1
|
Berro LF, Rowlett JK, Platt DM. GABAergic compounds for the treatment of alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:383-399. [PMID: 39523061 DOI: 10.1016/bs.irn.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Decades of research have implicated the gamma-aminobutyric acid (GABA)ergic system as one of the main mediators of the behavioral effects of alcohol. Of importance, the addiction-related effects of alcohol also have been shown to be mediated in part by GABAergic systems, raising the possibility that pharmacotherapies targeting GABAergic receptors may be promising candidates for the treatment of alcohol use disorder (AUD). Alcohol modulates the activity of GABAA and GABAB receptors, and studies show that compounds targeting some of those receptors may decrease the addiction-related behavioral effects of alcohol. Specifically, drugs that share similar pharmacological properties with alcohol, such as positive allosteric modulators (PAMs) of GABAA and GABAB receptors, have been proposed as substitution therapies for AUD. Available evidence also suggests that negative allosteric modulators (NAMs) of GABAergic receptors may be potential therapeutics for AUD, although this effect is selective for specific receptor subtypes. Therefore, this Chapter reviews the available evidence on the use of GABAergic compounds for the treatment of AUD. Several GABAA and GABAB ligands show promising results, with a particularly positive therapeutic profile demonstrated for α5GABAA receptor NAMs, α4/6δGABAA receptor modulators (both positive and negative, including neurosteroids), and GABAB receptor PAMs. As newer and better GABAergic compounds become available, future research should focus on understanding how these ligands can modulate different clinical symptoms of AUD, with potential new areas of research encompassing alcohol withdrawal syndrome and AUD-related insomnia.
Collapse
Affiliation(s)
- Laís F Berro
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS, United States.
| | - James K Rowlett
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS, United States
| | - Donna M Platt
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
2
|
Pareek T, Overton JS, Nguyen LT, Rahman MT, Sharmin D, Cook JM, Platt DM. Modeling cue-exposure therapy for alcohol use disorder in rhesus monkeys: Effects of putative cognitive enhancers. Drug Alcohol Depend 2023; 243:109735. [PMID: 36549228 PMCID: PMC9852009 DOI: 10.1016/j.drugalcdep.2022.109735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cue-exposure therapy (CET) is an effective approach for anxiety-related disorders, but its effectiveness for substance use disorders is less clear. One potential means of improving CET outcomes is to include a cognitive-enhancing pharmacotherapy. This study evaluated d-cycloserine (DCS) and RY-023, putative cognitive enhancers targeting glutamate and GABA systems, respectively, in a monkey model of CET for alcohol use disorder. METHODS Male rhesus monkeys (n = 4) underwent multiple cycles of the CET procedure. During baseline (Phase 1), monkeys self-administered an ethanol solution under a fixed-ratio schedule and limited access conditions such that every 5th response in a 3-h session resulted in 30-s access to a drinking spout and a change in ethanol-paired cue lights from white to red. Behavior then was extinguished (Phase 2) by omitting the ethanol solution yet retaining the ethanol-paired stimulus lights. Monkeys also received injections of vehicle, DCS (3 mg/kg), a partial agonist at the glycine modulatory site on glutamatergic NMDA receptors, or the α5GABAA receptor-selective inverse agonist RY-023 (0.03 or 0.3 mg/kg). Once responding declined, monkeys underwent a cue reactivity test (Phase 3), and then returned to self-administration the following day to assess reacquisition (Phase 4). RESULTS Through multiple cycles, self-administration remained stable. Compared to vehicle, DCS facilitated extinction of ethanol seeking (Phase 2) and delayed reacquisition of ethanol self-administration (Phase 4). In contrast, RY-023 facilitated extinction (Phase 2) and reduced cue reactivity (Phase 3). CONCLUSIONS Adjunctive pharmacotherapy can improve CET outcomes, but the choice of pharmacotherapy should be dependent on the outcome of interest.
Collapse
Affiliation(s)
- Tanya Pareek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - John S Overton
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Luat T Nguyen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Md Toufiqur Rahman
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Donna M Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
3
|
Szabó G, Éliás O, Erdélyi P, Potor A, Túrós GI, Károlyi BI, Varró G, Vaskó ÁG, Bata I, Kapus GL, Dohányos Z, Bobok AÁ, Fodor L, Thán M, Vastag M, Komlódi Z, Soukupné Kedves RÉ, Makó É, Süveges B, Greiner I. Multiparameter Optimization of Naphthyridine Derivatives as Selective α5-GABA A Receptor Negative Allosteric Modulators. J Med Chem 2022; 65:7876-7895. [PMID: 35584373 DOI: 10.1021/acs.jmedchem.2c00414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The discovery and characterization of novel naphthyridine derivatives with selective α5-GABAAR negative allosteric modulator (NAM) activity are disclosed. Utilizing a scaffold-hopping strategy, fused [6 + 6] bicyclic scaffolds were designed and synthesized. Among these, 1,6-naphthyridinones were identified as potent and selective α5-GABAAR NAMs with metabolic stability, cardiac safety, and beneficial intellectual property (IP) issues. Relocation of the oxo acceptor function and subsequent modulation of the physicochemical properties resulted in novel 1,6-naphthyridines with improved profile, combining good potency, selectivity, ADME, and safety properties. Besides this, compound 20, having the most balanced profile, provided in vivo proof of concept (POC) for the new scaffold in two animal models of cognitive impairment associated with schizophrenia (CIAS).
Collapse
Affiliation(s)
- György Szabó
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Olivér Éliás
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Péter Erdélyi
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Attila Potor
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - György I Túrós
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | | | - Gábor Varró
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Ágnes Gy Vaskó
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Imre Bata
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Gábor L Kapus
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Zoltán Dohányos
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Amrita Á Bobok
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - László Fodor
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Márta Thán
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Mónika Vastag
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | - Zsolt Komlódi
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | | | - Éva Makó
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| | | | - István Greiner
- Gedeon Richter Plc., 19-21. Gyömrői út, Budapest, 1103 Hungary
| |
Collapse
|
4
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
5
|
The Case for Clinical Trials with Novel GABAergic Drugs in Diabetes Mellitus and Obesity. Life (Basel) 2022; 12:life12020322. [PMID: 35207609 PMCID: PMC8876029 DOI: 10.3390/life12020322] [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: 01/22/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity and diabetes mellitus have become the surprising menaces of relative economic well-being worldwide. Gamma amino butyric acid (GABA) has a prominent role in the control of blood glucose, energy homeostasis as well as food intake at several levels of regulation. The effects of GABA in the body are exerted through ionotropic GABAA and metabotropic GABAB receptors. This treatise will focus on the pharmacologic targeting of GABAA receptors to reap beneficial therapeutic effects in diabetes mellitus and obesity. A new crop of drugs selectively targeting GABAA receptors has been under investigation for efficacy in stroke recovery and cognitive deficits associated with schizophrenia. Although these trials have produced mixed outcomes the compounds are safe to use in humans. Preclinical evidence is summarized here to support the rationale of testing some of these compounds in diabetic patients receiving insulin in order to achieve better control of blood glucose levels and to combat the decline of cognitive performance. Potential therapeutic benefits could be achieved (i) By resetting the hypoglycemic counter-regulatory response; (ii) Through trophic actions on pancreatic islets, (iii) By the mobilization of antioxidant defence mechanisms in the brain. Furthermore, preclinical proof-of-concept work, as well as clinical trials that apply the novel GABAA compounds in eating disorders, e.g., olanzapine-induced weight-gain, also appear warranted.
Collapse
|
6
|
Modulation of relapse-like drinking in male Sprague-Dawley rats by ligands targeting the α5GABA A receptor. Neuropharmacology 2021; 199:108785. [PMID: 34509495 DOI: 10.1016/j.neuropharm.2021.108785] [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: 04/12/2021] [Revised: 08/26/2021] [Accepted: 09/04/2021] [Indexed: 11/20/2022]
Abstract
Preclinical evidence suggests a key role for GABAA receptors containing the α5 subunit (i.e., α5GABAA receptors) in the abuse-related effects of alcohol, including the reinforcing and discriminative stimulus effects, as well as cue-induced alcohol-seeking behavior. However, the contribution of this GABAA receptor subtype to relapse-like drinking behavior remains unknown. The present study evaluated the capacity of ligands targeting α5GABAA receptors to modulate the alcohol deprivation effect (ADE), a model of relapse-like drinking. Groups of Sprague-Dawley rats underwent repeated cycles of long-term access to alcohol solutions (5%, 10%, 20% v/v) and water in the home cage followed by water only deprivation periods. Upon evidence that the ADE could be reliably expressed across cycles, drug treatment was initiated. One group received the α5GABAA receptor-preferring agonist QH-ii-066 and the other group received the α5GABAA receptor-selective inverse agonist L-655,708. At the end of ADE testing, rats underwent testing in the elevated zero maze under vehicle or L-655,708 treatment for assessment of anxiety-like behavior. The ADE was reliably expressed across repeated cycles of alcohol access/deprivation in a subset of rats. Low doses of QH-ii-066 enhanced expression of the ADE; whereas, L-655,708 dose-dependently inhibited expression of the ADE. L-655,708 did not engender anxiogenic effects in the elevated zero maze under the conditions evaluated. These findings suggest a key role for α5GABAA receptor mechanisms in relapse-like drinking. Moreover, they suggest that α5GABAA receptors may represent a novel pharmacological target for the development of medications to prevent or reduce alcohol relapse.
Collapse
|
7
|
Ghit A, Assal D, Al-Shami AS, Hussein DEE. GABA A receptors: structure, function, pharmacology, and related disorders. J Genet Eng Biotechnol 2021; 19:123. [PMID: 34417930 PMCID: PMC8380214 DOI: 10.1186/s43141-021-00224-0] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/08/2021] [Indexed: 02/03/2023]
Abstract
Background γ-Aminobutyric acid sub-type A receptors (GABAARs) are the most prominent inhibitory neurotransmitter receptors in the CNS. They are a family of ligand-gated ion channel with significant physiological and therapeutic implications. Main body GABAARs are heteropentamers formed from a selection of 19 subunits: six α (alpha1-6), three β (beta1-3), three γ (gamma1-3), three ρ (rho1-3), and one each of the δ (delta), ε (epsilon), π (pi), and θ (theta) which result in the production of a considerable number of receptor isoforms. Each isoform exhibits distinct pharmacological and physiological properties. However, the majority of GABAARs are composed of two α subunits, two β subunits, and one γ subunit arranged as γ2β2α1β2α1 counterclockwise around the center. The mature receptor has a central chloride ion channel gated by GABA neurotransmitter and modulated by a variety of different drugs. Changes in GABA synthesis or release may have a significant effect on normal brain function. Furthermore, The molecular interactions and pharmacological effects caused by drugs are extremely complex. This is due to the structural heterogeneity of the receptors, and the existence of multiple allosteric binding sites as well as a wide range of ligands that can bind to them. Notably, dysfunction of the GABAergic system contributes to the development of several diseases. Therefore, understanding the relationship between GABAA receptor deficits and CNS disorders thus has a significant impact on the discovery of disease pathogenesis and drug development. Conclusion To date, few reviews have discussed GABAA receptors in detail. Accordingly, this review aims to summarize the current understanding of the structural, physiological, and pharmacological properties of GABAARs, as well as shedding light on the most common associated disorders.
Collapse
Affiliation(s)
- Amr Ghit
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy. .,Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Dina Assal
- Department of Biotechnology, American University in Cairo (AUC), Cairo, Egypt
| | - Ahmed S Al-Shami
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.,Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Diaa Eldin E Hussein
- Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Port of Alexandria, Alexandria, Egypt
| |
Collapse
|
8
|
Orser BA. Anesthesiology: Resetting Our Sights on Long-term Outcomes: The 2020 John W. Severinghaus Lecture on Translational Science. Anesthesiology 2021; 135:18-30. [PMID: 33901279 DOI: 10.1097/aln.0000000000003798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Anesthesiologists have worked relentlessly to improve intraoperative anesthesia care. They are now well positioned to expand their horizons and address many of the longer-term adverse consequences of anesthesia and surgery. Perioperative neurocognitive disorders, chronic postoperative pain, and opioid misuse are not inevitable adverse outcomes; rather, they are preventable and treatable conditions that deserve attention. The author's research team has investigated why patients experience new cognitive deficits after anesthesia and surgery. Their animal studies have shown that anesthetic drugs trigger overactivity of "memory-blocking receptors" that persists after the drugs are eliminated, and they have discovered new strategies to preserve brain function by repurposing available drugs and developing novel therapeutics that inhibit these receptors. Clinical trials are in progress to examine the cognitive outcomes of such strategies. This work is just one example of how anesthesiologists are advancing science with the goal of improving the lives of patients.
Collapse
|
9
|
Basmisanil, a highly selective GABA A-α5 negative allosteric modulator: preclinical pharmacology and demonstration of functional target engagement in man. Sci Rep 2021; 11:7700. [PMID: 33833333 PMCID: PMC8032764 DOI: 10.1038/s41598-021-87307-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
GABAA-α5 subunit-containing receptors have been shown to play a key modulatory role in cognition and represent a promising drug target for cognitive dysfunction, as well as other disorders. Here we report on the preclinical and early clinical profile of a novel GABAA-α5 selective negative allosteric modulator (NAM), basmisanil, which progressed into Phase II trials for intellectual disability in Down syndrome and cognitive impairment associated with schizophrenia. Preclinical pharmacology studies showed that basmisanil is the most selective GABAA-α5 receptor NAM described so far. Basmisanil bound to recombinant human GABAA-α5 receptors with 5 nM affinity and more than 90-fold selectivity versus α1, α2, and α3 subunit-containing receptors. Moreover, basmisanil inhibited GABA-induced currents at GABAA-α5 yet had little or no effect at the other receptor subtypes. An in vivo occupancy study in rats showed dose-dependent target engagement and was utilized to establish the plasma exposure to receptor occupancy relationship. At estimated receptor occupancies between 30 and 65% basmisanil attenuated diazepam-induced spatial learning impairment in rats (Morris water maze), improved executive function in non-human primates (object retrieval), without showing anxiogenic or proconvulsant effects in rats. During the Phase I open-label studies, basmisanil showed good safety and tolerability in healthy volunteers at maximum GABAA-α5 receptor occupancy as confirmed by PET analysis with the tracer [11C]-Ro 15-4513. An exploratory EEG study provided evidence for functional activity of basmisanil in human brain. Therefore, these preclinical and early clinical studies show that basmisanil has an ideal profile to investigate potential clinical benefits of GABAA-α5 receptor negative modulation.
Collapse
|
10
|
Bhagat K, Singh JV, Pagare PP, Kumar N, Sharma A, Kaur G, Kinarivala N, Gandu S, Singh H, Sharma S, Bedi PMS. Rational approaches for the design of various GABA modulators and their clinical progression. Mol Divers 2021; 25:551-601. [PMID: 32170466 PMCID: PMC8422677 DOI: 10.1007/s11030-020-10068-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/28/2020] [Indexed: 12/20/2022]
Abstract
GABA (γ-amino butyric acid) is an important inhibitory neurotransmitter in the central nervous system. Attenuation of GABAergic neurotransmission plays an important role in the etiology of several neurological disorders including epilepsy, Alzheimer's disease, Huntington's chorea, migraine, Parkinson's disease, neuropathic pain, and depression. Increase in the GABAergic activity may be achieved through direct agonism at the GABAA receptors, inhibition of enzymatic breakdown of GABA, or by inhibition of the GABA transport proteins (GATs). These functionalities make GABA receptor modulators and GATs attractive drug targets in brain disorders associated with decreased GABA activity. There have been several reports of development of GABA modulators (GABA receptors, GABA transporters, and GABAergic enzyme inhibitors) in the past decade. Therefore, the focus of the present review is to provide an overview on various design strategies and synthetic approaches toward developing GABA modulators. Furthermore, mechanistic insights, structure-activity relationships, and molecular modeling inputs for the biologically active derivatives have also been discussed. Summary of the advances made over the past few years in the clinical translation and development of GABA receptor modulators is also provided. This compilation will be of great interest to the researchers working in the field of neuroscience. From the light of detailed literature, it can be concluded that numerous molecules have displayed significant results and their promising potential, clearly placing them ahead as potential future drug candidates.
Collapse
Affiliation(s)
- Kavita Bhagat
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India
| | - Jatinder V Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India
| | - Piyusha P Pagare
- Department of Medicinal Chemistry, School of Pharmacy and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Nitish Kumar
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India
| | - Anchal Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India
| | - Gurinder Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India
| | - Nihar Kinarivala
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Srinivasa Gandu
- Department of Cell Biology and Neuroscience, Cell and Development Biology Graduate Program, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India.
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India.
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY, 10065, USA.
| | - Preet Mohinder S Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, PB, 143005, India.
| |
Collapse
|
11
|
McGinnity CJ, Riaño Barros DA, Hinz R, Myers JF, Yaakub SN, Thyssen C, Heckemann RA, de Tisi J, Duncan JS, Sander JW, Lingford-Hughes A, Koepp MJ, Hammers A. Αlpha 5 subunit-containing GABA A receptors in temporal lobe epilepsy with normal MRI. Brain Commun 2021; 3:fcaa190. [PMID: 33501420 PMCID: PMC7811756 DOI: 10.1093/braincomms/fcaa190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 09/06/2020] [Accepted: 09/24/2020] [Indexed: 01/08/2023] Open
Abstract
GABAA receptors containing the α5 subunit mediate tonic inhibition and are widely expressed in the limbic system. In animals, activation of α5-containing receptors impairs hippocampus-dependent memory. Temporal lobe epilepsy is associated with memory impairments related to neuron loss and other changes. The less selective PET ligand [11C]flumazenil has revealed reductions in GABAA receptors. The hypothesis that α5 subunit receptor alterations are present in temporal lobe epilepsy and could contribute to impaired memory is untested. We compared α5 subunit availability between individuals with temporal lobe epilepsy and normal structural MRI ('MRI-negative') and healthy controls, and interrogated the relationship between α5 subunit availability and episodic memory performance, in a cross-sectional study. Twenty-three healthy male controls (median ± interquartile age 49 ± 13 years) and 11 individuals with MRI-negative temporal lobe epilepsy (seven males; 40 ± 8) had a 90-min PET scan after bolus injection of [11C]Ro15-4513, with arterial blood sampling and metabolite correction. All those with epilepsy and six controls completed the Adult Memory and Information Processing Battery on the scanning day. 'Bandpass' exponential spectral analyses were used to calculate volumes of distribution separately for the fast component [V F; dominated by signal from α1 (α2, α3)-containing receptors] and the slow component (V S; dominated by signal from α5-containing receptors). We made voxel-by-voxel comparisons between: the epilepsy and control groups; each individual case versus the controls. We obtained parametric maps of V F and V S measures from a single bolus injection of [11C]Ro15-4513. The epilepsy group had higher V S in anterior medial and lateral aspects of the temporal lobes, the anterior cingulate gyri, the presumed area tempestas (piriform cortex) and the insulae, in addition to increases of ∼24% and ∼26% in the ipsilateral and contralateral hippocampal areas (P < 0.004). This was associated with reduced V F:V S ratios within the same areas (P < 0.009). Comparisons of V S for each individual with epilepsy versus controls did not consistently lateralize the epileptogenic lobe. Memory scores were significantly lower in the epilepsy group than in controls (mean ± standard deviation -0.4 ± 1.0 versus 0.7 ± 0.3; P = 0.02). In individuals with epilepsy, hippocampal V S did not correlate with memory performance on the Adult Memory and Information Processing Battery. They had reduced V F in the hippocampal area, which was significant ipsilaterally (P = 0.03), as expected from [11C]flumazenil studies. We found increased tonic inhibitory neurotransmission in our cohort of MRI-negative temporal lobe epilepsy who also had co-morbid memory impairments. Our findings are consistent with a subunit shift from α1/2/3 to α5 in MRI-negative temporal lobe epilepsy.
Collapse
Affiliation(s)
- Colm J McGinnity
- Centre for Neuroscience, Department of Medicine, Imperial College London, London W12 0NN, UK
- MRC Clinical Sciences Centre, Hammersmith Hospital, London W12 0NN, UK
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Daniela A Riaño Barros
- Centre for Neuroscience, Department of Medicine, Imperial College London, London W12 0NN, UK
- MRC Clinical Sciences Centre, Hammersmith Hospital, London W12 0NN, UK
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester M20 3LJ, UK
| | - James F Myers
- Centre for Neuroscience, Department of Medicine, Imperial College London, London W12 0NN, UK
| | - Siti N Yaakub
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Charlotte Thyssen
- Medical Image and Signal Processing (MEDISIP), Department of Electronics and Information Systems, Faculty of Engineering and Architecture, Ghent University, 9000 Ghent, Belgium
| | - Rolf A Heckemann
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Jane de Tisi
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK, and Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
| | - John S Duncan
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK, and Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
| | - Josemir W Sander
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK, and Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede 2103SW, The Netherlands
| | - Anne Lingford-Hughes
- Neuropsychopharmacology Unit, Centre for Psychiatry, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Matthias J Koepp
- NIHR University College London Hospitals Biomedical Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK, and Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
| | - Alexander Hammers
- Centre for Neuroscience, Department of Medicine, Imperial College London, London W12 0NN, UK
- MRC Clinical Sciences Centre, Hammersmith Hospital, London W12 0NN, UK
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EH, UK
- Neurodis Foundation, CERMEP, Imagerie du Vivant, 69003 Lyon, France
| |
Collapse
|
12
|
Lewis A, Beresford A, Chambers MS, Clark G, Hartley DC, Hirst KL, Higashino M, Kawahadara S, Nakanishi M, Saito T, Imagawa A, Habashita H, Maidment S, Macleod AM, Owens AP, Rae A, Rouse C, Wishart G. Discovery of ONO-8590580: A novel, potent and selective GABA A α 5 negative allosteric modulator for the treatment of cognitive disorders. Bioorg Med Chem Lett 2020; 30:127536. [PMID: 32898695 DOI: 10.1016/j.bmcl.2020.127536] [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: 05/11/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
The identification and SAR development of a series of negative allosteric modulators of the GABAA α5 receptor is described. This novel series of compounds was optimised to provide analogues with high GABAA α5 binding affinity, high α5 negative allosteric modulatory activity, good functional subtype selectivity and low microsomal turnover, culminating in identification of ONO-8590580.
Collapse
Affiliation(s)
- A Lewis
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - A Beresford
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - M S Chambers
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - G Clark
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - D C Hartley
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - K L Hirst
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - M Higashino
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - S Kawahadara
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - M Nakanishi
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - T Saito
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - A Imagawa
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - H Habashita
- Ono Pharmaceutical Co. Ltd., Minase Research Institute, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - S Maidment
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - A M Macleod
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - A P Owens
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - A Rae
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom.
| | - C Rouse
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| | - G Wishart
- Charles River Discovery Research Services, Chesterford Research Park, Little Chesterford, Essex CB10 1XL, United Kingdom
| |
Collapse
|
13
|
Vinnakota C, Govindpani K, Tate WP, Peppercorn K, Anekal PV, Waldvogel HJ, Faull RLM, Kwakowsky A. An 5 GABAA Receptor Inverse Agonist, 5IA, Attenuates Amyloid Beta-Induced Neuronal Death in Mouse Hippocampal Cultures. Int J Mol Sci 2020; 21:ijms21093284. [PMID: 32384683 PMCID: PMC7247548 DOI: 10.3390/ijms21093284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder for which no cognition-restoring therapies exist. Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain. Increasing evidence suggests a remodeling of the GABAergic system in AD, which might represent an important therapeutic target. An inverse agonist of α5 subunit-containing GABAA receptors (α5GABAARs), 3-(5-Methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3–a]phthalazine (α5IA) has cognition-enhancing properties. This study aimed to characterize the effects of α5IA on amyloid beta (Aβ1–42)-induced molecular and cellular changes. Mouse primary hippocampal cultures were exposed to either Aβ1-42 alone, or α5IA alone, α5IA with Aβ1–42 or vehicle alone, and changes in cell viability and mRNA expression of several GABAergic signaling components were assessed. Treatment with 100 nM of α5IA reduced Aβ1–42-induced cell loss by 23.8% (p < 0.0001) after 6 h and by 17.3% after 5 days of treatment (p < 0.0001). Furthermore, we observed an Aβ1-42-induced increase in ambient GABA levels, as well as upregulated mRNA expression of the GABAAR α2,α5,β2/3 subunits and the GABABR R1 and R2 subunits. Such changes in GABARs expression could potentially disrupt inhibitory neurotransmission and normal network activity. Treatment with α5IA restored Aβ1-42-induced changes in the expression of α5GABAARs. In summary, this compound might hold neuroprotective potential and represent a new therapeutic avenue for AD.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Warren Perry Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Praju Vikas Anekal
- Biomedical Imaging Research Unit, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand;
| | - Henry John Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Richard Lewis Maxwell Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
- Correspondence: ; Tel.: +64-9923-9346
| |
Collapse
|
14
|
Matthey A, Daali Y, Curtin F, Poncet A, Desmeules J, Besson M. GABAergic modulation of secondary hyperalgesia: A randomized controlled 4-way crossover trial with the α2-subunit preferring GABA positive allosteric modulator, N-desmethyl-clobazam in healthy volunteers. Eur J Pain 2020; 24:1094-1106. [PMID: 32171038 DOI: 10.1002/ejp.1554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 11/10/2022]
Abstract
The antihyperalgesic and sedative effects of the α2-subunit preferring GABAA positive allosteric modulator (GAM), N-desmethyl-clobazam (NDMC), 20 and 60 mg, were assessed in a randomized, placebo and active-controlled (clonazepam 1,5 mg), 4-way crossover study, in healthy volunteers, using the ultraviolet B-induced experimental pain model. Single (20, 40, 60 mg) and repeated doses (20 mg over 15 days) of NDMC pharmacokinetics were evaluated. Thirty-two subjects participated in the study. Primary outcome parameter was maximal change in the area of cutaneous UVB irradiation-induced secondary hyperalgesia (ASH). ASH decreased under all treatments. Mean (SD) relative change was 79 (22)%, 83 (24)%, 77 (30)% and 92 (16)% for placebo, NDMC20, NDMC60 and clonazepam, respectively. Neither absolute change nor relative change in ASH was significantly different between NDMC60 and placebo (mean difference = 2.3 cm2 [95% CI 4.0-8.5], p = .462 and 0.4% [-11.9 to 12.6], p = .952, respectively). An overall treatment effect was found on level of sedation. Compared to placebo, sedation was higher under clonazepam (mean difference = 39 mm [30-49] on a visual analogue scale, p < .001) while NDMC was free of sedative effect. NDMC pharmacokinetics after single doses showed poor absorption, but was linear. Steady-state plasma concentrations of NDMC20 were attained within 14 days, with low between-subjects variability. Mean steady-state concentration (CS-S , SD) reached 209 (22) ng/ml. NDMC absence of sedative effect and its overall well-characterized safety coming from years of utilization as a metabolite from clobazam, raise the prospect of dose escalating trials in patients to quantify its clinical utility. SIGNIFICANCE: This article, presenting the Phase I data of the new antihyperalgesic compound, α2-subunit GABAA positive allosteric modulator, N-desmethyl-clobazam (NDMC) is exploring the modulation of a new target in the treatment of neuropathic pain. Based on these results and on its preclinical properties NDMC would qualify as a good tool compound to seek confirmation of the clinical utility of selective GABA allosteric modulators in neuropathic pain patients.
Collapse
Affiliation(s)
- Alain Matthey
- Division of Clinical Pharmacology and Toxicology, Multidisciplinary Pain Center, Geneva University Hospitals, Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Multidisciplinary Pain Center, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - François Curtin
- Division of Clinical Pharmacology and Toxicology, Multidisciplinary Pain Center, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Antoine Poncet
- Division of Clinical Epidemiology, Geneva University Hospitals, Geneva, Switzerland
| | - Jules Desmeules
- Division of Clinical Pharmacology and Toxicology, Multidisciplinary Pain Center, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Marie Besson
- Division of Clinical Pharmacology and Toxicology, Multidisciplinary Pain Center, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
15
|
Duchon A, Gruart A, Albac C, Delatour B, Zorrilla de San Martin J, Delgado-García JM, Hérault Y, Potier MC. Long-lasting correction of in vivo LTP and cognitive deficits of mice modelling Down syndrome with an α5-selective GABA A inverse agonist. Br J Pharmacol 2020; 177:1106-1118. [PMID: 31652355 PMCID: PMC7042104 DOI: 10.1111/bph.14903] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose Excessive GABAergic inhibition contributes to cognitive dysfunctions in Down syndrome (DS). Selective negative allosteric modulators (NAMs) of α5‐containing GABAA receptors such as the α5 inverse agonist (α5IA) restore learning and memory deficits in Ts65Dn mice, a model of DS. In this study we have assessed the long‐lasting effects of α5IA on in vivo LTP and behaviour in Ts65Dn mice. Experimental Approach We made in vivo LTP recordings for six consecutive days in freely moving Ts65Dn mice and their wild‐type littermates, treated with vehicle or α5IA. In parallel, Ts65Dn mice were assessed by various learning and memory tests (Y maze, Morris water maze, or the novel object recognition) for up to 7 days, following one single injection of α5IA or vehicle. Key Results LTP was not evoked in vivo in Ts65Dn mice at hippocampal CA3‐CA1 synapses. However, this deficit was sustainably reversed for at least six consecutive days following a single injection of α5IA. This long‐lasting effect of α5IA was also observed when assessing working and long‐term memory deficits in Ts65Dn mice. Conclusion and Implications We show for the first time in vivo LTP deficits in Ts65Dn mice. These deficits were restored for at least 6 days following acute treatment with α5IA and might be the substrate for the long‐lasting pharmacological effects of α5IA on spatial working and long‐term recognition and spatial memory tasks. Our results demonstrate the relevance of negative allosteric modulators of α5‐containing GABAA receptors to the treatment of cognitive deficits associated with DS.
Collapse
Affiliation(s)
- Arnaud Duchon
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Neuropôle, Université de Strasbourg, Illkirch, France
| | - Agnès Gruart
- División de Neurociencias, Universidad Pablo de Olavide, Seville, Spain
| | - Christelle Albac
- Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, Paris, France.,Centre National de la Recherche Scientifique, UMR7225, Hôpital de la Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Benoît Delatour
- Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, Paris, France.,Centre National de la Recherche Scientifique, UMR7225, Hôpital de la Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Javier Zorrilla de San Martin
- Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, Paris, France.,Centre National de la Recherche Scientifique, UMR7225, Hôpital de la Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | | | - Yann Hérault
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France.,Neuropôle, Université de Strasbourg, Illkirch, France
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, Paris, France.,Centre National de la Recherche Scientifique, UMR7225, Hôpital de la Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
16
|
Maramai S, Benchekroun M, Ward SE, Atack JR. Subtype Selective γ-Aminobutyric Acid Type A Receptor (GABAAR) Modulators Acting at the Benzodiazepine Binding Site: An Update. J Med Chem 2019; 63:3425-3446. [DOI: 10.1021/acs.jmedchem.9b01312] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Samuele Maramai
- Sussex Drug Discovery Centre, University of Sussex, Brighton BN1 9QJ, U.K
| | - Mohamed Benchekroun
- Sussex Drug Discovery Centre, University of Sussex, Brighton BN1 9QJ, U.K
- Équipe de Chimie Moléculaire, Laboratoire de Génomique Bioinformatique et Chimie Moléculaire, GBCM, EA7528, Conservatoire National des Arts et Métiers, 2 rue Conté, 75003 Paris, France
| | - Simon E. Ward
- Medicines Discovery Institute, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K
| | - John R. Atack
- Medicines Discovery Institute, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K
| |
Collapse
|
17
|
Gao T, Liu Y, Zhao Z, Luo Y, Wang L, Wang Y, Yin Y. L-655,708 Does not Prevent Isoflurane-induced Memory Deficits in Old Mice. Transl Neurosci 2019; 10:180-186. [PMID: 31410301 PMCID: PMC6689210 DOI: 10.1515/tnsci-2019-0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/21/2019] [Indexed: 01/07/2023] Open
Abstract
Background General anesthesia and increasing age are two main risk factors for postoperative cognitive dysfunction (POCD). Effective agents for the prevention or treatment of POCD are urgently needed. L-655,708, an inverse agonist of α5 subunit-containing γ-aminobutyric acid subtype A (α5GABAA) receptors, can prevent anesthesia-induced memory deficits in young animals. However, there is a lack of evidence of its efficacy in old animals. Methodology Young (3- to 5-month-old) and old (18- to 20-month-old) mice were given an inhalation of 1.33% isoflurane for 1 hour and their associative memory was evaluated 24 hours after anesthesia using fear-conditioning tests (FCTs). To evaluate the effect of L-655,708, mice received intraperitoneal injections of L-655,708 (0.7 mg/kg) or vehicle 30 minutes before anesthesia. Results Old mice exhibited impaired memory and lower hippocampal α5GABAA levels than young mice under physiological conditions. Pre-injections of L-655,708 significantly alleviated isoflurane-induced memory decline in young mice, but not in old mice. Conclusions L-655,708 is not as effective for the prevention of POCD in old mice as it is in young mice. The use of inverse agonists of α5GABAA in preventing POCD in old patients should be carefully considered.
Collapse
Affiliation(s)
- Teng Gao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yue Liu
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zifang Zhao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lifang Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yiqing Yin
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
18
|
Solomon VR, Tallapragada VJ, Chebib M, Johnston G, Hanrahan JR. GABA allosteric modulators: An overview of recent developments in non-benzodiazepine modulators. Eur J Med Chem 2019; 171:434-461. [DOI: 10.1016/j.ejmech.2019.03.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/17/2019] [Accepted: 03/17/2019] [Indexed: 01/13/2023]
|
19
|
Sequeira A, Shen K, Gottlieb A, Limon A. Human brain transcriptome analysis finds region- and subject-specific expression signatures of GABA AR subunits. Commun Biol 2019; 2:153. [PMID: 31069263 PMCID: PMC6494906 DOI: 10.1038/s42003-019-0413-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/03/2019] [Indexed: 11/19/2022] Open
Abstract
Altered expression of GABA receptors (GABAARs) has been implicated in neurological and psychiatric disorders, but limited information about region-specific GABAAR subunit expression in healthy human brains, heteromeric assembly of major isoforms, and their collective organization across healthy individuals, are major roadblocks to understanding their role in non-physiological states. Here, by using microarray and RNA-Seq datasets-from single cell nuclei to global brain expression-from the Allen Institute, we find that transcriptional expression of GABAAR subunits is anatomically organized according to their neurodevelopmental origin. The data show a combination of complementary and mutually-exclusive expression patterns that delineate major isoforms, and which is highly stereotypical across brains from control donors. We summarize the region-specific signature of GABAR subunits per subject and its variability in a control population sample that can be used as a reference for remodeling changes during homeostatic rearrangements of GABAAR subunits after physiological, pharmacological or pathological challenges.
Collapse
Affiliation(s)
- Adolfo Sequeira
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA USA
| | - Kevin Shen
- Department of Neurology, Mitchel Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX USA
| | - Assaf Gottlieb
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Agenor Limon
- Department of Neurology, Mitchel Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch, Galveston, TX USA
| |
Collapse
|
20
|
Zhao ZF, Du L, Gao T, Bao L, Luo Y, Yin YQ, Wang YA. Inhibition of α5 GABAA receptors has preventive but not therapeutic effects on isoflurane-induced memory impairment in aged rats. Neural Regen Res 2019; 14:1029-1036. [PMID: 30762015 PMCID: PMC6404482 DOI: 10.4103/1673-5374.250621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The α5 subunit-containing gamma-amino butyric acid type A receptors (α5 GABAARs) are a distinct subpopulation that are specifically distributed in the mammalian hippocampus and also mediate tonic inhibitory currents in hippocampal neurons. These tonic currents can be enhanced by low-dose isoflurane, which is associated with learning and memory impairment. Inverse agonists of α5 GABAARs, such as L-655,708, are able to reverse the short-term memory deficit caused by low-dose isoflurane in young animals. However, whether these negative allosteric modulators have the same effects on aged rats remains unclear. In the present study, we mainly investigated the effects of L-655,708 on low-dose (1.3%) isoflurane-induced learning and memory impairment in elderly rats. Young (3-month-old) and aged (24-month-old) Wistar rats were randomly assigned to receive L-655,708 0.5 hour before or 23.5 hours after 1.3% isoflurane anesthesia. The Morris Water Maze tests demonstrated that L-655,708 injected before or after anesthesia could reverse the memory deficit in young rats. But in aged rats, application of L-655,708 only before anesthesia showed similar effects. Reverse transcription-polymerase chain reaction showed that low-dose isoflurane decreased the mRNA expression of α5 GABAARs in aging hippocampal neurons but increased that in young animals. These findings indicate that L-655,708 prevented but could not reverse 1.3% isoflurane-induced spatial learning and memory impairment in aged Wistar rats. All experimental procedures and protocols were approved by the Experimental Animal Ethics Committee of Academy of Military Medical Science of China (approval No. NBCDSER-IACUC-2015128) in December 2015.
Collapse
Affiliation(s)
- Zi-Fang Zhao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Lei Du
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Teng Gao
- Department of Anesthesiology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Lin Bao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Yuan Luo
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Yi-Qing Yin
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Yong-An Wang
- Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Golovko AI, Ivanov MB, Golovko ES, Dolgo-Saburov VB, Zatsepin EP. The Neurochemical Mechanisms of the Pharmacological Activities of Inverse Agonists of the Benzodiazepine Binding Site. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418030042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Calvo-Flores Guzmán B, Vinnakota C, Govindpani K, Waldvogel HJ, Faull RL, Kwakowsky A. The GABAergic system as a therapeutic target for Alzheimer's disease. J Neurochem 2018; 146:649-669. [DOI: 10.1111/jnc.14345] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Beatriz Calvo-Flores Guzmán
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Chitra Vinnakota
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Karan Govindpani
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Henry J. Waldvogel
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Richard L.M. Faull
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| |
Collapse
|
23
|
Quercetin Reduces Cortical GABAergic Transmission and Alleviates MK-801-Induced Hyperactivity. EBioMedicine 2018; 34:201-213. [PMID: 30057312 PMCID: PMC6116474 DOI: 10.1016/j.ebiom.2018.07.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/11/2018] [Accepted: 07/20/2018] [Indexed: 02/06/2023] Open
Abstract
An imbalance between neuronal excitation and inhibition represents a core feature in multiple neuropsychiatry disorders, necessitating the development of novel strategies to calibrate the excitatory–inhibitory balance of therapeutics. Here we identify a natural compound quercetin that reduces prefrontal cortical GABAergic transmission and alleviates the hyperactivity induced by glutamatergic N-methyl-d-aspartate receptor antagonist MK-801. Quercetin markedly reduced the GABA-activated currents in a noncompetitive manner in cultured cortical neurons, and moderately inhibited spontaneous and electrically-evoked GABAergic inhibitory postsynaptic current in mouse prefrontal cortical slices. Notably, systemic and prefrontal-specific delivery of quercetin reduced basal locomotor activity in addition to alleviated the MK-801-induced hyperactivity. The effects of quercetin were not exclusively dependent on α5-subunit-containing A type GABA receptors (GABAARs), as viral-mediated, region-specific genetic knockdown of the α5-subunit in prefrontal cortex improved the MK-801-evoked psychotic symptom but reserved the pharmacological responsivity to quercetin. Both interventions together completely normalized the locomotor activity. Together, quercetin as a negative allosteric GABAAR modulator exerted antipsychotic activity, facilitating further therapeutic development for the excitatory–inhibitory imbalance disorders.
Collapse
|
24
|
Xu MY, Wong AHC. GABAergic inhibitory neurons as therapeutic targets for cognitive impairment in schizophrenia. Acta Pharmacol Sin 2018; 39:733-753. [PMID: 29565038 DOI: 10.1038/aps.2017.172] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022] Open
Abstract
Schizophrenia is considered primarily as a cognitive disorder. However, functional outcomes in schizophrenia are limited by the lack of effective pharmacological and psychosocial interventions for cognitive impairment. GABA (gamma-aminobutyric acid) interneurons are the main inhibitory neurons in the central nervous system (CNS), and they play a critical role in a variety of pathophysiological processes including modulation of cortical and hippocampal neural circuitry and activity, cognitive function-related neural oscillations (eg, gamma oscillations) and information integration and processing. Dysfunctional GABA interneuron activity can disrupt the excitatory/inhibitory (E/I) balance in the cortex, which could represent a core pathophysiological mechanism underlying cognitive dysfunction in schizophrenia. Recent research suggests that selective modulation of the GABAergic system is a promising intervention for the treatment of schizophrenia-associated cognitive defects. In this review, we summarized evidence from postmortem and animal studies for abnormal GABAergic neurotransmission in schizophrenia, and how altered GABA interneurons could disrupt neuronal oscillations. Next, we systemically reviewed a variety of up-to-date subtype-selective agonists, antagonists, positive and negative allosteric modulators (including dual allosteric modulators) for α5/α3/α2 GABAA and GABAB receptors, and summarized their pro-cognitive effects in animal behavioral tests and clinical trials. Finally, we also discuss various representative histone deacetylases (HDAC) inhibitors that target GABA system through epigenetic modulations, GABA prodrug and presynaptic GABA transporter inhibitors. This review provides important information on current potential GABA-associated therapies and future insights for development of more effective treatments.
Collapse
|
25
|
Chandler CM, Overton JS, Rüedi-Bettschen D, Platt DM. GABA A Receptor Subtype Mechanisms and the Abuse-Related Effects of Ethanol: Genetic and Pharmacological Evidence. Handb Exp Pharmacol 2018; 248:3-27. [PMID: 29204713 DOI: 10.1007/164_2017_80] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Ethanol's reinforcing and subjective effects, as well as its ability to induce relapse, are powerful factors contributing to its widespread use and abuse. A significant mediator of these behavioral effects is the GABAA receptor system. GABAA receptors are the target for γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter in the CNS. Structurally, they are pentameric, transmembrane chloride ion channels comprised of subunits from at least eight different families of distinct proteins. The contribution of different GABAA subunits to ethanol's diverse abuse-related effects is not clear and remains an area of research focus. This chapter details the clinical and preclinical findings supporting roles for different α, β, γ, and δ subunit-containing GABAA receptors in ethanol's reinforcing, subjective/discriminative stimulus, and relapse-inducing effects. The reinforcing properties of ethanol have been studied the most systematically, and convergent preclinical evidence suggests a key role for the α5 subunit in those effects. Regarding ethanol's subjective/discriminative stimulus effects, clinical and genetic findings support a primary role for the α2 subunit, whereas preclinical evidence implicates the α5 subunit. At present, too few studies investigating ethanol relapse exist to make any solid conclusions regarding the role of specific GABAA subunits in this abuse-related effect.
Collapse
Affiliation(s)
- Cassie M Chandler
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - John S Overton
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniela Rüedi-Bettschen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Donna M Platt
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
26
|
Vidal V, García-Cerro S, Martínez P, Corrales A, Lantigua S, Vidal R, Rueda N, Ozmen L, Hernández MC, Martínez-Cué C. Decreasing the Expression of GABA A α5 Subunit-Containing Receptors Partially Improves Cognitive, Electrophysiological, and Morphological Hippocampal Defects in the Ts65Dn Model of Down Syndrome. Mol Neurobiol 2017; 55:4745-4762. [PMID: 28717969 DOI: 10.1007/s12035-017-0675-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/27/2017] [Indexed: 12/31/2022]
Abstract
Trisomy 21 or Down syndrome (DS) is the most common cause of intellectual disability of a genetic origin. The Ts65Dn (TS) mouse, which is the most commonly used and best-characterized mouse model of DS, displays many of the cognitive, neuromorphological, and biochemical anomalies that are found in the human condition. One of the mechanisms that have been proposed to be responsible for the cognitive deficits in this mouse model is impaired GABA-mediated inhibition. Because of the well-known modulatory role of GABAA α5 subunit-containing receptors in cognitive processes, these receptors are considered to be potential targets for improving the intellectual disability in DS. The chronic administration of GABAA α5-negative allosteric modulators has been shown to be procognitive without anxiogenic or proconvulsant side effects. In the present study, we use a genetic approach to evaluate the contribution of GABAA α5 subunit-containing receptors to the cognitive, electrophysiological, and neuromorphological deficits in TS mice. We show that reducing the expression of GABAA α5 receptors by deleting one or two copies of the Gabra5 gene in TS mice partially ameliorated the cognitive impairments, improved long-term potentiation, enhanced neural differentiation and maturation, and normalized the density of the GABAergic synapse markers. Reducing the gene dosage of Gabra5 in TS mice did not induce motor alterations and anxiety or affect the viability of the mice. Our results provide further evidence of the role of GABAA α5 receptor-mediated inhibition in cognitive impairment in the TS mouse model of DS.
Collapse
Affiliation(s)
- Verónica Vidal
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Susana García-Cerro
- Departamento de Fundamentos Clínicos, Unidad de Farmacología, Universitat de Barcelona, Barcelona, Spain
| | - Paula Martínez
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Andrea Corrales
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Sara Lantigua
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Rebeca Vidal
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain.,Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), Santander, Spain.,Centro de Investigacion Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Noemí Rueda
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain
| | - Laurence Ozmen
- Pharma Research and Early Development, Hoffman-La Roche Ltd., Basel, Switzerland
| | | | - Carmen Martínez-Cué
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad deCantabria, Santander, Spain.
| |
Collapse
|
27
|
McGinnity CJ, Riaño Barros DA, Rosso L, Veronese M, Rizzo G, Bertoldo A, Hinz R, Turkheimer FE, Koepp MJ, Hammers A. Test-retest reproducibility of quantitative binding measures of [ 11C]Ro15-4513, a PET ligand for GABA A receptors containing alpha5 subunits. Neuroimage 2017; 152:270-282. [PMID: 28292717 PMCID: PMC5440177 DOI: 10.1016/j.neuroimage.2016.12.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/20/2016] [Accepted: 12/14/2016] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Alteration of γ-aminobutyric acid "A" (GABAA) receptor-mediated neurotransmission has been associated with various neurological and psychiatric disorders. [11C]Ro15-4513 is a PET ligand with high affinity for α5-subunit-containing GABAA receptors, which are highly expressed in limbic regions of the human brain (Sur et al., 1998). We quantified the test-retest reproducibility of measures of [11C]Ro15-4513 binding derived from six different quantification methods (12 variants). METHODS Five healthy males (median age 40 years, range 38-49 years) had a 90-min PET scan on two occasions (median interval 12 days, range 11-30 days), after injection of a median dose of 441 MegaBequerels of [11C]Ro15-4513. Metabolite-corrected arterial plasma input functions (parent plasma input functions, ppIFs) were generated for all scans. We quantified regional binding using six methods (12 variants), some of which were region-based (applied to the average time-activity curve within a region) and others were voxel-based: 1) Models requiring arterial ppIFs - regional reversible compartmental models with one and two tissue compartments (2kbv and 4kbv); 2) Regional and voxelwise Logan's graphical analyses (Logan et al., 1990), which required arterial ppIFs; 3) Model-free regional and voxelwise (exponential) spectral analyses (SA; (Cunningham and Jones, 1993)), which also required arterial ppIFs; 4) methods not requiring arterial ppIFs - voxelwise standardised uptake values (Kenney et al., 1941), and regional and voxelwise simplified reference tissue models (SRTM/SRTM2) using brainstem or alternatively cerebellum as pseudo-reference regions (Lammertsma and Hume, 1996; Gunn et al., 1997). To compare the variants, we sampled the mean values of the outcome parameters within six bilateral, non-reference grey matter regions-of-interest. Reliability was quantified in terms of median absolute percentage test-retest differences (MA-TDs; preferentially low) and between-subject coefficient of variation (BS-CV, preferentially high), both compounded by the intraclass correlation coefficient (ICC). These measures were compared between variants, with particular interest in the hippocampus. RESULTS Two of the six methods (5/12 variants) yielded reproducible data (i.e. MA-TD <10%): regional SRTMs and voxelwise SRTM2s, both using either the brainstem or the cerebellum; and voxelwise SA. However, the SRTMs using the brainstem yielded a lower median BS-CV (7% for regional, 7% voxelwise) than the other variants (8-11%), resulting in lower ICCs. The median ICCs across six regions were 0.89 (interquartile range 0.75-0.90) for voxelwise SA, 0.71 (0.64-0.84) for regional SRTM-cerebellum and 0.83 (0.70-0.86) for voxelwise SRTM-cerebellum. The ICCs for the hippocampus were 0.89 for voxelwise SA, 0.95 for regional SRTM-cerebellum and 0.93 for voxelwise SRTM-cerebellum. CONCLUSION Quantification of [11C]Ro15-4513 binding shows very good to excellent reproducibility with SRTM and with voxelwise SA which, however, requires an arterial ppIF. Quantification in the α5 subunit-rich hippocampus is particularly reliable. The very low expression of the α5 in the cerebellum (Fritschy and Mohler, 1995; Veronese et al., 2016) and the substantial α1 subunit density in this region may hamper the application of reference tissue methods.
Collapse
Affiliation(s)
- Colm J McGinnity
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK; Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK.
| | - Daniela A Riaño Barros
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Lula Rosso
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gaia Rizzo
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Federico E Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, UK; Epilepsy Society, Chalfont St Peter, UK
| | - Alexander Hammers
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK; Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK; The Neurodis Foundation, CERMEP - Imagerie du Vivant, Lyon, France
| |
Collapse
|
28
|
Abstract
The nomenclature of drugs is a critical aspect of science, since it can direct research and optimize treatment choices. Traditionally drugs acting on CNS receptors have been classified as either agonists or antagonists. Recently a new class of ligand, the inverse agonist, has been identified in some receptor systems. Inverse agonists have opposite actions to those of agonists but the effects of both of these can be blocked by antagonists. Pimavanserin is a new 5-HT2A receptor acting drug that has been given market authorization for psychosis in Parkinson׳s disease. The FDA have termed it an inverse agonist, but this conclusion is based on in-vitro data. In this paper we discuss the evidence for such a claim being made for pimavanserin in the human brain and conclude that this is not currently sufficient. It is therefore premature to conclude that the actions of pimavanserin in humans are due to inverse agonism, and we are of the opinion that it should be called a 5-HT2A antagonist until better evidence emerges.
Collapse
|
29
|
Lingford-Hughes A, Myers J, Watson B, Reid AG, Kalk N, Feeney A, Hammers A, Riaño-Barros DA, McGinnity CJ, Taylor LG, Rosso L, Brooks DJ, Turkheimer F, Nutt DJ. Using [(11)C]Ro15 4513 PET to characterise GABA-benzodiazepine receptors in opiate addiction: Similarities and differences with alcoholism. Neuroimage 2016; 132:1-7. [PMID: 26876472 PMCID: PMC4862962 DOI: 10.1016/j.neuroimage.2016.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 01/31/2016] [Accepted: 02/04/2016] [Indexed: 01/19/2023] Open
Abstract
The importance of the GABA-benzodiazepine receptor complex and its subtypes are increasingly recognised in addiction. Using the α1/α5 benzodiazepine receptor PET radioligand [11C]Ro15 4513, we previously showed reduced binding in the nucleus accumbens and hippocampus in abstinent alcohol dependence. We proposed that reduced [11C]Ro15 4513 binding in the nucleus accumbens was a marker of addiction whilst the reduction in hippocampus and positive relationship with memory was a consequence of chronic alcohol abuse. To examine this further we assessed [11C]Ro15 4513 binding in another addiction, opiate dependence, and used spectral analysis to estimate contributions of α1 and α5 subtypes to [11C]Ro15 4513 binding in opiate and previously acquired alcohol-dependent groups. Opiate substitute maintained opiate-dependent men (n = 12) underwent an [11C]Ro15 4513 PET scan and compared with matched healthy controls (n = 13). We found a significant reduction in [11C]Ro15 4513 binding in the nucleus accumbens in the opiate-dependent compared with the healthy control group. There was no relationship between [11C]Ro15 4513 binding in the hippocampus with memory. We found that reduced [11C]Ro15 4513 binding was associated with reduced α5 but not α1 subtypes in the opiate-dependent group. This was also seen in an alcohol-dependent group where an association between memory performance and [11C]Ro15 4513 binding was primarily driven by α5 and not α1 subtype. We suggest that reduced α5 levels in the nucleus accumbens are associated with addiction since we have now shown this in dependence to two pharmacologically different substances, alcohol and opiates. Lower [11C]Ro15 4513 binding is evident in the nucleus accumbens of opiate addicts. This appears primarily due to lower levels of α5 subtype of the GABA-A receptor. Lower [11C]Ro15 4513 binding is similarly found in alcoholism. Lower levels of α5 GABA-A receptor in nucleus accumbens may underpin addiction.
Collapse
Affiliation(s)
- Anne Lingford-Hughes
- Centre for Neuropsychopharmacology, Imperial College London, Du Cane Rd., London W12 0NN, United Kingdom; Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom; Hammersmith Imanet Ltd., Hammersmith Hospital, Du Cane Rd., London W12 0NN, United Kingdom.
| | - James Myers
- Centre for Neuropsychopharmacology, Imperial College London, Du Cane Rd., London W12 0NN, United Kingdom; Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Ben Watson
- Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom; Hammersmith Imanet Ltd., Hammersmith Hospital, Du Cane Rd., London W12 0NN, United Kingdom
| | - Alastair G Reid
- Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom; Hammersmith Imanet Ltd., Hammersmith Hospital, Du Cane Rd., London W12 0NN, United Kingdom
| | - Nicola Kalk
- Centre for Neuropsychopharmacology, Imperial College London, Du Cane Rd., London W12 0NN, United Kingdom; Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Adrian Feeney
- Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Alexander Hammers
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Daniela A Riaño-Barros
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Colm J McGinnity
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Lindsay G Taylor
- Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom
| | - Lula Rosso
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - David J Brooks
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom; Hammersmith Imanet Ltd., Hammersmith Hospital, Du Cane Rd., London W12 0NN, United Kingdom
| | - Federico Turkheimer
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - David J Nutt
- Centre for Neuropsychopharmacology, Imperial College London, Du Cane Rd., London W12 0NN, United Kingdom; Psychopharmacology Unit, University of Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom; MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| |
Collapse
|
30
|
Wiers CE, Cabrera E, Skarda E, Volkow ND, Wang GJ. PET imaging for addiction medicine: From neural mechanisms to clinical considerations. PROGRESS IN BRAIN RESEARCH 2015; 224:175-201. [PMID: 26822359 DOI: 10.1016/bs.pbr.2015.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Positron emission tomography (PET) has been shown to be an effective imaging technique to study neurometabolic and neurochemical processes involved in addiction. That is, PET has been used to research neurobiological differences in substance abusers versus healthy controls and the pharmacokinetics and pharmacodynamics of abused drugs. Over the past years, the research scope has shifted to investigating neurobiological effects of abstinence and treatment, and their predictive power for relapse and other clinical outcomes. This chapter provides an overview of PET methodology, recent human PET studies on drug addiction and their implications for clinical treatment.
Collapse
Affiliation(s)
- Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Elizabeth Cabrera
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emily Skarda
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA; National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
31
|
GABAergic modulation in central sensitization in humans: a randomized placebo-controlled pharmacokinetic-pharmacodynamic study comparing clobazam with clonazepam in healthy volunteers. Pain 2015; 156:397-404. [PMID: 25687539 DOI: 10.1097/01.j.pain.0000460331.33385.e8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Positive allosteric modulators of GABAA receptors (GAMs) acting at specific subtypes of GABAA receptors effectively restore compromised spinal pain control in rodents. Studies addressing a similar antihyperalgesic effect in humans are sparse and are hampered by sedative effects of nonselective GAMs available for use in humans. We present results from a randomized controlled double-blind crossover study in 25 healthy volunteers, which addressed potential antihyperalgesic actions of clobazam (CBZ) and clonazepam (CLN) at mildly sedating equianticonvulsive doses. Clobazam was chosen because of its relatively low sedative properties and CLN because of its use in neuropathic pain. Tolterodine (TLT) was used as an active placebo. The primary outcome parameter was a change in the area of cutaneous UVB irradiation-induced secondary hyperalgesia (ASH), which was monitored for 8 hours after drug application. Sedative effects were assessed in parallel to antihyperalgesia. Compared with TLT, recovery from hyperalgesia was significantly faster in the CBZ and CLN groups (P = 0.009). At the time point of maximum effect, the rate of recovery from hyperalgesia was accelerated by CBZ and CLN, relative to placebo by 15.7% (95% confidence interval [CI] 0.8-30.5), P = 0.040, and 28.6% (95% CI 4.5-52.6), P = 0.022, respectively. Active compounds induced stronger sedation than placebo, but these differences disappeared 8 hours after drug application. We demonstrate here that GAMs effectively reduce central sensitization in healthy volunteers. These results provide proof-of-principle evidence supporting efficacy of GAMs as antihyperalgesic agents in humans and should stimulate further research on compounds with improved subtype specificity.
Collapse
|
32
|
Stamenić TT, Joksimović S, Biawat P, Stanković T, Marković B, Cook JM, Savić MM. Negative modulation of α₅ GABAA receptors in rats may partially prevent memory impairment induced by MK-801, but not amphetamine- or MK-801-elicited hyperlocomotion. J Psychopharmacol 2015; 29:1013-24. [PMID: 26105958 PMCID: PMC4861997 DOI: 10.1177/0269881115590601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reportedly, negative modulation of α5 GABAA receptors may improve cognition in normal and pharmacologically-impaired animals, and such modulation has been proposed as an avenue for treatment of cognitive symptoms in schizophrenia. This study assessed the actions of PWZ-029, administered at doses (2, 5, and 10 mg/kg) at which it reached micromolar concentrations in brain tissue with estimated free concentrations adequate for selective modulation of α5 GABAA receptors, in three cognitive tasks in male Wistar rats acutely treated with the noncompetitive N-methyl-d-aspartate receptor antagonist, MK-801 (0.1 mg/kg), as well in tests of locomotor activity potentiated by MK-801 (0.2 mg/kg) or amphetamine (0.5 mg/kg). In a hormetic-like manner, only 5 mg/kg PWZ-029 reversed MK-801-induced deficits in novel object recognition test (visual recognition memory), whereas in the Morris water maze, the 2 mg/kg dose of PWZ-029 exerted partial beneficial effects on spatial learning impairment. PWZ-029 did not affect recognition memory deficits in social novelty discrimination procedure. Motor hyperactivity induced with MK-801 or amphetamine was not preventable by PWZ-029. Our results show that certain MK-801-induced memory deficits can be ameliorated by negative modulation of α5 GABAA receptors, and point to the need for further elucidation of their translational relevance to cognitive deterioration in schizophrenia.
Collapse
Affiliation(s)
| | - Srdjan Joksimović
- Department of Pharmacology, University of Belgrade, Belgrade, Serbia
| | - Poonam Biawat
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Tamara Stanković
- Department of Pharmacology, University of Belgrade, Belgrade, Serbia
| | - Bojan Marković
- Department of Pharmaceutical Chemistry, University of Belgrade, Belgrade, Serbia
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Miroslav M Savić
- Department of Pharmacology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
33
|
Inert gas narcosis disrupts encoding but not retrieval of long term memory. Physiol Behav 2015; 144:46-51. [PMID: 25725120 DOI: 10.1016/j.physbeh.2015.02.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 11/20/2022]
Abstract
Exposure to increased ambient pressure causes inert gas narcosis of which one symptom is long-term memory (LTM) impairment. Narcosis is posited to impair LTM by disrupting information encoding, retrieval (self-guided search), or both. The effect of narcosis on the encoding and retrieval of LTM was investigated by testing the effect of learning-recall pressure and levels of processing (LoP) on the free-recall of word lists in divers underwater. All participants (n=60) took part in four conditions in which words were learnt and then recalled at either low pressure (1.4-1.9atm/4-9msw) or high pressure (4.4-5.0atm/34-40msw), as manipulated by changes in depth underwater: low-low (LL), low-high(LH), high-high (HH), and high-low (HL). In addition, participants were assigned to either a deep or shallow processing condition, using LoP methodology. Free-recall memory ability was significantly impaired only when words were initially learned at high pressure (HH & HL conditions). When words were learned at low pressure and then recalled at low pressure (LL condition) or high pressure (LH condition) free-recall was not impaired. Although numerically superior in several conditions, deeper processing failed to significantly improve free-recall ability in any of the learning-recall conditions. This pattern of results support the hypothesis that narcosis disrupts encoding of information into LTM, while retrieval appears to be unaffected. These findings are discussed in relation to similar effects reported by some memory impairing drugs and the practical implications for workers in pressurised environments.
Collapse
|
34
|
Kell DB, Oliver SG. How drugs get into cells: tested and testable predictions to help discriminate between transporter-mediated uptake and lipoidal bilayer diffusion. Front Pharmacol 2014; 5:231. [PMID: 25400580 PMCID: PMC4215795 DOI: 10.3389/fphar.2014.00231] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/29/2014] [Indexed: 12/12/2022] Open
Abstract
One approach to experimental science involves creating hypotheses, then testing them by varying one or more independent variables, and assessing the effects of this variation on the processes of interest. We use this strategy to compare the intellectual status and available evidence for two models or views of mechanisms of transmembrane drug transport into intact biological cells. One (BDII) asserts that lipoidal phospholipid Bilayer Diffusion Is Important, while a second (PBIN) proposes that in normal intact cells Phospholipid Bilayer diffusion Is Negligible (i.e., may be neglected quantitatively), because evolution selected against it, and with transmembrane drug transport being effected by genetically encoded proteinaceous carriers or pores, whose “natural” biological roles, and substrates are based in intermediary metabolism. Despite a recent review elsewhere, we can find no evidence able to support BDII as we can find no experiments in intact cells in which phospholipid bilayer diffusion was either varied independently or measured directly (although there are many papers where it was inferred by seeing a covariation of other dependent variables). By contrast, we find an abundance of evidence showing cases in which changes in the activities of named and genetically identified transporters led to measurable changes in the rate or extent of drug uptake. PBIN also has considerable predictive power, and accounts readily for the large differences in drug uptake between tissues, cells and species, in accounting for the metabolite-likeness of marketed drugs, in pharmacogenomics, and in providing a straightforward explanation for the late-stage appearance of toxicity and of lack of efficacy during drug discovery programmes despite macroscopically adequate pharmacokinetics. Consequently, the view that Phospholipid Bilayer diffusion Is Negligible (PBIN) provides a starting hypothesis for assessing cellular drug uptake that is much better supported by the available evidence, and is both more productive and more predictive.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry, The University of Manchester Manchester, UK ; Manchester Institute of Biotechnology, The University of Manchester Manchester, UK
| | - Stephen G Oliver
- Department of Biochemistry, University of Cambridge Cambridge, UK ; Cambridge Systems Biology Centre, University of Cambridge Cambridge, UK
| |
Collapse
|
35
|
Campbell AE, Sumner P, Singh KD, Muthukumaraswamy SD. Acute effects of alcohol on stimulus-induced gamma oscillations in human primary visual and motor cortices. Neuropsychopharmacology 2014; 39:2104-13. [PMID: 24622470 PMCID: PMC4057932 DOI: 10.1038/npp.2014.58] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/04/2014] [Accepted: 02/24/2014] [Indexed: 11/09/2022]
Abstract
Alcohol is a rich drug affecting both the γ-amino butyric acid (GABA) and glutamatergic neurotransmitter systems. Recent findings from both modeling and pharmacological manipulation have indicated a link between GABAergic activity and oscillations measured in the gamma frequency range (30-80 Hz), but there are no previous reports of alcohol's modulation of gamma-band activity measured by magnetoencephalography (MEG) or electroencephalography (EEG). In this single-blind, placebo-controlled crossover study, 16 participants completed two study days, on one day of which they consumed a dose of 0.8 g/kg alcohol, and on the other day a placebo. MEG recordings of brain activity were taken before and after beverage consumption, using visual grating and finger abduction paradigms known to induce gamma-band activity in the visual and motor cortices respectively. Time-frequency analyses of beamformer source reconstructions in the visual cortex showed that alcohol increased peak gamma amplitude and decreased peak frequency. For the motor task, alcohol increased gamma amplitude in the motor cortex. These data support the notion that gamma oscillations are dependent, in part, on the balance between excitation and inhibition. Disruption of this balance by alcohol, by increasing GABAergic inhibition at GABAA receptors and decreasing glutamatergic excitation at N-methyl-D-aspartic acid receptors, alters both the amplitude and frequency of gamma oscillations. The findings provide further insight into the neuropharmacological action of alcohol.
Collapse
Affiliation(s)
- Anne E Campbell
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK,School of Psychology, Cardiff University, 70 Park Place, Cardiff CF10 3AT, UK, Tel: +44 29 20870470, Fax: +44 (0)29 20870339, E-mail:
| | - Petroc Sumner
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Krish D Singh
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Suresh D Muthukumaraswamy
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| |
Collapse
|
36
|
Martínez-Cué C, Delatour B, Potier MC. Treating enhanced GABAergic inhibition in Down syndrome: use of GABA α5-selective inverse agonists. Neurosci Biobehav Rev 2014; 46 Pt 2:218-27. [PMID: 24412222 DOI: 10.1016/j.neubiorev.2013.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 11/27/2022]
Abstract
Excess inhibition in the brain of individuals carrying an extra copy of chromosome 21 could be responsible for cognitive deficits observed throughout their lives. A change in the excitatory/inhibitory balance in adulthood would alter synaptic plasticity, potentially triggering learning and memory deficits. γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mature central nervous system and binds to GABAA receptors, opens a chloride channel, and reduces neuronal excitability. In this review we discuss methods to alleviate neuronal inhibition in a mouse model of Down syndrome, the Ts65Dn mouse, using either an antagonist (pentylenetetrazol) or two different inverse agonists selective for the α5-subunit containing receptor. Both inverse agonists, which reduce inhibitory GABAergic transmission, could rescue learning and memory deficits in Ts65Dn mice. We also discuss safety issues since modulation of the excitatory-inhibitory balance to improve cognition without inducing seizures remains particularly difficult when using GABA antagonists.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Benoît Delatour
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
37
|
Gill KM, Grace AA. The role of α5 GABAA receptor agonists in the treatment of cognitive deficits in schizophrenia. Curr Pharm Des 2014; 20:5069-76. [PMID: 24345268 PMCID: PMC4074253 DOI: 10.2174/1381612819666131216114612] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/10/2013] [Indexed: 11/22/2022]
Abstract
Currently available pharmacotherapies for the treatment of schizophrenia are ineffective in restoring the disrupted cognitive function associated with this disorder. As such, there is a continued search for more viable novel drug targets. Engaging in cognitive behaviors is associated with distinct coordinated oscillatory activity across brain regions, in particular the hippocampus and prefrontal cortex. In schizophrenia patients, pathological alterations in the functionality of GABAergic interneurons in the PFC and HPC responsible for generating network oscillations are thought to contribute to impaired cognition. Destabilized GABAergic interneuron activity in the HPC is further associated with aberrant increases in HPC output and enhanced dopamine neuron activity. Consequently, drugs directed at restoring HPC function could impact both oscillatory activity along with dopamine tone. There is compelling evidence from animal models of schizophrenia that allosteric modulation of the α5 subunit of the GABAA receptor is a viable means of resolving aberrant dopamine system activity through indirect alteration of HPC output. Consequently, these compounds are promising for their potential in also ameliorating cognitive deficits attributed to dysfunction in HPC network activity.
Collapse
Affiliation(s)
| | - Anthony A Grace
- University of Pittsburgh, Department of Neuroscience, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
38
|
Egawa K, Fukuda A. Pathophysiological power of improper tonic GABA(A) conductances in mature and immature models. Front Neural Circuits 2013; 7:170. [PMID: 24167475 PMCID: PMC3807051 DOI: 10.3389/fncir.2013.00170] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 09/28/2013] [Indexed: 11/25/2022] Open
Abstract
High-affinity extrasynaptic gamma-aminobutyric acid A (GABAA) receptors are tonically activated by low and consistent levels of ambient GABA, mediating chronic inhibition against neuronal excitability (tonic inhibition) and the modulation of neural development. Synaptic (phasic) inhibition is spatially and temporally precise compared with tonic inhibition, which provides blunt yet strong integral inhibitory force by shunting electrical signaling. Although effects of acute modification of tonic inhibition are known, its pathophysiological significance remains unclear because homeostatic regulation of neuronal excitability can compensate for long-term deficit of extrasynaptic GABAA receptor activation. Nevertheless, tonic inhibition is of great interest for its pathophysiological involvement in central nervous system (CNS) diseases and thus as a therapeutic target. Together with the development of experimental models for various pathological states, recent evidence demonstrates such pathological involvements of tonic inhibition in neuronal dysfunction. This review focuses on the recent progress of tonic activation of GABAA conductance on the development and pathology of the CNS. Findings indicate that neuronal function in various brain regions are exacerbated with a gain or loss of function of tonic inhibition by GABA spillover. Disturbance of tonic GABAA conductance mediated by non-synaptic ambient GABA may result in brain mal-development. Therefore, various pathological states (epilepsy, motor dysfunctions, psychiatric disorders, and neurodevelopmental disorders) may be partly attributable to abnormal tonic GABAA conductances. Thus, the tone of tonic conductance and level of ambient GABA may be precisely tuned to maintain the regular function and development of the CNS. Therefore, receptor expression and factors for regulating the ambient GABA concentration are highlighted to gain a deeper understanding of pathology and therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Kiyoshi Egawa
- Department of Neurology, Massachusetts General Hospital Charlestown, MA, USA ; Department of Pediatrics, Hokkaido University Graduate School of Medicine Sapporo, Japan
| | | |
Collapse
|
39
|
Rudolph U, Möhler H. GABAA receptor subtypes: Therapeutic potential in Down syndrome, affective disorders, schizophrenia, and autism. Annu Rev Pharmacol Toxicol 2013; 54:483-507. [PMID: 24160694 DOI: 10.1146/annurev-pharmtox-011613-135947] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The γ-aminobutyric acid (GABA) system plays a pivotal role in orchestrating the synchronicity of local networks and the functional coupling of different brain regions. Here we review the impact of the GABAA receptor subtypes on cognitive and emotional behavior, paying particular attention to five disease states: cognitive dysfunction and Down syndrome, anxiety disorders, depression, schizophrenia, and autism. Through the bidirectional modulation of tonic inhibition, α5-subunit-containing GABAA receptors permit the bidirectional modulation of cognitive processes, and a partial inverse agonist acting at the α5-subunit-containing GABAA receptor is in a clinical trial in individuals with Down syndrome. With regard to anxiety disorders, the viability of nonsedative anxiolytics based on the modulation of α2- and α3-subunit-containing GABAA receptors has been established in clinical proof-of-concept trials. Regarding the remaining three disease states, the GABA hypothesis of depression offers new options for antidepressant drug development; cognitive symptoms in schizophrenia are attributed to a cortical GABAergic deficit, and dysfunctional GABAergic inhibition is increasingly understood to contribute to the pathophysiology of autism spectrum disorders.
Collapse
Affiliation(s)
- Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, Massachusetts 02478;
| | | |
Collapse
|
40
|
Mendez MA, Horder J, Myers J, Coghlan S, Stokes P, Erritzoe D, Howes O, Lingford-Hughes A, Murphy D, Nutt D. The brain GABA-benzodiazepine receptor alpha-5 subtype in autism spectrum disorder: a pilot [(11)C]Ro15-4513 positron emission tomography study. Neuropharmacology 2013; 68:195-201. [PMID: 22546616 PMCID: PMC4489617 DOI: 10.1016/j.neuropharm.2012.04.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 11/19/2022]
Abstract
GABA (gamma-amino-butyric-acid) is the primary inhibitory neurotransmitter in the human brain. It has been proposed that the symptoms of autism spectrum disorders (ASDs) are the result of deficient GABA neurotransmission, possibly including reduced expression of GABAA receptors. However, this hypothesis has not been directly tested in living adults with ASD. In this preliminary investigation, we used Positron Emission Tomography (PET) with the benzodiazepine receptor PET ligand [(11)C]Ro15-4513 to measure α1 and α5 subtypes of the GABAA receptor levels in the brain of three adult males with well-characterized high-functioning ASD compared with three healthy matched volunteers. We found significantly lower [(11)C]Ro15-4513 binding throughout the brain of participants with ASD (p < 0.0001) compared with controls. Planned region of interest analyses also revealed significant reductions in two limbic brain regions, namely the amygdala and nucleus accumbens bilaterally. Further analysis suggested that these results were driven by lower levels of the GABAA α5 subtype. These results provide initial evidence of a GABAA α5 deficit in ASD and support further investigations of the GABA system in this disorder. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Maria Andreina Mendez
- King’s College London, Department of Forensic and Neurodevelopmental, Sciences, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, United Kingdom
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Division of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| | - Jamie Horder
- King’s College London, Department of Forensic and Neurodevelopmental, Sciences, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Jim Myers
- Psychopharmacology Unit, School of Social and Community Medicine, University of Bristol, BF1, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Suzanne Coghlan
- King’s College London, Department of Forensic and Neurodevelopmental, Sciences, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Paul Stokes
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Division of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| | - David Erritzoe
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Division of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| | - Oliver Howes
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| | - Anne Lingford-Hughes
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Division of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| | - Declan Murphy
- King’s College London, Department of Forensic and Neurodevelopmental, Sciences, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, United Kingdom
| | - David Nutt
- Neuropsychopharmacology Unit, Centre for Pharmacology and Therapeutics, Division of Experimental Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, UK W12 0NN
| |
Collapse
|
41
|
Andersson JD, Halldin C. PET radioligands targeting the brain GABAA/benzodiazepine receptor complex. J Labelled Comp Radiopharm 2013; 56:196-206. [DOI: 10.1002/jlcr.3008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 11/15/2012] [Accepted: 11/16/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Jan D. Andersson
- Department of Clinical Neuroscience; Center for Psychiatric Research and Education, Karolinska Institutet; Stockholm; Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience; Center for Psychiatric Research and Education, Karolinska Institutet; Stockholm; Sweden
| |
Collapse
|
42
|
Reducing GABAA α5 receptor-mediated inhibition rescues functional and neuromorphological deficits in a mouse model of down syndrome. J Neurosci 2013; 33:3953-66. [PMID: 23447605 DOI: 10.1523/jneurosci.1203-12.2013] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Down syndrome (DS) is associated with neurological complications, including cognitive deficits that lead to impairment in intellectual functioning. Increased GABA-mediated inhibition has been proposed as a mechanism underlying deficient cognition in the Ts65Dn (TS) mouse model of DS. We show that chronic treatment of these mice with RO4938581 (3-bromo-10-(difluoromethyl)-9H-benzo[f]imidazo[1,5-a][1,2,4]triazolo[1,5-d][1,4]diazepine), a selective GABA(A) α5 negative allosteric modulator (NAM), rescued their deficits in spatial learning and memory, hippocampal synaptic plasticity, and adult neurogenesis. We also show that RO4938581 normalized the high density of GABAergic synapse markers in the molecular layer of the hippocampus of TS mice. In addition, RO4938581 treatment suppressed the hyperactivity observed in TS mice without inducing anxiety or altering their motor abilities. These data demonstrate that reducing GABAergic inhibition with RO4938581 can reverse functional and neuromorphological deficits of TS mice by facilitating brain plasticity and support the potential therapeutic use of selective GABA(A) α5 NAMs to treat cognitive dysfunction in DS.
Collapse
|
43
|
PWZ-029, an inverse agonist selective for α₅ GABAA receptors, improves object recognition, but not water-maze memory in normal and scopolamine-treated rats. Behav Brain Res 2012; 241:206-13. [PMID: 23261875 DOI: 10.1016/j.bbr.2012.12.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 01/03/2023]
Abstract
Inverse agonism at the benzodiazepine site of α(5) subunit-containing GABA(A) receptors is an attractive approach for the development of putative cognition-enhancing compounds, which are still far from clinical application. Several ligands with binding and/or functional selectivity for α(5) GABA(A) receptors have been synthesized and tested in a few animal models. PWZ-029 is an α(5) GABA(A) selective inverse agonist whose memory enhancing effects were demonstrated in the passive avoidance task in rats and in Pavlovian fear conditioning in mice. In the present study we investigated the effects of PWZ-029 administration in novel object recognition test and Morris water maze, in normal and scopolamine-treated rats. All the three doses of PWZ-029 (2, 5 and 10 mg/kg) improved object recognition after the 24-h delay period, as shown by significant differences between the exploration times of the novel and old object, and the respective discrimination indices. PWZ-029 (2 mg/kg) also successfully reversed the 0.3 mg/kg scopolamine-induced deficit in recognition memory after the 1-h delay. In the Morris water maze test, PWZ-029 (5, 10 and 15 mg/kg) did not significantly influence swim patterns, either during five acquisition days or during the treatment-free probe trial. PWZ-029 (2, 5 and 10 mg/kg) also proved to be ineffective in the reversal of the 1mg/kg scopolamine-induced memory impairment in the water maze. The present mixed results encourage use of a variety of tests and experimental conditions in order to increase the predictability of preclinical testing of selective α(5) GABA(A) inverse agonists.
Collapse
|
44
|
Besson M, Daali Y, Di Lio A, Dayer P, Zeilhofer HU, Desmeules J. Antihyperalgesic effect of the GABA(A) ligand clobazam in a neuropathic pain model in mice: a pharmacokinetic-pharmacodynamic study. Basic Clin Pharmacol Toxicol 2012; 112:192-7. [PMID: 23006671 DOI: 10.1111/bcpt.12017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/10/2012] [Indexed: 11/30/2022]
Abstract
Facilitation of spinal GABAergic inhibition with benzodiazepines (BZDs) reverses pain sensitization in animals; however, the use of BZDs in man is limited by their sedative effect. The antihyperalgesic effects of GABA(A) agonists are mediated by GABA(A) receptors containing α2 subunits, whereas sedation is linked to α1 subunit-containing receptors. α2 and α3 selective GABA(A) receptor modulators have been tested in animals but are not yet available for use in human beings. Clobazam is a 1,5-BZD, which exhibits less cognitive side effects than other benzodiazepines. Here, we studied its antihyperalgesic effects in a mouse model of neuropathic pain. Clobazam showed a dose-dependent antihyperalgesic effect in the chronic constriction injury (CCI) model of neuropathic pain, peaking at 1 hr after administration and lasting for 4 hr with no relevant sedation at a dose of 3 mg/kg. At higher doses, the antihyperalgesic effect was stronger, but sedation became significant. The blood and brain kinetics of clobazam were linear over the range of doses tested with a short half-life of the parent compound and a ready penetration of the blood-brain barrier. Clobazam blood concentrations decreased rapidly, falling below the limit of detection at 120 min. after drug application. Its main metabolite, N-desmethyl-clobazam, showed more delayed and prolonged pharmacokinetics, partly explaining why antihyperalgesia persisted when clobazam was no longer detectable in the blood. Considering its therapeutic margin and its pharmacokinetic properties, clobazam would be a valuable compound to assess the role of the GABAergic pathway in pain transmission in human beings.
Collapse
Affiliation(s)
- Marie Besson
- Multidisciplinary Pain Center, Clinical Pharmacology and Toxicology, Geneva University Hospitals, Geneva, Switzerland.
| | | | | | | | | | | |
Collapse
|
45
|
Möhler H. Cognitive enhancement by pharmacological and behavioral interventions: the murine Down syndrome model. Biochem Pharmacol 2012; 84:994-9. [PMID: 22898099 DOI: 10.1016/j.bcp.2012.06.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/29/2012] [Accepted: 06/29/2012] [Indexed: 12/26/2022]
Abstract
The cognitive deficits in Down syndrome (DS) are attributed to an excessive hippocampal inhibition, which obstructs neuronal plasticity and normal learning and memory, a view which is largely based on studies of Ts65Dn mice, the best characterized mouse model of DS. The cognitive behavioral deficits of Ts65Dn mice can be rescued by reducing GABAergic inhibition, most selectively by partial inverse agonists acting on α(5) GABA-A receptors, of which one compound has recently entered clinical trials in DS. Most remarkably, the improved cognitive performance of Ts65Dn can persist for weeks and months after cessation of drug treatment, as demonstrated for the non-specific GABA antagonist pentylenetetrazole. The Alzheimer drugs, memantine and donepezil largely fail to show any benefit. Finally, repeated non-invasive sensory stimulation such as over-training or enriching the environment, are able to enhance the learning performance which underlines the reversibility of an obstructed neuronal plasticity in Ts65Dn mice.
Collapse
Affiliation(s)
- Hanns Möhler
- Institute of Pharmacology, University of Zurich and, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.
| |
Collapse
|
46
|
Redrobe JP, Elster L, Frederiksen K, Bundgaard C, de Jong IEM, Smith GP, Bruun AT, Larsen PH, Didriksen M. Negative modulation of GABAA α5 receptors by RO4938581 attenuates discrete sub-chronic and early postnatal phencyclidine (PCP)-induced cognitive deficits in rats. Psychopharmacology (Berl) 2012; 221:451-68. [PMID: 22124672 DOI: 10.1007/s00213-011-2593-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 11/15/2011] [Indexed: 11/29/2022]
Abstract
RATIONALE A growing body of evidence suggests that negative modulation of γ-aminobutyric acid (GABA) GABA(A) α5 receptors may be a promising strategy for the treatment of certain facets of cognitive impairment; however, selective modulators of GABA(A) α5 receptors have not yet been tested in "schizophrenia-relevant" cognitive assay/model systems in animals. OBJECTIVES The objectives of this study were to investigate the potential of RO4938581, a negative modulator of GABA(A) α5 receptors, and to attenuate cognitive impairments induced following sub-chronic (sub-PCP) and early postnatal PCP (neo-PCP) administration in the novel object recognition (NOR) and intra-extradimensional shift (ID/ED) paradigms in rats. Complementary in vitro, ex vivo and in vivo studies were performed to confirm negative modulatory activity of RO4938581 and to investigate animal model validity, concept validity and potential side effect issues, respectively. RESULTS In vitro studies confirmed the reported negative modulatory activity of RO4938581, whilst immunohistochemical analyses revealed significantly reduced parvalbumin-positive cells in the prefrontal cortex of sub-PCP- and neo-PCP-treated rats. RO4938581 (1 mg/kg) ameliorated both sub-PCP- and neo-PCP-induced cognitive deficits in NOR and ID/ED performance, respectively. In contrast, QH-II-066 (1 and 3 mg/kg), a GABA(A) α5 receptor positive modulator, impaired cognitive performance in the NOR task when administered to vehicle-treated animals. Additional studies revealed that both RO4938581 (1 mg/kg) and QH-II-066 (1 and 3 mg/kg) attenuated amphetamine-induced hyperactivity in rats. CONCLUSIONS Taken together, these novel findings suggest that negative modulation of GABA(A) α5 receptors may represent an attractive treatment option for the cognitive impairments, and potentially positive symptoms, associated with schizophrenia.
Collapse
Affiliation(s)
- John P Redrobe
- Synaptic Transmission I, Neuroscience Research DK, H Lundbeck A/S, Ottiliavej 9, 2500 Valby, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Myers JFM, Rosso L, Watson BJ, Wilson SJ, Kalk NJ, Clementi N, Brooks DJ, Nutt DJ, Turkheimer FE, Lingford-Hughes AR. Characterisation of the contribution of the GABA-benzodiazepine α1 receptor subtype to [(11)C]Ro15-4513 PET images. J Cereb Blood Flow Metab 2012; 32:731-44. [PMID: 22214903 PMCID: PMC3318150 DOI: 10.1038/jcbfm.2011.177] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This positron emission tomography (PET) study aimed to further define selectivity of [(11)C]Ro15-4513 binding to the GABARα5 relative to the GABARα1 benzodiazepine receptor subtype. The impact of zolpidem, a GABARα1-selective agonist, on [(11)C]Ro15-4513, which shows selectivity for GABARα5, and the nonselective benzodiazepine ligand [(11)C]flumazenil binding was assessed in humans. Compartmental modelling of the kinetics of [(11)C]Ro15-4513 time-activity curves was used to describe distribution volume (V(T)) differences in regions populated by different GABA receptor subtypes. Those with low α5 were best fitted by one-tissue compartment models; and those with high α5 required a more complex model. The heterogeneity between brain regions suggested spectral analysis as a more appropriate method to quantify binding as it does not a priori specify compartments. Spectral analysis revealed that zolpidem caused a significant V(T) decrease (~10%) in [(11)C]flumazenil, but no decrease in [(11)C]Ro15-4513 binding. Further analysis of [(11)C]Ro15-4513 kinetics revealed additional frequency components present in regions containing both α1 and α5 subtypes compared with those containing only α1. Zolpidem reduced one component (mean±s.d.: 71%±41%), presumed to reflect α1-subtype binding, but not another (13%±22%), presumed to reflect α5. The proposed method for [(11)C]Ro15-4513 analysis may allow more accurate selective binding assays and estimation of drug occupancy for other nonselective ligands.
Collapse
|
48
|
Brickley SG, Mody I. Extrasynaptic GABA(A) receptors: their function in the CNS and implications for disease. Neuron 2012; 73:23-34. [PMID: 22243744 DOI: 10.1016/j.neuron.2011.12.012] [Citation(s) in RCA: 509] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2011] [Indexed: 12/30/2022]
Abstract
Over the past two decades, research has identified extrasynaptic GABA(A) receptor populations that enable neurons to sense the low ambient GABA concentrations present in the extracellular space in order to generate a form of tonic inhibition not previously considered in studies of neuronal excitability. The importance of this tonic inhibition in regulating states of consciousness is highlighted by the fact that extrasynaptic GABA(A) receptors (GABA(A)Rs) are believed to be key targets for anesthetics, sleep-promoting drugs, neurosteroids, and alcohol. The neurosteroid sensitivity of these extrasynaptic GABA(A)Rs may explain their importance in stress-, ovarian cycle-, and pregnancy-related mood disorders. Moreover, disruptions in network dynamics associated with schizophrenia, epilepsy, and Parkinson's disease may well involve alterations in the tonic GABA(A)R-mediated conductance. Extrasynaptic GABA(A)Rs may therefore present a therapeutic target for treatment of these diseases, with the potential to enhance cognition and aid poststroke functional recovery.
Collapse
Affiliation(s)
- Stephen G Brickley
- Division of Cell & Molecular Biology, South Kensington Campus, Imperial College, London SW7 2AZ, UK.
| | | |
Collapse
|
49
|
Improved cognitive performance following supplementation with a mixed-grain diet in high school students: A randomized controlled trial. Nutrition 2012; 28:165-72. [DOI: 10.1016/j.nut.2011.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 01/21/2011] [Accepted: 05/28/2011] [Indexed: 11/19/2022]
|
50
|
Lingford-Hughes A, Reid AG, Myers J, Feeney A, Hammers A, Taylor LG, Rosso L, Turkheimer F, Brooks DJ, Grasby P, Nutt DJ. A [11C]Ro15 4513 PET study suggests that alcohol dependence in man is associated with reduced α5 benzodiazepine receptors in limbic regions. J Psychopharmacol 2012; 26:273-81. [PMID: 20870689 DOI: 10.1177/0269881110379509] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Preclinical evidence suggests the α5 subtype of the GABA-benzodiazepine receptor is involved in some of the actions of alcohol and in memory. The positron emission tomography (PET) tracer, [(11)C]Ro15 4513 shows relative selectivity in labelling the α5 subtype over the other GABA-benzodiazepine receptor subtypes in limbic regions of the brain. We used this tracer to investigate the distribution of α5 subtype availability in human alcohol dependence and its relationship to clinical variables. Abstinent (>6 weeks) alcohol-dependent men and healthy male controls underwent an [(11)C]Ro15 4513 PET scan. We report [(11)C]Ro15 4513 brain uptake for 8 alcohol-dependent men and 11 healthy controls. We found a significant reduction in [(11)C]Ro15 4513 binding in the nucleus accumbens, parahippocampal gyri, right hippocampus and amygdala in the alcohol-dependent compared with the healthy control group. Levels of [(11)C]Ro15 4513 binding in both hippocampi were significantly and positively associated with performance on a delayed verbal memory task in the alcohol-dependent but not the control group. We speculate that the reduced limbic [(11)C]Ro15 4513 binding seen here results from the effects of alcohol, though we cannot currently distinguish whether they are compensatory in nature or evidence of brain toxicity.
Collapse
Affiliation(s)
- Anne Lingford-Hughes
- Psychopharmacology Unit, Dorothy Hodgkin Building, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|