as images will help machine learning models to detect different biomarkers for AD classification. This marker has a more critical role for AD detection than other diseases because beta-amyloid can extract complex structures with some metal ions. Most researchers have focused on using 3D and 4D convolutional neural networks for AD classification due to reasonable amounts of data. Also, combination neuroimaging techniques like functional magnetic resonance imaging and positron emission tomography for AD detection have recently gathered much attention. However, gathering a combination of data can be expensive, complex, and tedious. For time consumption reasons, most patients prefer to throw one of the neuroimaging techniques. So, in this review article, we have surveyed different research studies with various neuroimaging techniques and ML methods to see the effect of using combined data as input. The result has shown that the use of the combination method would increase the accuracy of AD detection. Also, according to the sensitivity metrics from different machine learning methods, MRI and fMRI showed promising results.
Collapse
Affiliation(s)
- Morteza Amini
- Department of Cognitive Modeling, Institute for Cognitive Science Studies, Shahid Beheshti University, Tehran, Iran
| | - Mir Mohsen Pedram
- Department of Electrical and Computer Engineering, Faculty of Engineering, Kharazmi University, Tehran, Iran
- Department of Cognitive Modeling, Institute for Cognitive Science Studies, Tehran, Iran
| | - Alireza Moradi
- Department of Clinical Psychology, Faculty of Psychology and Educational Science, Kharazmi University, Tehran, Iran
- Department of Cognitive Psychology, Institute for Cognitive Science Studies, Tehran, Iran
| | - Mahdieh Jamshidi
- Department of Mathematical Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mahshad Ouchani
- Institute for Cognitive and Brain Science, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
20
|
Newman-Tancredi A, Depoortère RY, Kleven MS, Kołaczkowski M, Zimmer L. Translating biased agonists from molecules to medications: Serotonin 5-HT 1A receptor functional selectivity for CNS disorders. Pharmacol Ther 2021; 229:107937. [PMID: 34174274 DOI: 10.1016/j.pharmthera.2021.107937] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022]
Abstract
Biased agonism (or "functional selectivity") at G-protein-coupled receptors has attracted rapidly increasing interest as a means to improve discovery of more efficacious and safer pharmacotherapeutics. However, most studies are limited to in vitro tests of cellular signaling and few biased agonists have progressed to in vivo testing. As concerns 5-HT1A receptors, which exert a major control of serotonergic signaling in diverse CNS regions, study of biased agonism has previously been limited by the poor target selectivity and/or partial agonism of classically available ligands. However, a new generation of highly selective, efficacious and druggable agonists has advanced the study of biased agonism at this receptor and created new therapeutic opportunities. These novel agonists show differential properties for G-protein signaling, cellular signaling (particularly pERK), electrophysiological effects, neurotransmitter release, neuroimaging by PET and pharmacoMRI, and behavioral tests of mood, motor activity and side effects. Overall, NLX-101 (a.k.a. F15599) exhibits preferential activation of cortical and brain stem 5-HT1A receptors, whereas NLX-112 (a.k.a. befiradol or F13640) shows prominent activation of 5-HT1A autoreceptors in Raphe nuclei and in regions associated with motor control. Accordingly, NLX-101 is potently active in rodent models of depression and respiratory control, whereas NLX-112 shows promising activity in models of Parkinson's disease across several species - rat, marmoset and macaque. Moreover, NLX-112 has also been labeled with 18F to produce the first agonist PET radiopharmaceutical (known as [18F]-F13640) for investigation of the active state of 5-HT1A receptors in rodent, primate and human. The structure-functional activity relationships of biased agonists have been investigated by receptor modeling and novel compounds have been identified which exhibit increased affinity at 5-HT1A receptors and new profiles of cellular signaling bias, notably for β-arrestin recruitment versus pERK. Taken together, the data suggest that 5-HT1A receptor biased agonists constitute potentially superior pharmacological agents for treatment of CNS disorders involving serotonergic mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Luc Zimmer
- Université Claude Bernard Lyon1, Lyon, France; Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, CNRS-INSERM, France
| |
Collapse
|
21
|
Ozenil M, Aronow J, Millard M, Langer T, Wadsak W, Hacker M, Pichler V. Update on PET Tracer Development for Muscarinic Acetylcholine Receptors. Pharmaceuticals (Basel) 2021; 14:530. [PMID: 34199622 PMCID: PMC8229778 DOI: 10.3390/ph14060530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
The muscarinic cholinergic system regulates peripheral and central nervous system functions, and, thus, their potential as a therapeutic target for several neurodegenerative diseases is undoubted. A clinically applicable positron emission tomography (PET) tracer would facilitate the monitoring of disease progression, elucidate the role of muscarinic acetylcholine receptors (mAChR) in disease development and would aid to clarify the diverse natural functions of mAChR regulation throughout the nervous system, which still are largely unresolved. Still, no mAChR PET tracer has yet found broad clinical application, which demands mAChR tracers with improved imaging properties. This paper reviews strategies of mAChR PET tracer design and summarizes the binding properties and preclinical evaluation of recent mAChR tracer candidates. Furthermore, this work identifies the current major challenges in mAChR PET tracer development and provides a perspective on future developments in this area of research.
Collapse
Affiliation(s)
- Marius Ozenil
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (M.O.); (J.A.); (W.W.); (M.H.)
| | - Jonas Aronow
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (M.O.); (J.A.); (W.W.); (M.H.)
| | - Marlon Millard
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, 1090 Wien, Austria; (M.M.); (T.L.)
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, 1090 Wien, Austria; (M.M.); (T.L.)
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (M.O.); (J.A.); (W.W.); (M.H.)
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (M.O.); (J.A.); (W.W.); (M.H.)
| | - Verena Pichler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, 1090 Wien, Austria; (M.M.); (T.L.)
| |
Collapse
|
22
|
Opposite alterations of 5-HT 2A receptor brain density in subjects with schizophrenia: relevance of radiotracers pharmacological profile. Transl Psychiatry 2021; 11:302. [PMID: 34016955 PMCID: PMC8137947 DOI: 10.1038/s41398-021-01430-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
The status of serotonin 5-HT2A receptors (5-HT2ARs) in schizophrenia has been controversial. In vivo positron emission tomography neuroimaging and in vitro post-mortem binding studies have reported conflicting results about 5-HT2AR density. Radiotracers bind different receptor conformations depending on their agonist, antagonist or inverse agonist properties. This study investigates 5-HT2AR density in the post-mortem prefrontal cortex from subjects with schizophrenia and controls using three radiotracers with a different pharmacological profile. The specific binding parameters of the inverse agonist [18F]altanserin, the agonist [3H]lysergic acid diethylamide (LSD) and the antagonist [3H]MDL100907 to brain cortex membranes from 20 subjects with schizophrenia and 20 individually matched controls were evaluated under similar methodological conditions. Ten schizophrenia subjects were antipsychotic-free at death. Saturation curve analyses were performed by non-linear regression to obtain a maximal density of binding sites (Bmax) and the affinity of the respective radiotracers (Kd). In schizophrenia subjects, 5-HT2AR density was decreased when quantified by [18F]altanserin binding, whereas increased when evaluated by [3H]LSD binding. However, [3H]MDL100907 binding was unaltered. A slight loss of affinity (higher Kd) was observed exclusively in [3H]LSD binding. The findings were more evident in antipsychotic-free subjects than in antipsychotic-treated subjects. In conclusion, a higher proportion of the 5-HT2AR-active functional conformation, which is rather identified by agonist radiotracers, was observed in schizophrenia patients. A consequent reduction of the inactive 5-HT2AR conformation, which is preferentially identified by inverse agonist radiotracers, was also obtained. Antagonist radiotracers do not distinguish between molecular conformations of the receptor, and accordingly, the absence of changes was shown. These results are compatible with the proposed increased functional activity of brain cortical 5-HT2ARs in schizophrenia.
Collapse
|
23
|
Functional approaches to the study of G-protein-coupled receptors in postmortem brain tissue: [ 35S]GTPγS binding assays combined with immunoprecipitation. Pharmacol Rep 2021; 73:1079-1095. [PMID: 33876404 DOI: 10.1007/s43440-021-00253-z] [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: 02/08/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 10/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) have an enormous biochemical importance as they bind to diverse extracellular ligands and regulate a variety of physiological and pathological responses. G-protein activation measures the functional consequence of receptor occupancy at one of the earliest receptor-mediated events. Receptor coupling to G-proteins promotes the GDP/GTP exchange on Gα subunits. Thus, modulation of the binding of the poorly hydrolysable GTP analog [35S]GTPγS to the Gα-protein subunit can be used as a functional approach to quantify GPCR interaction with agonist, antagonist or inverse agonist drugs. In order to determine receptor-mediated selective activation of the different Gα-proteins, [35S]GTPγS binding assays combined with immunodetection by specific antibodies have been developed and applied to physiological and pathological brain conditions. Currently, immunoprecipitation with magnetic beads and scintillation proximity assays are the most habitual techniques for this purpose. The present review summarizes the different procedures, advantages and limitations of the [35S]GTPγS binding assays combined with selective Gα-protein sequestration methods. Experience of functional coupling of several GPCRs to different Gα-proteins and recommendations for optimal performance in brain membranes are described. One of the biggest opportunities opened by these techniques is that they enable evaluation of biased agonism in the native tissue, which results in high interest in drug discovery. The available results derived from application of these functional methodologies to study GPCR dysfunctions in neuro-psychiatric disorders are also described. In conclusion, [35S]GTPγS binding combined with antibody-mediated immunodetection represents an useful method to separately evaluate the functional activity of drugs acting on GPCRs over each Gα-protein subtype.
Collapse
|
24
|
Colom M, Vidal B, Fieux S, Redoute J, Costes N, Lavenne F, Mérida I, Irace Z, Iecker T, Bouillot C, Billard T, Newman-Tancredi A, Zimmer L. [ 18F]F13640, a 5-HT 1A Receptor Radiopharmaceutical Sensitive to Brain Serotonin Fluctuations. Front Neurosci 2021; 15:622423. [PMID: 33762906 PMCID: PMC7982540 DOI: 10.3389/fnins.2021.622423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/17/2021] [Indexed: 12/30/2022] Open
Abstract
Introduction Serotonin is involved in a variety of physiological functions and brain disorders. In this context, efforts have been made to investigate the in vivo fluctuations of this neurotransmitter using positron emission tomography (PET) imaging paradigms. Since serotonin is a full agonist, it binds preferentially to G-protein coupled receptors. In contrast, antagonist PET ligands additionally interact with uncoupled receptors. This could explain the lack of sensitivity to serotonin fluctuations of current 5-HT1A radiopharmaceuticals which are mainly antagonists and suggests that agonist radiotracers would be more appropriate to measure changes in neurotransmitter release. The present study evaluated the sensitivity to endogenous serotonin release of a recently developed, selective 5-HT1A receptor PET radiopharmaceutical, the agonist [18F]F13640 (a.k.a. befiradol or NLX-112). Materials and Methods Four cats each underwent three PET scans with [18F]F13640, i.e., a control PET scan of 90 min, a PET scan preceded 30 min before by an intravenous injection 1 mg/kg of d-fenfluramine, a serotonin releaser (blocking challenge), and a PET scan comprising the intravenous injection of 1 mg/kg of d-fenfluramine 30 min after the radiotracer injection (displacement challenge). Data were analyzed with regions of interest and voxel-based approaches. A lp-ntPET model approach was implemented to determine the dynamic of serotonin release during the challenge study. Results D-fenfluramine pretreatment elicited a massive inhibition of [18F]F13640 labeling in regions known to express 5-HT1A receptors, e.g., raphe nuclei, hippocampus, thalamus, anterior cingulate cortex, caudate putamen, occipital, frontal and parietal cortices, and gray matter of cerebellum. Administration of d-fenfluramine during PET acquisition indicates changes in occupancy from 10% (thalamus) to 31% (gray matter of cerebellum) even though the dissociation rate of [18F]F13640 over the 90 min acquisition time was modest. The lp-ntPET simulation succeeded in differentiating the control and challenge conditions. Conclusion The present findings demonstrate that labeling of 5-HT1A receptors with [18F]F13640 is sensitive to serotonin concentration fluctuations in vivo. Although the data underline the need to perform longer PET scan to ensure accurate measure of displacement, they support clinical development of [18F]F13640 as a tool to explore experimental paradigms involving physiological or pathological (neurological or neuropsychiatric pathologies) fluctuations of extracellular serotonin.
Collapse
Affiliation(s)
- Matthieu Colom
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Lyon, France
| | - Benjamin Vidal
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Sylvain Fieux
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | | | | | | | | | | | | | - Luc Zimmer
- Lyon Neuroscience Research Center, INSERM, CNRS, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Lyon, France.,CERMEP-Imagerie du Vivant, Bron, France.,Institut National des Sciences et Techniques Nucléaires, Gif-sur-Yvette, France
| |
Collapse
|
25
|
Zhang K, Mizuma H, Zhang X, Takahashi K, Jin C, Song F, Gao Y, Kanayama Y, Wu Y, Li Y, Ma L, Tian M, Zhang H, Watanabe Y. PET imaging of neural activity, β-amyloid, and tau in normal brain aging. Eur J Nucl Med Mol Imaging 2021; 48:3859-3871. [PMID: 33674892 DOI: 10.1007/s00259-021-05230-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Normal brain aging is commonly associated with neural activity alteration, β-amyloid (Aβ) deposition, and tau aggregation, driving a progressive cognitive decline in normal elderly individuals. Positron emission tomography (PET) with radiotracers targeting these age-related changes has been increasingly employed to clarify the sequence of their occurrence and the evolution of clinically cognitive deficits. Herein, we reviewed recent literature on PET-based imaging of normal human brain aging in terms of neural activity, Aβ, and tau. Neural hypoactivity reflected by decreased glucose utilization with PET imaging has been predominately reported in the frontal, cingulate, and temporal lobes of the normal aging brain. Aβ PET imaging uncovers the pathophysiological association of Aβ deposition with cognitive aging, as well as the potential mechanisms. Tau-associated cognitive changes in normal aging are likely independent of but facilitated by Aβ as indicated by tau and Aβ PET imaging. Future longitudinal studies using multi-radiotracer PET imaging combined with other neuroimaging modalities, such as magnetic resonance imaging (MRI) morphometry, functional MRI, and magnetoencephalography, are essential to elucidate the neuropathological underpinnings and interactions in normal brain aging.
Collapse
Affiliation(s)
- Kai Zhang
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan. .,Interntional Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan.
| | - Hiroshi Mizuma
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan.,Kavli Institute for the Physics and Mathematics of the Universe, The University of Tokyo, Chiba, Kashiwa, 277-8583, Japan
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Kayo Takahashi
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Fahuan Song
- Department of Nuclear Medicine, Zhejiang Province People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Yuanxue Gao
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Yousuke Kanayama
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan.,Kavli Institute for the Physics and Mathematics of the Universe, The University of Tokyo, Chiba, Kashiwa, 277-8583, Japan
| | - Yuping Wu
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Yuting Li
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Lijuan Ma
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, 310007, China. .,The College of Biomedical Engineering and Instrument Science of Zhejiang University, Hangzhou, Zhejiang, 310007, China.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan.
| |
Collapse
|
26
|
Kilbourn MR. 11C- and 18F-Radiotracers for In Vivo Imaging of the Dopamine System: Past, Present and Future. Biomedicines 2021; 9:108. [PMID: 33499179 PMCID: PMC7912183 DOI: 10.3390/biomedicines9020108] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022] Open
Abstract
The applications of positron emission tomography (PET) imaging to study brain biochemistry, and in particular the aspects of dopamine neurotransmission, have grown significantly over the 40 years since the first successful in vivo imaging studies in humans. In vivo PET imaging of dopaminergic functions of the central nervous system (CNS) including dopamine synthesis, vesicular storage, synaptic release and receptor binding, and reuptake processes, are now routinely used for studies in neurology, psychiatry, drug abuse and addiction, and drug development. Underlying these advances in PET imaging has been the development of the unique radiotracers labeled with positron-emitting radionuclides such as carbon-11 and fluorine-18. This review focuses on a selection of the more accepted and utilized PET radiotracers currently available, with a look at their past, present and future.
Collapse
Affiliation(s)
- Michael R Kilbourn
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
27
|
Neuronal Dopamine D3 Receptors: Translational Implications for Preclinical Research and CNS Disorders. Biomolecules 2021; 11:biom11010104. [PMID: 33466844 PMCID: PMC7830622 DOI: 10.3390/biom11010104] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Dopamine (DA), as one of the major neurotransmitters in the central nervous system (CNS) and periphery, exerts its actions through five types of receptors which belong to two major subfamilies such as D1-like (i.e., D1 and D5 receptors) and D2-like (i.e., D2, D3 and D4) receptors. Dopamine D3 receptor (D3R) was cloned 30 years ago, and its distribution in the CNS and in the periphery, molecular structure, cellular signaling mechanisms have been largely explored. Involvement of D3Rs has been recognized in several CNS functions such as movement control, cognition, learning, reward, emotional regulation and social behavior. D3Rs have become a promising target of drug research and great efforts have been made to obtain high affinity ligands (selective agonists, partial agonists and antagonists) in order to elucidate D3R functions. There has been a strong drive behind the efforts to find drug-like compounds with high affinity and selectivity and various functionality for D3Rs in the hope that they would have potential treatment options in CNS diseases such as schizophrenia, drug abuse, Parkinson’s disease, depression, and restless leg syndrome. In this review, we provide an overview and update of the major aspects of research related to D3Rs: distribution in the CNS and periphery, signaling and molecular properties, the status of ligands available for D3R research (agonists, antagonists and partial agonists), behavioral functions of D3Rs, the role in neural networks, and we provide a summary on how the D3R-related drug research has been translated to human therapy.
Collapse
|
28
|
Razakarivony O, Newman-Tancredi A, Zimmer L. Towards in vivo imaging of functionally active 5-HT 1A receptors in schizophrenia: concepts and challenges. Transl Psychiatry 2021; 11:22. [PMID: 33414418 PMCID: PMC7791062 DOI: 10.1038/s41398-020-01119-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 12/18/2022] Open
Abstract
The serotonin 5-HT1A receptor has attracted wide attention as a target for treatment of psychiatric disorders. Although this receptor is important in the pharmacological mechanisms of action of new-generation antipsychotics, its characterization remains incomplete. Studies based on in vitro molecular imaging on brain tissue by autoradiography, and more recently in vivo PET imaging, have not yielded clear results, in particular due to the limitations of current 5-HT1A radiotracers, which lack specificity and/or bind to all 5-HT1A receptors, regardless of their functional status. The new concept of PET neuroimaging of functionally active G-protein-coupled receptors makes it possible to revisit PET brain exploration by enabling new research paradigms. For the 5-HT1A receptor it is now possible to use [18F]-F13640, a 5-HT1A receptor radioligand with high efficacy agonist properties, to specifically visualize and quantify functionally active receptors, and to relate this information to subjects' pathophysiological or pharmacological state. We therefore propose imaging protocols to follow changes in the pattern of functional 5-HT1A receptors in relation to mood deficits or cognitive processes. This could allow improved discrimination of different schizophrenia phenotypes and greater understanding of the basis of therapeutic responses to antipsychotic drugs. Finally, as well as targeting functionally active receptors to gain insights into the role of 5-HT1A receptors, the concept can also be extended to the study of other receptors involved in the pathophysiology or therapy of psychiatric disorders.
Collapse
Affiliation(s)
- Oriane Razakarivony
- grid.25697.3f0000 0001 2172 4233Université de Lyon, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Lyon, France
| | | | - Luc Zimmer
- Université de Lyon, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France. .,Hospices Civils de Lyon, Lyon, France. .,CERMEP-Imagerie du Vivant, Bron, France. .,French National Institute for Nuclear Science and Technology, CEA Saclay, Gif-sur-Yvette, France.
| |
Collapse
|
29
|
Hamano S, Tomokiyo A, Hasegawa D, Yuda A, Sugii H, Yoshida S, Mitarai H, Wada N, Maeda H. Functions of beta2-adrenergic receptor in human periodontal ligament cells. J Cell Biochem 2020; 121:4798-4808. [PMID: 32115771 DOI: 10.1002/jcb.29706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022]
Abstract
Adrenergic receptors (ARs) are receptors of noradrenalin and adrenalin, of which there are nine different subtypes. In particular, β2 adrenergic receptor (β2-AR) is known to be related to the restoration and maintenance of homeostasis in bone and cardiac tissues; however, the functional role of signaling through β2-AR in periodontal ligament (PDL) tissue has not been fully examined. In this report, we investigated that β2-AR expression in PDL tissues and their features in PDL cells. β2-AR expressed in rat PDL tissues and human PDL cells (HPDLCs) derived from two different patients (HPDLCs-2G and -3S). Rat PDL tissue with occlusal loading showed high β2-AR expression, while its expression was downregulated in that without loading. In HPDLCs, β2-AR expression was increased exposed to stretch loading. The gene expression of PDL-related molecules was investigated in PDL clone cells (2-23 cells) overexpressing β2-AR. Their gene expression and intracellular cyclic adenosine monophosphate (cAMP) levels were also investigated in HPDLCs treated with a specific β2-AR agonist, fenoterol (FEN). Overexpression of β2-AR significantly promoted the gene expression of PDL-related molecules in 2 to 23 cells. FEN led to an upregulation in the expression of PDL-related molecules and increased intracellular cAMP levels in HPDLCs. In both HPDLCs, inhibition of cAMP signaling by using protein kinase A inhibitor suppressed the FEN-induced gene expression of α-smooth muscle actin. Our findings suggest that the occlusal force is important for β2-AR expression in PDL tissue and β2-AR is involved in fibroblastic differentiation and collagen synthesis of PDL cells. The signaling through β2-AR might be important for restoration and homeostasis of PDL tissue.
Collapse
Affiliation(s)
- Sayuri Hamano
- Division of Oral Rehabilitation, Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of OBT Research Center, Kyushu University, Fukuoka, Japan
| | - Atsushi Tomokiyo
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Daigaku Hasegawa
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Asuka Yuda
- Division of General Dentistry, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hideki Sugii
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Shinichiro Yoshida
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hiromi Mitarai
- Division of General Dentistry, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- Division of General Dentistry, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hidefumi Maeda
- Division of Oral Rehabilitation, Department of Endodontology and Operative Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Endodontology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| |
Collapse
|
30
|
Synthesis, Biological, and Computational Evaluation of Antagonistic, Chiral Hydrobenzoin Esters of Arecaidine Targeting mAChR M1. Pharmaceuticals (Basel) 2020; 13:ph13120437. [PMID: 33266067 PMCID: PMC7760838 DOI: 10.3390/ph13120437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Muscarinic acetylcholine receptors (mAChRs) are a pivotal constituent of the central and peripheral nervous system. Yet, therapeutic and diagnostic applications thereof are hampered by the lack of subtype selective ligands. Within this work, we synthesized and chemically characterized three different stereoisomers of hydrobenzoin esters of arecaidine by NMR, HR-MS, chiral chromatography, and HPLC-logP. All compounds are structurally eligible for carbon-11 labeling and show appropriate stability in Dulbecco’s phosphate-buffered saline (DPBS) and F12 cell culture medium. A competitive radioligand binding assay on Chinese hamster ovary cell membranes comprising the human mAChR subtypes M1-M5 showed the highest orthosteric binding affinity for subtype M1 and a strong influence of stereochemistry on binding affinity, which corresponds to in silico molecular docking experiments. Ki values toward M1 were determined as 99 ± 19 nM, 800 ± 200 nM, and 380 ± 90 nM for the (R,R)-, (S,S)-, and racemic (R,S)-stereoisomer, respectively, highlighting the importance of stereochemical variations in mAChR ligand development. All three stereoisomers were shown to act as antagonists toward mAChR M1 using a Fluo-4 calcium efflux assay. With respect to future positron emission tomography (PET) tracer development, the (R,R)-isomer appears especially promising as a lead structure due to its highest subtype selectivity and lowest Ki value.
Collapse
|
31
|
Optimizing clozapine for chemogenetic neuromodulation of somatosensory cortex. Sci Rep 2020; 10:6001. [PMID: 32265461 PMCID: PMC7138833 DOI: 10.1038/s41598-020-62923-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 11/25/2022] Open
Abstract
Clozapine (CLZ) has been proposed as an agonist for Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to replace Clozapine-N-oxide (CNO); however, there are no reliable guidelines for the use of CLZ for chemogenetic neuromodulation. We titrated the optimal dose of CLZ required to evoke changes in neural activity whilst avoiding off-target effects. We also performed [18F]Fluoro-deoxy-glucose micro positron emission tomography (FDG-microPET) scans to determine the global effect of CLZ-induced hM3D(Gq) DREADD activation in the rat brain. Our results show that low doses of CLZ (0.1 and 0.01 mg/kg) successfully induced neural responses without off-target effects. CLZ at 1 mg/kg evoked a stronger and longer-lasting neural response but produced off-target effects, observed as changes in locomotor behavior and FDG-microPET imaging. Unexpectedly, FDG-microPET imaging failed to demonstrate an increase in regional glucose metabolism in the stimulated cortex during CLZ chemogenetic neuromodulation. Therefore, caution should be used when interpreting FDG-PET images in the context of cortical chemogenetic activation.
Collapse
|