1
|
Jari S, Ratne N, Tadas M, Katariya R, Kale M, Umekar M, Taksande B. Imidazoline receptors as a new therapeutic target in Huntington's disease: A preclinical overview. Ageing Res Rev 2024; 101:102482. [PMID: 39236858 DOI: 10.1016/j.arr.2024.102482] [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: 05/16/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
An autosomal dominant neurodegenerative disease called Huntington's disease (HD) is characterized by motor dysfunction, cognitive decline, and a variety of psychiatric symptoms due to the expansion of polyglutamine in the Huntingtin gene. The disease primarily affects the striatal neurons within the basal ganglia, leading to significant neuronal loss and associated symptoms such as chorea and dystonia. Current therapeutic approaches focus on symptom management without altering the disease's progression, highlighting a pressing need for novel treatment strategies. Recent studies have identified imidazoline receptors (IRs) as promising targets for neuroprotective and disease-modifying interventions in HD. IRs, particularly the I1 and I2 subtypes, are involved in critical physiological processes such as neurotransmission, neuronal excitability, and cell survival. Activation of these receptors has been shown to modulate neurotransmitter release and provide neuroprotective effects in preclinical models of neurodegeneration. This review discusses the potential of IR-targeted therapies to not only alleviate multiple symptoms of HD but also possibly slow the progression of the disease. We emphasize the necessity for ongoing research to further elucidate the role of IRs in HD and develop selective ligands that could lead to effective and safe treatments, thereby significantly improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Sakshi Jari
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Nandini Ratne
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
2
|
Bagán A, Rodriguez-Arévalo S, Taboada-Jara T, Griñán-Ferré C, Pallàs M, Brocos-Mosquera I, Callado LF, Morales-García JA, Pérez B, Diaz C, Fernández-Godino R, Genilloud O, Beljkas M, Oljacic S, Nikolic K, Escolano C. Preclinical Evaluation of an Imidazole-Linked Heterocycle for Alzheimer's Disease. Pharmaceutics 2023; 15:2381. [PMID: 37896141 PMCID: PMC10610545 DOI: 10.3390/pharmaceutics15102381] [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: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 10/29/2023] Open
Abstract
Humanity is facing a vast prevalence of neurodegenerative diseases, with Alzheimer's disease (AD) being the most dominant, without efficacious drugs, and with only a few therapeutic targets identified. In this scenario, we aim to find molecular entities that modulate imidazoline I2 receptors (I2-IRs) that have been pointed out as relevant targets in AD. In this work, we explored structural modifications of well-established I2-IR ligands, giving access to derivatives with an imidazole-linked heterocycle as a common key feature. We report the synthesis, the affinity in human I2-IRs, the brain penetration capabilities, the in silico ADMET studies, and the three-dimensional quantitative structure-activity relationship (3D-QSAR) studies of this new bunch of I2-IR ligands. Selected compounds showed neuroprotective properties and beneficial effects in an in vitro model of Parkinson's disease, rescued the human dopaminergic cell line SH-SY5Y from death after treatment with 6-hydroxydopamine, and showed crucial anti-inflammatory effects in a cellular model of neuroinflammation. After a preliminary pharmacokinetic study, we explored the action of our representative 2-(benzo[b]thiophen-2-yl)-1H-imidazole LSL33 in a mouse model of AD (5xFAD). Oral administration of LSL33 at 2 mg/Kg for 4 weeks ameliorated 5XFAD cognitive impairment and synaptic plasticity, as well as reduced neuroinflammation markers. In summary, this new I2-IR ligand that promoted beneficial effects in a well-established AD mouse model should be considered a promising therapeutic strategy for neurodegeneration.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| | - Sergio Rodriguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| | - Teresa Taboada-Jara
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (I.B.-M.); (L.F.C.)
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, 28029 Madrid, Spain
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (I.B.-M.); (L.F.C.)
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, 28029 Madrid, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - José A. Morales-García
- Department of Cell Biology, School of Medicine, Complutense University (UCM), 28040 Madrid, Spain;
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain;
| | - Caridad Diaz
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Rosario Fernández-Godino
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Olga Genilloud
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Milan Beljkas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| |
Collapse
|
3
|
Singh P, Singh D, Srivastava P, Mishra G, Tiwari AK. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Dev Res 2023; 84:484-513. [PMID: 36779375 DOI: 10.1002/ddr.22040] [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: 10/23/2022] [Revised: 12/12/2022] [Accepted: 12/31/2022] [Indexed: 02/14/2023]
Abstract
The inadequate information about the in vivo pathological, physiological, and neurological impairments, as well as the absence of in vivo tools for assessing brain penetrance and the efficiency of newly designed drugs, has hampered the development of new techniques for the treatment for variety of new central nervous system (CNS) diseases. The searching sites such as Science Direct and PubMed were used to find out the numerous distinct tracers across 16 CNS targets including tau, synaptic vesicle glycoprotein, the adenosine 2A receptor, the phosphodiesterase enzyme PDE10A, and the purinoceptor, among others. Among the most encouraging are [18 F]FIMX for mGluR imaging, [11 C]Martinostat for Histone deacetylase, [18 F]MNI-444 for adenosine 2A imaging, [11 C]ER176 for translocator protein, and [18 F]MK-6240 for tau imaging. We also reviewed the findings for each tracer's features and potential for application in CNS pathophysiology and therapeutic evaluation investigations, including target specificity, binding efficacy, and pharmacokinetic factors. This review aims to present a current evaluation of modern positron emission tomography tracers for CNS targets, with a focus on recent advances for targets that have newly emerged for imaging in humans.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Pooja Srivastava
- Division of Cyclotron and Radiopharmaceuticals Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhananad College, University of Delhi, Alipur, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
4
|
Bagán A, Morales-García JA, Griñán-Ferré C, Díaz C, Pérez del Palacio J, Ramos MC, Vicente F, Pérez B, Brea J, Loza MI, Pallàs M, Escolano C. Insights into the Pharmacokinetics and In Vitro Cell-Based Studies of the Imidazoline I 2 Receptor Ligand B06. Int J Mol Sci 2022; 23:ijms23105408. [PMID: 35628219 PMCID: PMC9141032 DOI: 10.3390/ijms23105408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
The impact of neurodegenerative diseases (ND) is becoming unbearable for humankind due to their vast prevalence and the lack of efficacious treatments. In this scenario, we focused on imidazoline I2 receptors (I2-IR) that are widely distributed in the brain and are altered in patients with brain disorders. We took the challenge of modulating I2-IR by developing structurally new molecules, in particular, a family of bicyclic α-iminophosphonates, endowed with high affinity and selectivity to these receptors. Treatment of two murine models, one for age-related cognitive decline and the other for Alzheimer's disease (AD), with representative compound B06 ameliorated their cognitive impairment and improved their behavioural condition. Furthermore, B06 revealed beneficial in vitro ADME-Tox properties. The pharmacokinetics (PK) and metabolic profile are reported to de-risk B06 for progressing in the preclinical development. To further characterize the pharmacological properties of B06, we assessed its neuroprotective properties and beneficial effect in an in vitro model of Parkinson's disease (PD). B06 rescued the human dopaminergic cell line SH-SY5Y from death after treatment with 6-hydroxydopamine (6-OHDA) and showed a crucial anti-inflammatory effect in a cellular model of neuroinflammation. This research reveals B06 as a putative candidate for advancing in the difficult path of drug discovery and supports the modulation of I2-IR as a fresh approach for the therapy of ND.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain;
| | - José A. Morales-García
- The Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, School of Medicine, Complutense University (UCM), 28040 Madrid, Spain;
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
| | - Caridad Díaz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - José Pérez del Palacio
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Maria C. Ramos
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, 08193 Barcelona, Spain;
| | - José Brea
- Innopharma Screening Platform, BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15706 Santiago de Compostela, Spain; (J.B.); (M.I.L.)
| | - María Isabel Loza
- Innopharma Screening Platform, BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15706 Santiago de Compostela, Spain; (J.B.); (M.I.L.)
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain;
- Correspondence:
| |
Collapse
|
5
|
Brumberg J, Varrone A. New PET radiopharmaceuticals for imaging CNS diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
6
|
Vasilopoulou F, Griñán-Ferré C, Rodríguez-Arévalo S, Bagán A, Abás S, Escolano C, Pallàs M. I 2 imidazoline receptor modulation protects aged SAMP8 mice against cognitive decline by suppressing the calcineurin pathway. GeroScience 2020; 43:965-983. [PMID: 33128688 PMCID: PMC8110656 DOI: 10.1007/s11357-020-00281-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Brain aging and dementia are current problems that must be solved. The levels of imidazoline 2 receptors (I2-IRs) are increased in the brain in Alzheimer's disease (AD) and other neurodegenerative diseases. We tested the action of the specific and selective I2-IR ligand B06 in a mouse model of accelerated aging and AD, the senescence-accelerated mouse prone 8 (SAMP8) model. Oral administration of B06 for 4 weeks improved SAMP8 mouse behavior and cognition and reduced AD hallmarks, oxidative stress, and apoptotic and neuroinflammation markers. Likewise, B06 regulated glial excitatory amino acid transporter 2 and N-methyl-D aspartate 2A and 2B receptor subunit protein levels. Calcineurin (CaN) is a phosphatase that controls the phosphorylation levels of cAMP response element-binding (CREB), apoptotic mediator BCL-2-associated agonist of cell death (BAD) and GSK3β, among other molecules. Interestingly, B06 was able to reduce the levels of the CaN active form (CaN A). Likewise, CREB phosphorylation, BAD gene expression, and other factors were modified after B06 treatment. Moreover, phosphorylation of a target of CaN, nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1), was increased in B06-treated mice, impeding the transcription of genes related to neuroinflammation and neural plasticity. In summary, this I2 imidazoline ligand can exert its beneficial effects on age-related conditions by modulating CaN pathway action and affecting several molecular pathways, playing a neuroprotective role in SAMP8 mice.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain.
| |
Collapse
|
7
|
Kotagale N, Dixit M, Garmelwar H, Bhondekar S, Umekar M, Taksande B. Agmatine reverses memory deficits induced by Aβ1–42 peptide in mice: A key role of imidazoline receptors. Pharmacol Biochem Behav 2020; 196:172976. [DOI: 10.1016/j.pbb.2020.172976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
|
8
|
Abás S, Rodríguez-Arévalo S, Bagán A, Griñán-Ferré C, Vasilopoulou F, Brocos-Mosquera I, Muguruza C, Pérez B, Molins E, Luque FJ, Pérez-Lozano P, de Jonghe S, Daelemans D, Naesens L, Brea J, Loza MI, Hernández-Hernández E, García-Sevilla JA, García-Fuster MJ, Radan M, Djikic T, Nikolic K, Pallàs M, Callado LF, Escolano C. Bicyclic α-Iminophosphonates as High Affinity Imidazoline I2 Receptor Ligands for Alzheimer’s Disease. J Med Chem 2020; 63:3610-3633. [DOI: 10.1021/acs.jmedchem.9b02080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Foteini Vasilopoulou
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Carolina Muguruza
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, E-08193 Barcelona, Spain
| | - Elies Molins
- Institut de Ciència de Materials de Barcelona (CSIC), Campus UAB, E-08193 Cerdanyola, Spain
| | - F. Javier Luque
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain
| | - Pilar Pérez-Lozano
- Unit of Pharmaceutical Technology, Pharmacy and Pharmaceutical Technology, and Physical Chemistry Department, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Steven de Jonghe
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Lieve Naesens
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - José Brea
- Innopharma screening platform, BioFarma research group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - M. Isabel Loza
- Innopharma screening platform, BioFarma research group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Elena Hernández-Hernández
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Jesús A. García-Sevilla
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - M. Julia García-Fuster
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Milica Radan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Teodora Djikic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Luis F. Callado
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| |
Collapse
|
9
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
10
|
Tyacke RJ, Myers JFM, Venkataraman A, Mick I, Turton S, Passchier J, Husbands SM, Rabiner EA, Gunn RN, Murphy PS, Parker CA, Nutt DJ. Evaluation of 11C-BU99008, a PET Ligand for the Imidazoline 2 Binding Site in Human Brain. J Nucl Med 2018; 59:1597-1602. [PMID: 29523627 DOI: 10.2967/jnumed.118.208009] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/21/2018] [Indexed: 11/16/2022] Open
Abstract
The imidazoline2 binding site (I2BS) is thought to be expressed in glia and implicated in the regulation of glial fibrillary acidic protein. A PET ligand for this target would be important for the investigation of neurodegenerative and neuroinflammatory diseases. 11C-BU99008 has previously been identified as a putative PET radioligand. Here, we present the first in vivo characterization of this PET radioligand in humans and assess its test-retest reproducibility. Methods: Fourteen healthy male volunteers underwent dynamic PET imaging with 11C-BU99008 and arterial sampling. Six subjects were used in a test-retest assessment, and 8 were used in a pharmacologic evaluation, undergoing a second or third heterologous competition scan with the mixed I2BS/α2-adrenoceptor drug idazoxan (n = 8; 20, 40, 60, and 80 mg) and the mixed irreversible monoamine oxidase type A/B inhibitor isocarboxazid (n = 4; 50 mg). Regional time-activity data were generated from arterial plasma input functions corrected for metabolites using the most appropriate model to derive the outcome measure VT (regional distribution volume). All image processing and kinetic analyses were performed in MIAKAT. Results: Brain uptake of 11C-BU99008 was good, with reversible kinetics and a heterogeneous distribution consistent with known I2BS expression. Model selection criteria indicated that the 2-tissue-compartment model was preferred. VT estimates were high in the striatum (105 ± 21 mL⋅cm-3), medium in the cingulate cortex (62 ± 10 mL⋅cm-3), and low in the cerebellum (41 ± 7 mL⋅cm-3). Test-retest reliability was reasonable. The uptake was dose-dependently reduced throughout the brain by pretreatment with idazoxan, with an average block across all regions of about 60% (VT, ∼30 mL⋅cm-3) at the highest dose (80 mg). The median effective dose for idazoxan was 28 mg. Uptake was not blocked by pretreatment with the monoamine oxidase inhibitor isocarboxazid. Conclusion:11C-BU99008 in human PET studies demonstrates good brain delivery, reversible kinetics, heterogeneous distribution, specific binding signal consistent with I2BS distribution, and good test-retest reliability.
Collapse
Affiliation(s)
- Robin J Tyacke
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jim F M Myers
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Ashwin Venkataraman
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Inge Mick
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Samuel Turton
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jan Passchier
- Imanova Limited, Imperial College London, London, United Kingdom
| | - Stephen M Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | - Roger N Gunn
- Imanova Limited, Imperial College London, London, United Kingdom
- Restorative Neurosciences, Imperial College London, London, United Kingdom; and
| | - Philip S Murphy
- Experimental Medicine Imaging, GlaxoSmithKline Research and Development Limited, Stevenage, United Kingdom
| | - Christine A Parker
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
- Experimental Medicine Imaging, GlaxoSmithKline Research and Development Limited, Stevenage, United Kingdom
| | - David J Nutt
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Xuanfei L, Hao C, Zhujun Y, Yanming L, Jianping G. Imidazoline I2 receptor inhibitor idazoxan regulates the progression of hepatic fibrosis via Akt-Nrf2-Smad2/3 signaling pathway. Oncotarget 2017; 8:21015-21030. [PMID: 28423499 PMCID: PMC5400562 DOI: 10.18632/oncotarget.15472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/07/2017] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is a global health problem and its relationship with imidazoline I2 receptor has not been reported. This study aimed to investigate the effects and underlying mechanisms of imidazoline I2 receptor (I2R) inhibitor idazoxan (IDA) on carbon tetrachloride (CCl4)-induced liver fibrosis. In vivo liver fibrosis in mice was induced by intraperitoneally injections of CCl4 for eight weeks, and in vitro studies were performed on activated LX2 cells treated with transforming growth factor-β (TGF-β). Our results showed that IDA significantly improved liver inflammation, ameliorated hepatic stellate cells activation and reduced collagen accumulation by suppressing the pro-fibrogenic signaling of TGF-β/Smad. Further investigation showed that IDA significantly balanced oxidative stress through improving the expressions and activities of anti-oxidant and detoxifying enzymes and activating Nrf2-the key defender against oxidative stress with anti-fibrotic potentials. Even more impressively, knock out of Nrf2 or suppression of Akt by perifosine (PE) eliminated the anti-oxidant and anti-fibrotic effects of IDA in vivo and in vitro, suggesting that Akt/Nrf2 constitutes a critical component of IDA's protective functions. Taken together, IDA exhibits potent effects against liver fibrosis via Akt-Nrf2-Smad2/3 signaling pathway, which suggests that specifically targeting I2R may be a potentially useful therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Li Xuanfei
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Chen Hao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Yi Zhujun
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Liu Yanming
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430041, Hubei, P. R. China
| | - Gong Jianping
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
12
|
Kawamura K, Shimoda Y, Yui J, Zhang Y, Yamasaki T, Wakizaka H, Hatori A, Xie L, Kumata K, Fujinaga M, Ogawa M, Kurihara Y, Nengaki N, Zhang MR. A useful PET probe [ 11C]BU99008 with ultra-high specific radioactivity for small animal PET imaging of I 2-imidazoline receptors in the hypothalamus. Nucl Med Biol 2016; 45:1-7. [PMID: 27835825 DOI: 10.1016/j.nucmedbio.2016.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 01/23/2023]
Abstract
INTRODUCTION A positron emission tomography (PET) probe with ultra-high specific radioactivity (SA) enables measuring high receptor specific binding in brain regions by avoiding mass effect of the PET probe itself. It has been reported that PET probe with ultra-high SA can detect small change caused by endogenous or exogenous ligand. Recently, Kealey et al. developed [11C]BU99008, a more potent PET probe for I2-imidazoline receptors (I2Rs) imaging, with a conventional SA (mean 76GBq/μmol) showed higher specific binding in the brain. Here, to detect small change of specific binding for I2Rs caused by endogenous or exogenous ligand in an extremely small region, such as hypothalamus in the brain, we synthesized and evaluated [11C]BU99008 with ultra-high SA as a useful PET probe for small-animal PET imaging of I2Rs. METHODS [11C]BU99008 was prepared by [11C]methylation of N-desmethyl precursor with [11C]methyl iodide. Biodistribution, metabolite analysis, and brain PET studies were conducted in rats. RESULTS [11C]BU99008 with ultra-high SA in the range of 5400-16,600GBq/μmol were successfully synthesized (n=7), and had appropriate radioactivity for in vivo study. In the biodistribution study, the mean radioactivity levels in all investigated tissues except for the kidney did not show significant difference between [11C]BU99008 with ultra-high SA and that with conventional SA. In the metabolite analysis, the percentage of unchanged [11C]BU99008 at 30min after the injection of probes with ultra-high and conventional SA was similar in rat brain and plasma. In the PET study of rats' brain, radioactivity level (AUC30-60 min) in the hypothalamus of rats injected with [11C]BU99008 with ultra-high SA (64 [SUV ∙ min]) was significantly higher than that observed for that with conventional SA (50 [SUV ∙ min]). The specific binding of [11C]BU99008 with ultra-high SA (86% of total binding) for I2R was higher than that of conventional SA (76% of total binding). CONCLUSION A PET study using [11C]BU99008 with ultra-high SA would thus contribute to the detection of small changes in or small regions with I2R expression and hence may be useful in elucidating new functions of I2R.
Collapse
Affiliation(s)
- Kazunori Kawamura
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan.
| | - Yoko Shimoda
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Joji Yui
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Hidekatsu Wakizaka
- Department of Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Akiko Hatori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masanao Ogawa
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Yusuke Kurihara
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Nobuki Nengaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
13
|
Caprioli G, Mammoli V, Ricciutelli M, Sagratini G, Ubaldi M, Domi E, Mennuni L, Sabatini C, Galimberti C, Ferrari F, Milia C, Comi E, Lanza M, Giannella M, Pigini M, Del Bello F. Biological profile and bioavailability of imidazoline compounds on morphine tolerance modulation. Eur J Pharmacol 2015; 769:219-24. [PMID: 26593429 DOI: 10.1016/j.ejphar.2015.11.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 11/20/2022]
Abstract
Tolerance to opioid administration represents a serious medical alert in different chronic conditions. This study compares the effects of the imidazoline compounds 1, 2, and 3 on morphine tolerance in an animal model of inflammatory pain in the rat. 1, 2, and 3 have been selected in that, although bearing a common scaffold, preferentially bind to α2-adrenoceptors, imidazoline I2 receptors, or both systems, respectively. Such compounds have been tested in vivo by measuring the paw withdrawal threshold to mechanical pressure after complete Freund's adjuvant injection. To determine the ligand levels in rat plasma, an HPLC-mass spectrometry method has been developed. All the compounds significantly reduced the induction of morphine tolerance, showing different potency and duration of action. Indeed, the selective imidazoline I2 receptor interaction (2) restored the analgesic response by maintaining the same time-dependent profile observed after a single morphine administration. Differently, the selective α2C-adrenoceptor activation (1) or the combination between α2C-adrenoceptor activation and imidazoline I2 receptor engagement (3) promoted a change in the temporal profile of morphine analgesia by maintaining a mild but long lasting analgesic effect. Interestingly, the kinetics of compounds in rat plasma supported the pharmacodynamic data. Therefore, this study highlights that both peculiar biological profile and bioavailability of such ligands complement each other to modulate the reduction of morphine tolerance. Based on these observations, 1-3 can be considered useful leads in the design of new drugs able to turn off the undesired tolerance induced by opioids.
Collapse
Affiliation(s)
- Giovanni Caprioli
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Valerio Mammoli
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Massimo Ricciutelli
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Gianni Sagratini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| | - Esi Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 62032 Camerino, Italy
| | - Laura Mennuni
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy
| | - Chiara Sabatini
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy
| | - Chiara Galimberti
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy
| | - Flora Ferrari
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy
| | - Chiara Milia
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy; PhD program in Neuroscience, University of Milan-Bicocca, Italy
| | - Eleonora Comi
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy; PhD program in Neuroscience, University of Milan-Bicocca, Italy
| | - Marco Lanza
- Department of Pharmacology & Toxicology, Rottapharm Biotech S.r.l., 20900 Monza, Italy
| | - Mario Giannella
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Maria Pigini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy.
| |
Collapse
|
14
|
In vivo evaluation of a new 18F-labeled PET ligand, [18F]FEBU, for the imaging of I2-imidazoline receptors. Nucl Med Biol 2015; 42:406-12. [DOI: 10.1016/j.nucmedbio.2014.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/01/2014] [Accepted: 12/19/2014] [Indexed: 11/20/2022]
|
15
|
Kealey S, Turner EM, Husbands SM, Salinas CA, Jakobsen S, Tyacke RJ, Nutt DJ, Parker CA, Gee AD. Imaging imidazoline-I2 binding sites in porcine brain using 11C-BU99008. J Nucl Med 2012; 54:139-44. [PMID: 23223380 DOI: 10.2967/jnumed.112.108258] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Changes in the density of imidazoline-I(2) binding sites have been observed in a range of neurologic disorders including Alzheimer's disease, Huntington's chorea, and glial tumor; however, the precise function of these sites remains unclear. A PET probe for I(2) binding sites would further our understanding of the target and may find application as a biomarker for early disease diagnosis. Compound BU99008 has previously been identified as a promising I(2) ligand from autoradiography studies, displaying high affinity and good selectivity toward the target. In this study, BU99008 was radiolabeled with (11)C in order to image the I(2) binding sites in vivo using PET. METHODS (11)C-BU99008 was radiolabeled by N-alkylation of the desmethyl precursor using (11)C-methyl iodide. A series of PET experiments was performed to investigate the binding of (11)C-BU99008 in porcine brains, in the presence or absence of a nonradiolabeled, competing I(2) ligand, BU224. RESULTS (11)C-BU99008 was obtained in good yield and specific activity. In vivo, (11)C-BU99008 displayed good brain penetration and gave a heterogeneous distribution with high uptake in the thalamus and low uptake in the cortex and cerebellum. (11)C-BU99008 brain kinetics were well described by the 1-tissue-compartment model, which was used to provide estimates for the total volume of distribution (V(T)) across brain regions of interest. Baseline V(T) values were ranked in the following order: thalamus > striatum > hippocampus > frontal cortex ≥ cerebellum, consistent with the known distribution and concentration of I(2) binding sites. Administration of a selective I(2) binding site ligand, BU224, reduced the V(T) to near-homogeneous levels in all brain regions. CONCLUSION (11)C-BU99008 appears to be a suitable PET radioligand for imaging the I(2) binding sites in vivo.
Collapse
Affiliation(s)
- Steven Kealey
- Institute of Psychiatry, De Crespigny Park, King's College London, London, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Tyacke RJ, Fisher A, Robinson ESJ, Grundt P, Turner EM, Husbands SM, Hudson AL, Parker CA, Nutt DJ. Evaluation and initial in vitro and ex vivo characterization of the potential positron emission tomography ligand, BU99008 (2-(4,5-dihydro-1H-imidazol-2-yl)-1- methyl-1H-indole), for the imidazoline₂ binding site. Synapse 2012; 66:542-51. [PMID: 22290740 DOI: 10.1002/syn.21541] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/12/2012] [Accepted: 01/23/2012] [Indexed: 11/06/2022]
Abstract
The density of the Imidazoline₂ binding site (I₂BS) has been shown to change in psychiatric conditions such as depression and addiction, along with neurodegenerative disorders such as Alzheimer's disease and Huntington's chorea. The presence of I₂BS on glial cells and the possibility that they may in some way regulate glial fibrillary acidic protein has led to increased interest into the role of I₂BS and I₂BS ligands in conditions characterized by marked gliosis. In addition, it has been suggested that I₂BS may be a marker for human glioblastomas. Therefore, the development of a positron emission tomography (PET) radioligand for the I₂BS would be of major benefit in our understanding of these conditions. We now report the successful synthesis and initial pharmacological evaluation of potential PET radioligands for the I₂BS as well as the tritiation and characterization of the most favorable of the series, BU99008 (6), both in vitro and ex vivo in rat. The series as a whole demonstrated excellent affinity and selectivity for the I₂BS, with BU99008 (6) selected as the lead candidate to be taken forward for in vivo assessment. BU99008 (6) showed very good affinity for the I₂BS (K(i) of 1.4 nM; K(d) = 1.3 nM), good selectivity compared with the α₂ -adrenoceptor (909-fold). In addition, following peripheral administration, [³H]BU99008 demonstrated a heterogenous uptake into the rat brain consistent with the known distribution of the I₂BS in vivo. This, and the amenability of BU99008 (6) to radiolabeling with a positron-emitting radioisotope, indicates its potential as a PET radioligand for imaging the I₂BS in vivo.
Collapse
Affiliation(s)
- Robin J Tyacke
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kawamura K, Kimura Y, Yui J, Wakizaka H, Yamasaki T, Hatori A, Kumata K, Fujinaga M, Yoshida Y, Ogawa M, Nengaki N, Fukumura T, Zhang MR. PET study using [11C]FTIMD with ultra-high specific activity to evaluate I2-imidazoline receptors binding in rat brains. Nucl Med Biol 2012; 39:199-206. [DOI: 10.1016/j.nucmedbio.2011.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 06/29/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
|
18
|
Synthesis and evaluation of PET probes for the imaging of I2 imidazoline receptors in peripheral tissues. Nucl Med Biol 2012; 39:89-99. [DOI: 10.1016/j.nucmedbio.2011.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 06/02/2011] [Accepted: 06/07/2011] [Indexed: 11/21/2022]
|
19
|
Kawamura K, Maeda J, Hatori A, Okauchi T, Nagai Y, Higuchi M, Suhara T, Fukumura T, Zhang MR. In vivo and in vitro imaging of I2 imidazoline receptors in the monkey brain. Synapse 2011; 65:452-5. [DOI: 10.1002/syn.20897] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
20
|
Imaging of I2-imidazoline receptors by small-animal PET using 2-(3-fluoro-[4-11C]tolyl)-4,5-dihydro-1H-imidazole ([11C]FTIMD). Nucl Med Biol 2010; 37:625-35. [DOI: 10.1016/j.nucmedbio.2010.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 02/26/2010] [Accepted: 02/28/2010] [Indexed: 11/23/2022]
|
21
|
Kimura A, Tyacke RJ, Robinson JJ, Husbands SM, Minchin MC, Nutt DJ, Hudson AL. Identification of an imidazoline binding protein: creatine kinase and an imidazoline-2 binding site. Brain Res 2009; 1279:21-8. [PMID: 19410564 PMCID: PMC2722693 DOI: 10.1016/j.brainres.2009.04.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 03/28/2009] [Accepted: 04/22/2009] [Indexed: 12/11/2022]
Abstract
Drugs that bind to imidazoline binding proteins have major physiological actions. To date, three subtypes of such proteins, I(1), I(2) and I(3), have been proposed, although characterisations of these binding proteins are lacking. I(2) binding sites are found throughout the brain, particularly dense in the arcuate nucleus of the hypothalamus. Selective I(2) ligands demonstrate antidepressant-like activity and the identity of the proteins that respond to such ligands remained unknown until now. Here we report the isolation of a approximately 45 kDa imidazoline binding protein from rabbit and rat brain using a high affinity ligand for the I(2) subtype, 2-BFI, to generate an affinity column. Following protein sequencing of the isolated approximately 45 kDa imidazoline binding protein, we identified it to be brain creatine kinase (B-CK). B-CK shows high binding capacity to selective I(2) ligands; [(3)H]-2-BFI (5 nM) specifically bound to B-CK (2330+/-815 fmol mg protein(-1)). We predicted an I(2) binding pocket near the active site of B-CK using molecular modelling. Furthermore, B-CK activity was inhibited by a selective I(2) irreversible ligand, where 20 microM BU99006 reduced the enzyme activity by 16%, confirming the interaction between B-CK and the I(2) ligand. In summary, we have identified B-CK to be the approximately 45 kDa imidazoline binding protein and we have demonstrated the existence of an I(2) binding site within this enzyme. The importance of B-CK in regulating neuronal activity and neurotransmitter release may well explain the various actions of I(2) ligands in brain and the alterations in densities of I(2) binding sites in psychiatric disorders.
Collapse
Key Words
- 2-bfi, 2-(2-benzofuranyl)2-imidazoline
- bu224, 2-(4,5-dihydroimidaz-2-yl)quinoline
- bu99006, 5-isothiocyanoato-2-benzofuranyl-2-imidazoline
- b-ck, brain creatine kinase
- ck, creatine kinase
- gold, genetic optimisation for ligand docking
- gr, glucose-responsive
- i2, imidazoline-2 subtype
- katp channel, atp sensitive potassium channel
- mao, monoamine oxidase
- moe, molecular operating environment
- imidazoline binding protein
- creatine kinase
- 2-bfi
- harmane and psychiatric disorders
Collapse
Affiliation(s)
- Atsuko Kimura
- Psychopharmacology Unit, University of Bristol, BS1 3NY, UK
| | | | - James J. Robinson
- Department of Pharmacy and Pharmacology, University of Bath, BA2 7AY, UK
| | | | | | - David J. Nutt
- Psychopharmacology Unit, University of Bristol, BS1 3NY, UK
| | - Alan L. Hudson
- Department of Pharmacology, 9-70 Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| |
Collapse
|
22
|
Idazoxan attenuates spinal cord injury by enhanced astrocytic activation and reduced microglial activation in rat experimental autoimmune encephalomyelitis. Brain Res 2009; 1253:198-209. [DOI: 10.1016/j.brainres.2008.11.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 11/03/2008] [Accepted: 11/03/2008] [Indexed: 11/20/2022]
|
23
|
Saczewski F, Tabin P, Tyacke RJ, Maconie A, Saczewski J, Kornicka A, Nutt DJ, Hudson AL. 2-(4,5-Dihydroimidazol-2-yl)benzimidazoles as highly selective imidazoline I2/adrenergic α2 receptor ligands. Bioorg Med Chem 2006; 14:6679-85. [PMID: 16782348 DOI: 10.1016/j.bmc.2006.05.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Revised: 05/19/2006] [Accepted: 05/31/2006] [Indexed: 11/25/2022]
Abstract
2-(4,5-Dihydroimidazol-2-yl)benzimidazoles have been identified as selective imidazoline I2/alpha2-adrenoceptor ligands. 4-Methyl (2) and 4-chloro (4) derivatives display I2 affinity at nanomolar concentration (Ki=4.4 and 17.7 nM, respectively) and high I2/alpha2 selectivity ratio=4226 and 5649, respectively. An evidence has been obtained that pKa value influences considerably the I2/alpha2-selectivity ratio of this class of imidazoline I2 receptor ligands.
Collapse
Affiliation(s)
- Francieszek Saczewski
- Department of Chemical Technology of Drugs, Medical University of Gdańsk, 80-416 Gdańsk, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Gentili F, Cardinaletti C, Carrieri A, Ghelfi F, Mattioli L, Perfumi M, Vesprini C, Pigini M. Involvement of I2-imidazoline binding sites in positive and negative morphine analgesia modulatory effects. Eur J Pharmacol 2006; 553:73-81. [PMID: 17081513 DOI: 10.1016/j.ejphar.2006.09.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 09/05/2006] [Accepted: 09/07/2006] [Indexed: 11/26/2022]
Abstract
Some studies, suggesting the involvement of I(2)-imidazoline binding sites (I(2)-IBS) in morphine analgesia modulation, prompted us to examine on mice antinociceptive assays the effect produced by 1 (phenyzoline), that in view of its high I(2)-IBS affinity and high I(2)-IBS selectivity with regard to I(1)-IBS, alpha(2)-adrenoreceptors and mu-opioid receptors might be considered the first interesting I(2)-IBS ligand. The study was also applied to its ortho phenyl derivative 2 (diphenyzoline), designed and prepared in order to produce a possible modification of the biological profile of 1. Diphenyzoline (2) retains a significant I(2)-IBS selectivity with regard to I(1)-IBS, alpha(2)-adrenoreceptors and mu-opioid receptors. Moreover, by the functional assays 1 and 2 proved inactive at all alpha(2)-adrenoreceptors subtypes up to 10(-3) M. As expected, phenyzoline and diphenyzoline, which are structurally related, highlighted an interesting "positive" or "negative", respectively, morphine analgesia modulatory effect. In fact, 1 (s.c. 10 mg/kg) enhanced morphine analgesia (60% and 40% in mouse tail-flick and mouse hot-plate, respectively), while 2 (s.c. 10 mg/kg) decreased it (-41% and -20%, respectively). The ability to decrease morphine analgesia had never been observed before in I(2)-IBS ligands. These effects were not affected by i.p. treatment of animals with yohimbine (a selective alpha(2)-adrenoreceptor antagonist, 0.625 mg/kg) or efaroxan (an I(1)-IBS/alpha(2)-adrenoreceptor antagonist, 1.0 mg/kg). In contrast, they were completely reversed by i.p. treatment of animals with idazoxan (an I(2)-IBS/alpha(2)-adrenoreceptor antagonist, 2 mg/kg). Moreover, compound 2, in mouse tail-flick test, was able to potentiate by 23% the naloxone-induced decrease of morphine analgesia. Therefore, the results of this study indicate the crucial involvement of I(2)-IBS in the morphine analgesia modulatory effects of 1 and 2.
Collapse
Affiliation(s)
- Francesco Gentili
- Dipartimento di Scienze Chimiche, Università degli Studi di Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Tanabe M, Kino Y, Honda M, Ono H. Presynaptic I1-imidazoline receptors reduce GABAergic synaptic transmission in striatal medium spiny neurons. J Neurosci 2006; 26:1795-802. [PMID: 16467528 PMCID: PMC6793622 DOI: 10.1523/jneurosci.4642-05.2006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Imidazoline receptors are expressed widely in the CNS. In the present study, whole-cell patch-clamp recordings were made from medium spiny neurons in dorsal striatum slices from the rat brain, and the roles of I1-imidazoline receptors in the modulation of synaptic transmission were studied. Moxonidine, an I1-imidazoline receptor agonist, decreased the GABAA receptor-mediated IPSCs in a concentration-dependent manner. However, glutamate-mediated EPSCs were hardly affected. The depression of IPSCs by moxonidine was antagonized by either idazoxan or efaroxan, which are both imidazoline receptor antagonists containing an imidazoline moiety. In contrast, yohimbine and SKF86466 (6-chloro-2,3,4,5-tetrahydro-3-methyl-1H-3-benzazepine), which are alpha2-adrenergic receptor antagonists with no affinity for imidazoline receptors, did not affect the moxonidine-induced inhibition of IPSCs. Moxonidine increased the paired-pulse ratio and reduced the frequency of miniature IPSCs without affecting their amplitude, indicating that this agent inhibits IPSCs via presynaptic mechanisms. Moreover, the sulfhydryl alkylating agent N-ethylmaleimide (NEM) significantly reduced the moxonidine-induced inhibition of IPSCs. Thus, the activation of presynaptic I1-imidazoline receptors decreases GABA-mediated inhibition of medium spiny neurons in the striatum, in which NEM-sensitive proteins such as G(i/o)-type G-proteins play an essential role. The adenylate cyclase activator forskolin partly opposed IPSC inhibition elicited by subsequently applied moxonidine. Furthermore, the protein kinase C (PKC) activator phorbol 12,13-dibutyrate attenuated and the PKC inhibitor chelerythrine potentiated the moxonidine-induced inhibition of IPSCs. These results suggest that IPSC inhibition via presynaptic I1-imidazoline receptors involves intracellular adenylate cyclase activity and is influenced by static PKC activity in the striatum.
Collapse
Affiliation(s)
- Mitsuo Tanabe
- Laboratory of CNS Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| | | | | | | |
Collapse
|
26
|
Anderson NJ, Lupo PA, Nutt DJ, Hudson AL, Robinson ESJ. Characterisation of imidazoline I2 binding sites in pig brain. Eur J Pharmacol 2005; 519:68-74. [PMID: 16109401 DOI: 10.1016/j.ejphar.2005.06.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2005] [Accepted: 06/28/2005] [Indexed: 11/29/2022]
Abstract
The imidazoline I2 binding sites in the central nervous system have previously been described in several different species including rat, mouse, rabbit and frog. The present study has investigated the imidazoline I2 binding site, and its relationship to the monoamine oxidase isoforms, in pig whole brain and compared the results obtained with data from other species. Results from saturation binding studies revealed that the imidazoline I2-selective ligand, [3H]2BFI (2-(2-benzofuranyl)-2-imidazoline) labelled a single saturable population of sites with a KD=6.6 nM and Bmax=771.7 fmol/mg protein. The pharmacological characterisation of the sites was similar to that previously reported with a rank order of potency for the imidazoline I2 ligands of 2BFI>BU224>Idazoxan>BU226. Displacement by the imidazoline I1 ligands was low affinity and the monoamine oxidase inhibitors displaced with micromolar affinity. The majority of compounds displaced the binding in a monophasic manner, however, displacement by the putative endogenous ligand, harmane was biphasic. The relative populations of the two monoamine oxidase isoforms revealed a 10 fold greater expression of monoamine oxidase B relative to monoamine oxidase A. These data confirm the presence of imidazoline I2 binding sites in pig brain and show that their pharmacology is characteristic of that seen in other species. The proportion of monoamine oxidase A and B expressed in the pig brain is similar to that seen in the human brain therefore, given the association between imidazoline I2 binding sites and monoamine oxidase, the pig may provide a more useful model for human imidazoline I2 binding sites than other species such as the rat.
Collapse
Affiliation(s)
- Neil J Anderson
- Department of Pharmacology, School of Medical Sciences, University Walk, Clifton, Bristol BS8 1TD, UK
| | | | | | | | | |
Collapse
|
27
|
Parker CA, Anderson NJ, Robinson ESJ, Price R, Tyacke RJ, Husbands SM, Dillon MP, Eglen RM, Hudson AL, Nutt DJ, Crump MP, Crosby J. Harmane and harmalan are bioactive components of classical clonidine-displacing substance. Biochemistry 2005; 43:16385-92. [PMID: 15610033 DOI: 10.1021/bi048584v] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Elucidation of the structure of the endogenous ligand(s) for imidazoline binding sites, clonidine-displacing substance (CDS), has been a major goal for many years. Crude CDS from bovine lung was purified by reverse-phase high-pressure liquid chromatography. Electrospray mass spectrometry (ESMS) and nuclear magnetic resonance ((1)H NMR) analysis revealed the presence of L-tryptophan and 1-carboxy-1-methyltetrahydro-beta-carboline in the active CDS extract. Competition radioligand binding studies, however, failed to show displacement of specific [(3)H]clonidine binding to rat brain membranes for either compound. Further purification of the bovine lung extract allowed the isolation of the beta-carbolines harmane and harmalan as confirmed by ESMS, (1)H NMR, and comparison with synthetic standards. Both compounds exhibited a high (nanomolar) affinity for both type 1 and type 2 imidazoline binding sites, and the synthetic standards were shown to coelute with the active classical CDS extracts. We therefore propose that the beta-carbolines harmane and harmalan represent active components of classical CDS. The identification of these compounds will allow us to establish clear physiological roles for CDS.
Collapse
|
28
|
MacInnes N, Duty S. Locomotor effects of imidazoline I2-site-specific ligands and monoamine oxidase inhibitors in rats with a unilateral 6-hydroxydopamine lesion of the nigrostriatal pathway. Br J Pharmacol 2004; 143:952-9. [PMID: 15545290 PMCID: PMC1575965 DOI: 10.1038/sj.bjp.0706019] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The present study examined the ability of the selective imidazoline I(2)-site ligands 2-(-2-benzofuranyl)-2-imidazoline (2-BFI) and 2-[4,5-dihydroimidaz-2-yl]-quinoline (BU224) and selected monoamine oxidase (MAO) inhibitors to evoke locomotor activity in rats bearing a lesion of the nigrostriatal pathway. Male Sprague-Dawley rats were injected with 12.5 microg 6-hydroxydopamine (6-OHDA) into the right median forebrain bundle to induce a unilateral lesion of the nigrostriatal tract. After 6 weeks, test drugs were administered either alone or in combination with L-DOPA (l-3,4-dihydroxyphenylamine) and the circling behaviour of animals was monitored as an index of anti-Parkinsonian activity. Intraperitoneal (i.p.) administration of the irreversible MAO-B inhibitor deprenyl (20 mg kg(-1)) or the imidazoline I(2)-site ligands BU224 (14 mg kg(-1)) and 2-BFI (7 and 14 mg kg(-1)) produced significant increases in ipsiversive rotations compared to vehicle controls totaling, at the highest respective doses tested, 521 +/-120, 131 +/- 37 and 92.5 +/- 16.3 net contraversive rotations in 30 (deprenyl) or 60 (BU224 and 2-BFI) min. In contrast, the reversible MAO-A inhibitor moclobemide (2.5-10 mg kg(-1)) and the reversible MAO-B inhibitor lazabemide (2.5-10 mg kg(-1)) failed to instigate significant rotational behaviour compared to vehicle. Coadministration of lazabemide (10 mg kg(-1)), moclobemide (10 mg kg(-1)) or 2-BFI (14 mg kg(-1)) with L-DOPA (20 mg kg(-1)) significantly increased either the duration or total number of contraversive rotations emitted over the testing period in comparison to L-DOPA alone. These data suggest that I(2)-specific ligands have dual effects in the 6-OHDA-lesioned rat model of Parkinson's disease; a first effect associated with an increase in activity in the intact hemisphere, probably via an increase in striatal dopamine content, and a secondary action which, through the previously documented inhibition of MAO-A and/or MAO-B, increases the availability of dopamine produced by L-DOPA.
Collapse
Affiliation(s)
- Nicholas MacInnes
- Wolfson Centre for Age-Related Diseases, G20 Wolfson Wing, Hodgkin Building, Guy's Campus, GKT School of Biomedical Sciences, King's College London, London SE1 1UL
| | - Susan Duty
- Wolfson Centre for Age-Related Diseases, G20 Wolfson Wing, Hodgkin Building, Guy's Campus, GKT School of Biomedical Sciences, King's College London, London SE1 1UL
- Author for correspondence:
| |
Collapse
|
29
|
Saczewski F, Hudson AL, Tyacke RJ, Nutt DJ, Man J, Tabin P, Saczewski J. 2-(4,5-Dihydro-1H-imidazol-2-yl)indazole (indazim) derivatives as selective I2 imidazoline receptor ligands. Eur J Pharm Sci 2003; 20:201-8. [PMID: 14550886 DOI: 10.1016/s0928-0987(03)00182-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of variously substituted 2-(4,5-dihydro-1H-imidazol-2-yl)indazoles 3a-j and 2-(4,5-dihydro-1H-imidazol-2-yl)-4,5,6,7-tetrahydroindazole 6 were prepared by the regiospecific heteroalkylation of corresponding indazoles 1a-k with 2-chloro-4,5-dihydroimidazole (2). Their affinity to imidazoline I(2) receptors and alpha(2)-adrenergic receptors was determined by radioligand binding assay carried out on P(2) membrane preparations obtained from rat whole brains. 4-Chloro-2-(4,5-dihydro-1H-imidazol-2-yl)indazole (3f, 4-Cl-indazim) showed a 3076-fold difference in affinity for the [(3)H]2BFI-labeled imidazoline I(2) receptors relative to the [(3)H]RX821001-labeled alpha(2)-adrenergic receptors. This highly selective compound should prove to be useful tool in further understanding the functions of the imidazoline I(2) receptors.
Collapse
Affiliation(s)
- F Saczewski
- Department of Chemical Technology of Drugs, Medical University of Gdańsk, Al Gen Hallera 107, 80-416 Gdańsk, Poland.
| | | | | | | | | | | | | |
Collapse
|
30
|
Robinson ESJ, Tyacke RJ, Nutt DJ, Hudson AL. Distribution of [(3)H]BU224, a selective imidazoline I(2) binding site ligand, in rat brain. Eur J Pharmacol 2002; 450:55-60. [PMID: 12176109 DOI: 10.1016/s0014-2999(02)02076-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BU224 (2-(4,5-dihydroimidaz-2-yl)-quinoline) is a selective imidazoline I(2) binding site ligand characterised in both competition binding assays and functional studies. However, in some studies, BU224 has been reported to have a different functional effect from that seen with another selective imidazoline I(2) binding site ligand 2-BFI (2-(2-benzofuranyl)-2-imidazoline). This effect may reflect differing efficacies of the ligands or a difference in their brain distribution. The present study has investigated the distribution of the tritiated form of BU224 in rat brain and correlated this distribution with other imidazoline I(2) binding site ligands, [(3)H]idazoxan and [(3)H]2-BFI. Saturation studies revealed binding of [(3)H]BU224 was of high affinity and saturable. The central distribution of [(3)H]BU224 was similar to that previous reported for imidazoline I(2) binding site in rat brain. Autoradiography revealed that the highest levels of binding were in the arcuate nucleus, interpeduncular nucleus, area postrema, pineal gland and ependymal cell layer lining the ventricles. Correlation analysis of the binding distribution with our previous published studies revealed a highly significant correlation between [(3)H]BU224 and both [(3)H]idazoxan (r=0.94) and [(3)H]2-BFI (r=0.96). These data indicate [(3)H]BU224 labels the same population of imidazoline I(2) binding site in rat brain as seen with [(3)H]idazoxan and [(3)H]2-BFI. Therefore, the differences in functional effects observed with these compounds may reflect agonist and antagonist properties.
Collapse
Affiliation(s)
- Emma S J Robinson
- Psychopharmacology Unit, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | | | | | | |
Collapse
|
31
|
Husbands SM, Glennon RA, Gorgerat S, Gough R, Tyacke R, Crosby J, Nutt DJ, Lewis JW, Hudson AL. beta-carboline binding to imidazoline receptors. Drug Alcohol Depend 2001; 64:203-8. [PMID: 11543990 DOI: 10.1016/s0376-8716(01)00123-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A series of beta-carbolines were prepared and their affinities for imidazoline (I(1) and I(2)) sites evaluated. Selected compounds were also examined at alpha(2)-adrenoceptors. Some of the beta-carbolines were found to bind with high affinity to I(2)-sites and this affinity was dependent on both the planarity of the molecule and the presence of the aryl ring substituents. Good I(1)-affinity was observed with two of the compounds but none of the tested compounds bound to alpha(2)-adrenoceptors. The hallucinogenic properties of beta-carbolines have been linked to activity at 5-HT receptors, in particular 5-HT(2), however, it is apparent from this study that many of these compounds display substantially higher affinity for the imidazoline sites. This finding, and those showing modulation of some behavioural effects of morphine by I(2)-ligands, suggests that imidazoline sites may be interesting new targets in drug abuse research.
Collapse
Affiliation(s)
- S M Husbands
- Department of Pharmacy and Pharmacology, University of Bath, BA2 7AY, Bath, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Callado LF, Maeztu AI, Ballesteros J, Gutiérrez M, Meana JJ. Differential [(3)H]idazoxan and [(3)H]2-(2-benzofuranyl)-2-imidazoline (2-BFI) binding to imidazoline I(2) receptors in human postmortem frontal cortex. Eur J Pharmacol 2001; 423:109-14. [PMID: 11448473 DOI: 10.1016/s0014-2999(01)01097-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
[(3)H]2-(2-benzofuranyl)-2-imidazoline (2-BFI) and [(3)H]idazoxan are the most used tools to characterise imidazoline I(2) receptors. We evaluated the binding of both radioligands to human postmortem frontal cortex membranes. Saturation binding analyses revealed that [(3)H]idazoxan (in the presence of 2 microM efaroxan to avoid radioligand binding to alpha(2)-adrenoceptors and imidazoline I(1) receptors) and [(3)H]2-BFI bound with high affinity to an apparent single population of sites. However, in competition studies whereas [(3)H]idazoxan (10 nM) binding was displaced monophasically by idazoxan and 2-BFI, both drugs displayed biphasic curves for [(3)H]2-BFI (1 nM). The proportion of the low-affinity binding site increased from 17% to 25% when 10 nM [(3)H]2-BFI was displaced by idazoxan. Amiloride inhibited [(3)H]2-BFI (10 nM) binding with low affinity and in a monophasic way. These data indicate that [(3)H]2-BFI recognises in human postmortem brain membranes a second binding site different from the imidazoline I(2) receptors labelled by [(3)H]idazoxan.
Collapse
Affiliation(s)
- L F Callado
- Department of Pharmacology, University of the Basque Country, E-48940 Leioa, Bizkaia, Spain.
| | | | | | | | | |
Collapse
|
33
|
Ulibarri I, Soto J, Ruiz J, Ballesteros J, Jaúregui JV, Meana JJ. I2-imidazoline receptors in platelets of patients with Parkinson's disease and Alzheimer's type dementia. Ann N Y Acad Sci 1999; 881:199-202. [PMID: 10415916 DOI: 10.1111/j.1749-6632.1999.tb09360.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- I Ulibarri
- Department of Pharmacology, University of the Basque Country, Bizkaia, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Major depression, opioid addiction, neurodegenerative diseases, and glial tumors are associated with disturbances of imidazoline receptors (IR) in the human brain. In depression, the level of a 45-kD IR protein (putative I1-IR) is increased in the brain of suicide victims (51%) and in platelets of depressed patients (40%). The density of platelet I1-IR ([125I]-p-iodoclonidine binding) is also increased in depression (135%). The 29/30-kD IR protein (putative I2B-IR) is downregulated (19%) in suicide victims in parallel with a reduction (40%) in the density of I2B-IR ([3H]idazoxan binding). Antidepressant drugs induce downregulation of 45-kD IR protein and I1-sites in platelets of depressed patients and upregulation of I2-sites in rat brain. The densities of I2B-IR and the related 29/30-kD IR protein are decreased (39% and 28%) in the brain of heroin addicts. The density of I2B-IR is increased in Alzheimer's disease (63%) and decreased in Huntington's disease (56%). Brain I2B-IR is not altered in Parkinson's disease. The level of I2-IR in glial tumors is increased (two-fivefold) in parallel with the abundance of the related 29/30-kD IR protein (39%), whereas the level of 45-kD IR protein is decreased (39%). The possible functional relevance of these findings in the context of the pathogenesis of these disorders remains to be elucidated.
Collapse
Affiliation(s)
- J A García-Sevilla
- Institute of Neurobiology Ramón y Cajal/CSIC, Department of Biology, University of the Balearic Islands, Palma de Mallorca, Spain
| | | | | |
Collapse
|
35
|
Vauquelin G, De Backer JP, Ladure P, Flamez A. Identification of I1 and I2 imidazoline receptors in striatum membranes from different species. Ann N Y Acad Sci 1999; 881:135-43. [PMID: 10415909 DOI: 10.1111/j.1749-6632.1999.tb09353.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The alpha 2-adrenergic agonist [3H]clonidine and antagonist [3H]idazoxan also label I1 and I2 imidazoline receptors in striatum membranes. They are investigated here in striata from the dog, rat, mouse, rabbit, calf, monkey, and human. I1 receptors were barely detected in the dog, rat, and mouse and only further examined by competition binding experiments in calf, rabbit, and human. I2 receptors were further examined in all species. The centrally acting vasodilators clonidine and rilmenidine were more potent than moxonidine at the I1 receptors. They displayed low potency for the I2 receptors in all species except the rat. In all species examined, the nonsubstituted imidazoline derivatives idazoxan and RX801077 displayed high affinity for the I1 and I2 receptors. Conversely, both stereoisomers of the alkoxy-substituted imidazoline-derivative efaroxan displayed low affinity. The matching binding characteristics of these compounds further stress the structural similarity of the ligand binding sites of I1 and I2 receptors.
Collapse
Affiliation(s)
- G Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Sint-Genesius-Rode, Belgium
| | | | | | | |
Collapse
|
36
|
Eglen RM, Hudson AL, Kendall DA, Nutt DJ, Morgan NG, Wilson VG, Dillon MP. 'Seeing through a glass darkly': casting light on imidazoline 'I' sites. Trends Pharmacol Sci 1998; 19:381-90. [PMID: 9786027 DOI: 10.1016/s0165-6147(98)01244-9] [Citation(s) in RCA: 195] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although imidazoline sites have been the subject of research for several years, there is still controversy about their structure, diversity and physiology. The I1 site is thought to exist principally as a binding site and is widely purported to play a role in controlling systemic blood pressure, although this is still unclear. The majority of I2 sites are widely accepted as being allosteric sites on monoamine oxidase; however, even with selective ligands, their exact function remains to be determined. A putative I3 site modulates insulin secretion and could represent the first functional site to be pharmacologically defined with selective agonists and antagonists. The structure and relevance of the proposed endogenous ligand 'clonidine-displacing substance' remains elusive. A potential candidate for this substance is agmatine; however, although it is capable of displacing bound clonidine from imidazoline sites, it lacks the functionality ascribed to the clonidine-displacing substance. In this review, Richard M. Eglen and colleagues assess our knowledge of imidazoline sites in the light of recent data.
Collapse
Affiliation(s)
- R M Eglen
- Medicinal Chemistry Department, Roche Bioscience, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Lione LA, Nutt DJ, Hudson AL. Characterisation and localisation of [3H]2-(2-benzofuranyl)-2-imidazoline binding in rat brain: a selective ligand for imidazoline I2 receptors. Eur J Pharmacol 1998; 353:123-35. [PMID: 9721049 DOI: 10.1016/s0014-2999(98)00389-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In rat whole brain homogenates, saturation binding analysis revealed that both [3H]2-BFI (2-(2-benzofuranyl)-2-imidazoline) and [3H]idazoxan (in the presence of 5 microM rauwolscine) bound with high affinity to an apparent single population of sites. However, the Kd for [3H]2-BFI (1.74+/-0.14 nM) was significantly (P < 0.01) less than that for [3H]idazoxan (10.4+/-2.68 nM). In competition studies idazoxan, 2-BFI, BU224 (2-(4,5-dihydroimidaz-2-yl)-quinoline), amiloride and guanabenz displayed high affinity (Ki values = 7.32, 1.71, 2.08, 21.80 and 14.90 nM, respectively) for 70-80% of sites, and low microM affinity for the remaining 20-30% of sites labelled by [3H]2-BFI. In contrast, several alpha2-adrenoceptor, imidazoline I1 receptor and histamine receptor ligands exhibited only micromolar affinity for the [3H]2-BFI labelled site. Quantitative receptor autoradiography revealed high binding by [3H]2-BFI to discrete brain nuclei, notably the area postrema, interpeduncular nucleus, arcuate nucleus, mammillary peduncle, ependyma and pineal gland. These data indicate that [3H]2-BFI recognises imidazoline I2 receptors in rat brain with higher affinity and selectivity than [3H]idazoxan and thus represents a superior radioligand to [3H]idazoxan for the study of imidazoline I2 receptors.
Collapse
Affiliation(s)
- L A Lione
- Psychopharmacology Unit, School of Medical Sciences, University of Bristol, UK
| | | | | |
Collapse
|
38
|
Abstract
There is growing evidence that noradrenergic inputs to the prefrontal cortex (PFC) play an important role in regulating its function. This paper reviews the pharmacological control of noradrenaline (NA) release in this region, with particular reference to our studies using brain microdialysis, and also describes how NA levels are modulated by antidepressant and antipsychotic drugs. The suggestion that atypical antipsychotics such as clozapine and risperidone may produce clinical benefits by their ability to increase NA release is discussed. Finally, a new class of drugs, which show selectivity for imidazoline receptors is described. These compounds are shown to similarly increase extracellular NA in the PFC. Their potential utility as clinical treatments is discussed.
Collapse
Affiliation(s)
- D J Nutt
- Psychopharmacology Unit, School of Medical Sciences, University of Bristol, UK.
| | | | | | | |
Collapse
|