1
|
Lewter LA, Woodhouse K, Tiruveedhula VVNPB, Cook JM, Li JX. Antinociceptive Effects of α2/ α3-Subtype-Selective GABA A Receptor Positive Allosteric Modulators KRM-II-81 and NS16085 in Male Rats: Behavioral Specificity. J Pharmacol Exp Ther 2024; 391:389-398. [PMID: 38670800 PMCID: PMC11585310 DOI: 10.1124/jpet.123.002070] [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] [Received: 12/20/2023] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Recent studies suggest that among the gamma-aminobutyric acid type A (GABAA)receptor subtype heterogeneity, α2/α3 subunits of GABAA receptors mediate pain processing. Therefore, α2/α3 subtype-selective GABAA receptor-positive allosteric modulators (PAMs) may be candidate analgesics. Antinociceptive effects of α2/α3 subtype-selective GABAA receptor PAMs have been reported, but the behavioral effects of these compounds have not been systematically evaluated. This study examined the behavioral effects of two α2/α3 subtype-selective GABAA receptor PAMs, KRM-II-81 and NS16085, in male rats. The antinociceptive effects of KRM-II-81 and NS16085 were examined using rat models of inflammatory (complete Freund's adjuvant) and neuropathic pain (chronic constriction injury). The effect of KRM-II-81 on affective pain was measured using the place escape/avoidance paradigm (PEAP). Rate-response of food-maintained operant responding, horizontal wire test, and the spontaneous alternation T-maze were assessed to study the side-effect profiles of KRM-II-81 and NS16085. The benzodiazepine midazolam was used as a comparator in these studies. KRM-II-81 and NS16085 attenuated mechanical allodynia but not thermal hyperalgesia in both pain states, and their effects were attenuated by the benzodiazepine receptor antagonist flumazenil. KRM-II-81 attenuated affective pain-related behavior in the PEAP test. In the operant responding procedure and horizontal wire test, only midazolam produced significant effects at the dose that produced maximal antinociception. In the T-maze assay, only midazolam significantly decreased the percentage of alternation at an antinociceptive dose. Thus, KRM-II-81 and NS16085 but not midazolam selectively produced antinociceptive effects. Collectively, these data suggest that α2/α3 subtype-selective GABAA PAMs could be a novel class of analgesics and warrant further investigation. SIGNIFICANCE STATEMENT: This study demonstrates that α2/α3 subtype-selective GABAA PAMs KRM-II-81 and NS16085 produce selective antinociceptive effects devoid of sedation, myorelaxation, and cognitive impairment in two rat models of persistent pain. This study supports the development of α2/α3 subtype-selective GABAA PAMs, rather than classical benzodiazepines, as safe and novel analgesics for pain management.
Collapse
Affiliation(s)
- Lakeisha A Lewter
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York (L.A.L., K.W., J.-X.L.); and Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin (V.V.N.P.B.T., J.M.C.)
| | - Kristen Woodhouse
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York (L.A.L., K.W., J.-X.L.); and Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin (V.V.N.P.B.T., J.M.C.)
| | - V V N Phani Babu Tiruveedhula
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York (L.A.L., K.W., J.-X.L.); and Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin (V.V.N.P.B.T., J.M.C.)
| | - James M Cook
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York (L.A.L., K.W., J.-X.L.); and Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin (V.V.N.P.B.T., J.M.C.)
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York (L.A.L., K.W., J.-X.L.); and Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin (V.V.N.P.B.T., J.M.C.)
| |
Collapse
|
2
|
Girotti M, Bulin SE, Carreno FR. Effects of chronic stress on cognitive function - From neurobiology to intervention. Neurobiol Stress 2024; 33:100670. [PMID: 39295772 PMCID: PMC11407068 DOI: 10.1016/j.ynstr.2024.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
Exposure to chronic stress contributes considerably to the development of cognitive impairments in psychiatric disorders such as depression, generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), post-traumatic stress disorder (PTSD), and addictive behavior. Unfortunately, unlike mood-related symptoms, cognitive impairments are not effectively treated by available therapies, a situation in part resulting from a still incomplete knowledge of the neurobiological substrates that underly cognitive domains and the difficulty in generating interventions that are both efficacious and safe. In this review, we will present an overview of the cognitive domains affected by stress with a specific focus on cognitive flexibility, behavioral inhibition, and working memory. We will then consider the effects of stress on neuronal correlates of cognitive function and the factors which may modulate the interaction of stress and cognition. Finally, we will discuss intervention strategies for treatment of stress-related disorders and gaps in knowledge with emerging new treatments under development. Understanding how cognitive impairment occurs during exposure to chronic stress is crucial to make progress towards the development of new and effective therapeutic approaches.
Collapse
Affiliation(s)
| | - Sarah E. Bulin
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| | - Flavia R. Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, 7703 Floyd Curl Dr., San Antonio, TX, 78229, USA
| |
Collapse
|
3
|
Arora I, Mal P, Arora P, Paul A, Kumar M. GABAergic implications in anxiety and related disorders. Biochem Biophys Res Commun 2024; 724:150218. [PMID: 38865810 DOI: 10.1016/j.bbrc.2024.150218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/05/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024]
Abstract
Evidence indicates that anxiety disorders arise from an imbalance in the functioning of brain circuits that govern the modulation of emotional responses to possibly threatening stimuli. The circuits under consideration in this context include the amygdala's bottom-up activity, which signifies the existence of stimuli that may be seen as dangerous. Moreover, these circuits encompass top-down regulatory processes that originate in the prefrontal cortex, facilitating the communication of the emotional significance associated with the inputs. Diverse databases (e.g., Pubmed, ScienceDirect, Web of Science, Google Scholar) were searched for literature using a combination of different terms e.g., "anxiety", "stress", "neuroanatomy", and "neural circuits", etc. A decrease in GABAergic activity is present in both anxiety disorders and severe depression. Research on cerebral functional imaging in depressive individuals has shown reduced levels of GABA within the cortical regions. Additionally, animal studies demonstrated that a reduction in the expression of GABAA/B receptors results in a behavioral pattern resembling anxiety. The amygdala consists of inhibitory networks composed of GABAergic interneurons, responsible for modulating anxiety responses in both normal and pathological conditions. The GABAA receptor has allosteric sites (e.g., α/γ, γ/β, and α/β) which enable regulation of neuronal inhibition in the amygdala. These sites serve as molecular targets for anxiolytic medications such as benzodiazepine and barbiturates. Alterations in the levels of naturally occurring regulators of these allosteric sites, along with alterations to the composition of the GABAA receptor subunits, could potentially act as mechanisms via which the extent of neuronal inhibition is diminished in pathological anxiety disorders.
Collapse
Affiliation(s)
- Indu Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pankaj Mal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Poonam Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Anushka Paul
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
4
|
Daher F, Villalobos NA, Hanley M, Atack JR, Popa MO, Fogaça MV. Basmisanil, an α5-GABA AR negative allosteric modulator, produces rapid and sustained antidepressant-like responses in male mice. Neurosci Lett 2024; 833:137828. [PMID: 38772437 PMCID: PMC11146097 DOI: 10.1016/j.neulet.2024.137828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024]
Abstract
There is a critical need for safer and better-tolerated alternatives to address the current limitations of antidepressant treatments for major depressive disorder. Recently, drugs targeting the GABA system via α5-containing GABAA receptors (α5-GABAAR) as negative allosteric modulators (α5-NAMs) have shown promise in alleviating stress-related behaviors in preclinical studies, suggesting that α5-NAMs may have translational relevance as novel antidepressant medications. Here, we evaluated the efficacy of Basmisanil, an α5-NAM that has been evaluated in Phase 2 clinical studies as a cognitive enhancer, in a battery of behavioral tests relevant to coping strategies, motivation, and aversion in male mice, along with plasma and brain pharmacokinetic measurements. Our findings reveal that Basmisanil induces dose-dependent rapid antidepressant-like responses in the forced swim test and sucrose splash test without promoting locomotor stimulating effects. Furthermore, Basmisanil elicits sustained behavioral responses in the female urine sniffing test and sucrose splash test, observed 24 h and 48 h post-treatment, respectively. Bioanalysis of plasma and brain samples confirms effective blood-brain barrier penetration by Basmisanil and extrapolation to previously published data suggest that effects were observed at doses (10 and 30 mg/kg i.p.) corresponding to relatively modest levels of α5-GABAAR occupancy (40-65 %). These results suggest that Basmisanil exhibits a combination of rapid and sustained antidepressant-like effects highlighting the potential of α5-NAMs as a novel therapeutic strategy for depression.
Collapse
Affiliation(s)
- Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Narcy A Villalobos
- Medicines Discovery Institute, Cardiff University, Park Place, CF10 3AT, Cardiff, Wales, United Kingdom
| | - Marcus Hanley
- Medicines Discovery Institute, Cardiff University, Park Place, CF10 3AT, Cardiff, Wales, United Kingdom
| | - John R Atack
- Medicines Discovery Institute, Cardiff University, Park Place, CF10 3AT, Cardiff, Wales, United Kingdom
| | - Mariana O Popa
- Medicines Discovery Institute, Cardiff University, Park Place, CF10 3AT, Cardiff, Wales, United Kingdom
| | - Manoela V Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
5
|
Pharmacological Signature and Target Specificity of Inhibitory Circuits Formed by Martinotti Cells in the Mouse Barrel Cortex. J Neurosci 2023; 43:14-27. [PMID: 36384682 PMCID: PMC9838699 DOI: 10.1523/jneurosci.1661-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/13/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
In the neocortex, fast synaptic inhibition orchestrates both spontaneous and sensory-evoked activity. GABAergic interneurons (INs) inhibit pyramidal neurons (PNs) directly, modulating their output activity and thus contributing to balance cortical networks. Moreover, several IN subtypes also inhibit other INs, forming specific disinhibitory circuits, which play crucial roles in several cognitive functions. Here, we studied a subpopulation of somatostatin-positive INs, the Martinotti cells (MCs) in layer 2/3 of the mouse barrel cortex (both sexes). MCs inhibit the distal portion of PN apical dendrites, thus controlling dendrite electrogenesis and synaptic integration. Yet, it is poorly understood whether MCs inhibit other elements of the cortical circuits, and the connectivity properties with non-PN targets are unknown. We found that MCs have a strong preference for PN dendrites, but they also considerably connect with parvalbumin-positive, vasoactive intestinal peptide-expressing, and layer 1 (L1) INs. Remarkably, GABAergic synapses from MCs exhibited clear cell type-specific short-term plasticity. Moreover, whereas the biophysical properties of MC-PN synapses were consistent with distal dendritic inhibition, MC-IN synapses exhibited characteristics of fast perisomatic inhibition. Finally, MC-PN connections used α5-containing GABAA receptors (GABAARs), but this subunit was not expressed by the other INs targeted by MCs. We reveal a specialized connectivity blueprint of MCs within different elements of superficial cortical layers. In addition, our results identify α5-GABAARs as the molecular fingerprint of MC-PN dendritic inhibition. This is of critical importance, given the role of α5-GABAARs in cognitive performance and their involvement in several brain diseases.SIGNIFICANCE STATEMENT Martinotti cells (MCs) are a prominent, broad subclass of somatostatin-expressing GABAergic interneurons, specialized in controlling distal dendrites of pyramidal neurons (PNs) and taking part in several cognitive functions. Here we characterize the connectivity pattern of MCs with other interneurons in the superficial layers (L1 and L2/3) of the mouse barrel cortex. We found that the connectivity pattern of MCs with PNs as well as parvalbumin, vasoactive intestinal peptide, and L1 interneurons exhibit target-specific plasticity and biophysical properties. The specificity of α5-GABAARs at MC-PN synapses and the lack or functional expression of this subunit by other cell types define the molecular identity of MC-PN connections and the exclusive involvement of this inhibitory circuits in α5-dependent cognitive tasks.
Collapse
|
6
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
7
|
Amalyan S, Tamboli S, Lazarevich I, Topolnik D, Bouman LH, Topolnik L. Enhanced motor cortex output and disinhibition in asymptomatic female mice with C9orf72 genetic expansion. Cell Rep 2022; 40:111043. [PMID: 35793625 DOI: 10.1016/j.celrep.2022.111043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 04/29/2022] [Accepted: 06/12/2022] [Indexed: 11/28/2022] Open
Abstract
Information and action coding by cortical circuits relies on a balanced dialogue between excitation and inhibition. Circuit hyperexcitability is considered a potential pathophysiological mechanism in various brain disorders, but the underlying deficits, especially at early disease stages, remain largely unknown. We report that asymptomatic female mice carrying the chromosome 9 open reading frame 72 (C9orf72) repeat expansion, which represents a high-prevalence genetic abnormality for human amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) spectrum disorder, exhibit abnormal motor cortex output. The number of primary motor cortex (M1) layer 5 pyramidal neurons is reduced in asymptomatic mice, with the surviving neurons receiving a decreased inhibitory drive that results in a higher M1 output, specifically during high-speed animal locomotion. Importantly, using deep-learning algorithms revealed that speed-dependent M1 output predicts the likelihood of C9orf72 genetic expansion. Our data link early circuit abnormalities with a gene mutation in asymptomatic ALS/FTLD carriers.
Collapse
Affiliation(s)
- Sona Amalyan
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Suhel Tamboli
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Ivan Lazarevich
- École Normale Supérieure, Laboratoire de Neurosciences Cognitives, Group for Neural Theory, Paris, France
| | - Dimitry Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Leandra Harriet Bouman
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Santrač A, Batinić B, Stamenić TT, Aranđelović J, Sharmin D, Knutson DE, Cook JM, Savić MM. Positive modulation of α5GABAA receptors leads to dichotomous effects in rats on memory pattern and GABRA5 expression in prefrontal cortex and hippocampus. Behav Brain Res 2022; 416:113578. [PMID: 34508769 DOI: 10.1016/j.bbr.2021.113578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/22/2021] [Accepted: 09/06/2021] [Indexed: 01/04/2023]
Abstract
Positive allosteric modulators (PAMs) of α5GABAA receptors (α5GABAARs) are emerging as potential therapeutics for a range of neuropsychiatric disorders. However, their role in memory processing of healthy animals is not sufficiently examined. We tested the effects of MP-III-022 (1 mg/kg, 2.5 mg/kg and 10 mg/kg), a PAM known to be selective for α5GABAARs and devoid of prominent side-effects, in different behavioral paradigms (Morris water maze, novel object recognition test and social novelty discrimination) and on GABRA5 expression in Wistar rats, 30 min and 24 h after intraperitoneal treatment administration. The lowest dose tested worsened short-term object memory. The same dose, administered two times in a span of 24 h, improved spatial and impaired object and, at a trend level, social memory. The highest dose had a detrimental effect on all types of long-term memory (object memory at a trend level) and short-term spatial memory, but improved short-term object and social memory. Distinct sets of expression changes were detected in both prefrontal cortex and two regions of the hippocampus, but the latter ones could be assessed as more consequential. An increase of GABRA5 mRNA in CA2 occurred in parallel with improvement of object and social, but impairment of spatial memory, while the opposite happened with a trend level change in CA1. Our study demonstrates the variability of the roles of the α5GABAAR based on its level of expression and localization, in dependence on the type and protocol of cognitive tasks, as well as the respective timing of pharmacological modulation and testing.
Collapse
Affiliation(s)
- Anja Santrač
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Bojan Batinić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Tamara Timić Stamenić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Jovana Aranđelović
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee and the Milwaukee Institute of Drug Discovery, P.O.Box 413, Milwaukee, WI 53201, USA
| | - Daniel E Knutson
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee and the Milwaukee Institute of Drug Discovery, P.O.Box 413, Milwaukee, WI 53201, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee and the Milwaukee Institute of Drug Discovery, P.O.Box 413, Milwaukee, WI 53201, USA
| | - Miroslav M Savić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| |
Collapse
|
11
|
Yuan C, Gao A, Xu Q, Zhang B, Xue R, Dou Y, Yu C. A multi-dosing regimen to enhance the spatial memory of normal rats with α5-containing GABA A receptor negative allosteric modulator L-655,708. Psychopharmacology (Berl) 2021; 238:3375-3389. [PMID: 34389882 DOI: 10.1007/s00213-021-05951-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 08/03/2021] [Indexed: 01/20/2023]
Abstract
RATIONALE AND OBJECTIVES The reported inconsistent effects of negative allosteric modulators of α5-containing GABAA receptors on learning and memory may be attributed to receptor selectivity, effective plasma concentration maintenance, and administration time. This study aimed to compare the effects of L-655,708 administered by single-dosing regimen versus multi-dosing regimen on spatial memory, signaling molecules, and brain functional connectivity. METHODS After comparing the maintenance time of the effective plasma concentration of L-655,708 between multi-dosing and single-dosing regimens, we further compared the effects of the administration of the two regimens at different phases (before-learning, during-learning, and before-probe) of the Morris water maze (MWM) test on the performance of learning and memory and the levels of signaling molecules related to learning and memory in hippocampal tissues. Functional connectivity analyses between hippocampal and cortical regions were performed to further clarify the effects of the multi-dosing regimen. RESULTS The multi-dosing regimen could maintain the effective plasma concentration of L-655,708 much longer than the single-dosing regimen. Only the multi-dosing regimen for L-655,708 administration during the learning period led to significant improvement in spatial memory in the MWM test and increases in levels of glutamate receptors and phosphorylated signaling molecules (p-PKAα, p-CaMKII, and p-CREB-1). Compared with the vehicle control, the multi-dosing regimen increased the functional connectivity of the left hippocampal CA1 with cingulate and motor cortices. CONCLUSIONS A multi-dosing regimen for L-655,708 administered during the learning period is an effective strategy to improve spatial memory, increase signaling molecule levels, and enhance the functional connectivity of the hippocampus.
Collapse
Affiliation(s)
- Congcong Yuan
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - An Gao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Qiang Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Beibei Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Rui Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
12
|
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
|
13
|
Nuwer JL, Brady ML, Povysheva NV, Coyne A, Jacob TC. Sustained treatment with an α5 GABA A receptor negative allosteric modulator delays excitatory circuit development while maintaining GABAergic neurotransmission. Neuropharmacology 2021; 197:108724. [PMID: 34284042 DOI: 10.1016/j.neuropharm.2021.108724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023]
Abstract
α5 subunit GABA type A receptor (GABAAR) preferring negative allosteric modulators (NAMs) are cognitive enhancers with antidepressant-like effects. α5-NAM success in treating mouse models of neurodevelopmental disorders with excessive inhibition have led to Phase 2 clinical trials for Down syndrome. Despite in vivo efficacy, no study has examined the effects of continued α5-NAM treatment on inhibitory and excitatory synapse plasticity to identify mechanisms of action. Here we used L-655,708, an imidazobenzodiazepine that acts as a highly selective but weak α5-NAM, to investigate the impact of sustained treatment on hippocampal neuron synapse and dendrite development. We show that 2-day pharmacological reduction of α5-GABAAR signaling from DIV12-14, when GABAARs contribute to depolarization, delays dendritic spine maturation and the NMDA receptor (NMDAR) GluN2B/GluN2A developmental shift. In contrast, α5-NAM treatment from DIV19-21, when hyperpolarizing GABAAR signaling predominates, enhances surface synaptic GluN2A while decreasing GluN2B. Despite changes in NMDAR subtype surface levels and localization, total levels of key excitatory synapse proteins were largely unchanged, and mEPSCs were unaltered. Importantly, 2-day α5-NAM treatment does not alter the total surface levels or distribution of α5-GABAARs, reduce the gephyrin inhibitory synaptic scaffold, or impair phasic or tonic inhibition. Furthermore, α5-NAM inhibition of the GABAAR tonic current in mature neurons is maintained after 2-day α5-NAM treatment, suggesting reduced tolerance liability, in contrast to other clinically relevant GABAAR-targeting drugs such as benzodiazepines. Together, these results show that α5-GABAARs contribute to dendritic spine maturation and excitatory synapse development via a NMDAR dependent mechanism without perturbing overall neuronal excitability.
Collapse
Affiliation(s)
- Jessica L Nuwer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Megan L Brady
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadya V Povysheva
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amanda Coyne
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Garakani A, Murrough JW, Freire RC, Thom RP, Larkin K, Buono FD, Iosifescu DV. Pharmacotherapy of Anxiety Disorders: Current and Emerging Treatment Options. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2021; 19:222-242. [PMID: 34690588 PMCID: PMC8475923 DOI: 10.1176/appi.focus.19203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
(Appeared originally in Frontiers in Psychiatry 2020 Dec 23; 11:595584)
Collapse
|
15
|
Takamura Y, Kakuta H. In Vivo Receptor Visualization and Evaluation of Receptor Occupancy with Positron Emission Tomography. J Med Chem 2021; 64:5226-5251. [PMID: 33905258 DOI: 10.1021/acs.jmedchem.0c01714] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Positron emission tomography (PET) is useful for noninvasive in vivo visualization of disease-related receptors, for evaluation of receptor occupancy to determine an appropriate drug dosage, and for proof-of-concept of drug candidates in translational research. For these purposes, the specificity of the PET tracer for the target receptor is critical. Here, we review work in this area, focusing on the chemical structures of reported PET tracers, their Ki/Kd values, and the physical properties relevant to target receptor selectivity. Among these physical properties, such as cLogP, cLogD, molecular weight, topological polar surface area, number of hydrogen bond donors, and pKa, we focus especially on LogD and LogP as important physical properties that can be easily compared across a range of studies. We discuss the success of PET tracers in evaluating receptor occupancy and consider likely future developments in the field.
Collapse
Affiliation(s)
- Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
16
|
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
|
17
|
Kwakowsky A, Waldvogel HJ, Faull RLM. Therapeutic potential of alpha 5 subunit containing GABA A receptors in Alzheimer's disease. Neural Regen Res 2021; 16:1550-1551. [PMID: 33433477 PMCID: PMC8323681 DOI: 10.4103/1673-5374.300987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Joy MT, Carmichael ST. Encouraging an excitable brain state: mechanisms of brain repair in stroke. Nat Rev Neurosci 2021; 22:38-53. [PMID: 33184469 PMCID: PMC10625167 DOI: 10.1038/s41583-020-00396-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2020] [Indexed: 02/02/2023]
Abstract
Stroke induces a plastic state in the brain. This period of enhanced plasticity leads to the sprouting of new axons, the formation of new synapses and the remapping of sensory-motor functions, and is associated with motor recovery. This is a remarkable process in the adult brain, which is normally constrained in its levels of neuronal plasticity and connectional change. Recent evidence indicates that these changes are driven by molecular systems that underlie learning and memory, such as changes in cellular excitability during memory formation. This Review examines circuit changes after stroke, the shared mechanisms between memory formation and brain repair, the changes in neuronal excitability that underlie stroke recovery, and the molecular and pharmacological interventions that follow from these findings to promote motor recovery in animal models. From these findings, a framework emerges for understanding recovery after stroke, central to which is the concept of neuronal allocation to damaged circuits. The translation of the concepts discussed here to recovery in humans is underway in clinical trials for stroke recovery drugs.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Manzo MA, Wang DS, Li WW, Pinguelo A, Popa MO, Khodaei S, Atack JR, Ross RA, Orser BA. Inhibition of a tonic inhibitory conductance in mouse hippocampal neurones by negative allosteric modulators of α5 subunit-containing γ-aminobutyric acid type A receptors: implications for treating cognitive deficits. Br J Anaesth 2020; 126:674-683. [PMID: 33388140 DOI: 10.1016/j.bja.2020.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Multiple cognitive and psychiatric disorders are associated with an increased tonic inhibitory conductance that is generated by α5 subunit-containing γ-aminobutyric acid type A (α5 GABAA) receptors. Negative allosteric modulators that inhibit α5 GABAA receptors (α5-NAMs) are being developed as treatments for these disorders. The effects of α5-NAMs have been studied on recombinant GABAA receptors expressed in non-neuronal cells; however, no study has compared drug effects on the tonic conductance generated by native GABAA receptors in neurones, which was the goal of this study. METHODS The effects of five α5-NAMs (basmisanil, Ono-160, L-655,708, α5IA, and MRK-016) on tonic current evoked by a low concentration of GABA were studied using whole-cell recordings in cultured mouse hippocampal neurones. Drug effects on current evoked by a saturating concentration of GABA and on miniature inhibitory postsynaptic currents (mIPSCs) were also examined. RESULTS The α5-NAMs caused a concentration-dependent decrease in tonic current. The potencies varied as the inhibitory concentration for 50% inhibition (IC50) of basmisanil (127 nM) was significantly higher than those of the other compounds (0.4-0.8 nM). In contrast, the maximal efficacies of the drugs were similar (35.5-51.3% inhibition). The α5-NAMs did not modify current evoked by a saturating GABA concentration or mIPSCs. CONCLUSIONS Basmisanil was markedly less potent than the other α5-NAMs, an unexpected result based on studies of recombinant α5 GABAA receptors. Studying the effects of α5 GABAA receptor-selective drugs on the tonic inhibitory current in neurones could inform the selection of compounds for future clinical trials.
Collapse
Affiliation(s)
- Marc A Manzo
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Dian-Shi Wang
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Winston W Li
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Arsène Pinguelo
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Mariana O Popa
- Medicines Discovery Institute, Cardiff University, Cardiff, Wales
| | - Shahin Khodaei
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - John R Atack
- Medicines Discovery Institute, Cardiff University, Cardiff, Wales
| | - Ruth A Ross
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Beverley A Orser
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada; Department of Anesthesia, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| |
Collapse
|
20
|
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
|
21
|
Petrache AL, Khan AA, Nicholson MW, Monaco A, Kuta-Siejkowska M, Haider S, Hilton S, Jovanovic JN, Ali AB. Selective Modulation of α5 GABA A Receptors Exacerbates Aberrant Inhibition at Key Hippocampal Neuronal Circuits in APP Mouse Model of Alzheimer's Disease. Front Cell Neurosci 2020; 14:568194. [PMID: 33262690 PMCID: PMC7686552 DOI: 10.3389/fncel.2020.568194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Selective negative allosteric modulators (NAMs), targeting α5 subunit-containing GABAA receptors (GABAARs) as potential therapeutic targets for disorders associated with cognitive deficits, including Alzheimer's disease (AD), continually fail clinical trials. We investigated whether this was due to the change in the expression of α5 GABAARs, consequently altering synaptic function during AD pathogenesis. Using medicinal chemistry and computational modeling, we developed aqueous soluble hybrids of 6,6-dimethyl-3-(2-hydroxyethyl) thio-1-(thiazol-2-yl)-6,7-dihydro-2-benzothiophene-4(5H)-one, that demonstrated selective binding and high negative allosteric modulation, specifically for the α5 GABAAR subtypes in constructed HEK293 stable cell-lines. Using a knock-in mouse model of AD (APP NL-F/NL-F), which expresses a mutant form of human amyloid-β (Aβ), we performed immunofluorescence studies combined with electrophysiological whole-cell recordings to investigate the effects of our key molecule, α5-SOP002 in the hippocampal CA1 region. In aged APP NL-F/NL-F mice, selective preservation of α5 GABAARs was observed in, calretinin- (CR), cholecystokinin- (CCK), somatostatin- (SST) expressing interneurons, and pyramidal cells. Previously, we reported that CR dis-inhibitory interneurons, specialized in regulating other interneurons displayed abnormally high levels of synaptic inhibition in the APP NL-F/NL-F mouse model, here we show that this excessive inhibition was "normalized" to control values with bath-applied α5-SOP002 (1 μM). However, α5-SOP002, further impaired inhibition onto CCK and pyramidal cells that were already largely compromised by exhibiting a deficit of inhibition in the AD model. In summary, using a multi-disciplinary approach, we show that exposure to α5 GABAAR NAMs may further compromise aberrant synapses in AD. We, therefore, suggest that the α5 GABAAR is not a suitable therapeutic target for the treatment of AD or other cognitive deficits due to the widespread neuronal-networks that use α5 GABAARs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Afia B. Ali
- UCL School of Pharmacy, London, United Kingdom
| |
Collapse
|
22
|
Xu Y, Zhao M, Han Y, Zhang H. GABAergic Inhibitory Interneuron Deficits in Alzheimer's Disease: Implications for Treatment. Front Neurosci 2020; 14:660. [PMID: 32714136 PMCID: PMC7344222 DOI: 10.3389/fnins.2020.00660] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized clinically by severe cognitive deficits and pathologically by amyloid plaques, neuronal loss, and neurofibrillary tangles. Abnormal amyloid β-protein (Aβ) deposition in the brain is often thought of as a major initiating factor in AD neuropathology. However, gamma-aminobutyric acid (GABA) inhibitory interneurons are resistant to Aβ deposition, and Aβ decreases synaptic glutamatergic transmission to decrease neural network activity. Furthermore, there is now evidence suggesting that neural network activity is aberrantly increased in AD patients and animal models due to functional deficits in and decreased activity of GABA inhibitory interneurons, contributing to cognitive deficits. Here we describe the roles played by excitatory neurons and GABA inhibitory interneurons in Aβ-induced cognitive deficits and how altered GABA interneurons regulate AD neuropathology. We also comprehensively review recent studies on how GABA interneurons and GABA receptors can be exploited for therapeutic benefit. GABA interneurons are an emerging therapeutic target in AD, with further clinical trials urgently warranted.
Collapse
Affiliation(s)
- Yilan Xu
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Manna Zhao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Yuying Han
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
23
|
Bi D, Wen L, Wu Z, Shen Y. GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease. Alzheimers Dement 2020; 16:1312-1329. [PMID: 32543726 DOI: 10.1002/alz.12088] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/28/2020] [Accepted: 02/10/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To propose a new hypothesis that GABAergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease (AD). BACKGROUND Synaptic dysfunction and E/I imbalance emerge decades before the appearance of cognitive decline in AD patients, which contribute to neurodegeneration. Initially, E/I imbalance was thought to occur first, due to dysfunction of the glutamatergic and cholinergic systems. However, new evidence has demonstrated that the GABAergic system, the counterpart of E/I balance and the major inhibitory neurotransmitter system in the central nervous system, is altered enormously and that this contributes to E/I imbalance and further AD pathogenesis. NEW HYPOTHESIS Alterations to the GABAergic system, induced by multiple AD pathogenic or risk factors, contribute to E/I imbalance and AD pathogenesis. MAJOR CHALLENGES FOR THE HYPOTHESIS This GABAergic hypothesis accounts for many critical questions and common challenges confronting a new hypothesis of AD pathogenesis. More specifically, it explains why amyloid beta (Aβ), β-secretase (BACE1), apolipoprotein E4 gene (APOE ε4), hyperactive glia cells, contributes to AD pathogenesis and why age and sex are the risk factors of AD. GABAergic dysfunction promotes the spread of Aβ pathology throughout the AD brain and associated cognitive impairments, and the induction of dysfunction induced by these varied risk factors shares this common neurobiology leading to E/I imbalance. In turn, some of these factors exacerbate GABAergic dysfunction and E/I imbalance. Moreover, the GABAergic system modulates various brain functions and thus, the GABAergic hypothesis accounts for nonamnestic manifestations. Furthermore, corrections of E/I balance through manipulation of GABAergic functions have shown positive outcomes in preclinical and clinical studies, suggesting the potential of the GABAergic system as a therapeutic target in AD. LINKAGE TO OTHER MAJOR THEORIES Dysfunction of the GABAergic system is induced by multiple critical signaling pathways, which include the existing major theories of AD pathogenesis, such as the Aβ and neuroinflammation hypotheses. In a new perspective, this GABAergic hypothesis accounts for the E/I imbalance and related excitotoxicity, which contribute to cognitive decline and AD pathogenesis. Therefore, the GABAergic system could be a key target to restore, at least partially, the E/I balance and cognitive function in AD patients.
Collapse
Affiliation(s)
- Danlei Bi
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lang Wen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zujun Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, University of Sciences and Technology of China, Hefei, China.,Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
24
|
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
|
25
|
Koh MT, Branch A, Haberman R, Gallagher M. Significance of inhibitory recruitment in aging with preserved cognition: limiting gamma-aminobutyric acid type A α5 function produces memory impairment. Neurobiol Aging 2020; 91:1-4. [PMID: 32240868 DOI: 10.1016/j.neurobiolaging.2020.02.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/30/2019] [Accepted: 02/22/2020] [Indexed: 01/27/2023]
Abstract
Numerous aging studies have identified a shift in the excitatory/inhibitory (E/I) balance with heightened hippocampal neural activity associated with age-related memory impairment across species, including rats, monkeys, and humans. Neurobiological investigations directed at the hippocampal formation have demonstrated that unimpaired aged rats performing on par with young adult rats in a spatial memory task exhibit gene expression profiles, mechanisms for plasticity, and altered circuit/network function, which are distinct from younger rats. Particularly striking is a convergence of observational evidence that aged unimpaired rats augment recruitment of mechanisms associated with neural inhibition, a finding that may represent an adaptive homeostatic adjustment necessary to maintain neural plasticity and memory function in aging. In this study, we test the effect of limiting inhibition via administration of TB21007, a negative allosteric modulator of the alpha 5 subtype of gamma-aminobutyric acid type A α5 receptor, on a radial arm maze assessment of memory function. Impaired memory performance produced by this intervention in otherwise high-performing aged rats supports an adaptive role for gamma-aminobutyric acid in the functional maintenance of intact cognition in aging.
Collapse
Affiliation(s)
- Ming Teng Koh
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Audrey Branch
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Rebecca Haberman
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA.
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
26
|
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
|
27
|
Garakani A, Murrough JW, Freire RC, Thom RP, Larkin K, Buono FD, Iosifescu DV. Pharmacotherapy of Anxiety Disorders: Current and Emerging Treatment Options. Front Psychiatry 2020; 11:595584. [PMID: 33424664 PMCID: PMC7786299 DOI: 10.3389/fpsyt.2020.595584] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Anxiety disorders are the most prevalent psychiatric disorders and a leading cause of disability. While there continues to be expansive research in posttraumatic stress disorder (PTSD), depression and schizophrenia, there is a relative dearth of novel medications under investigation for anxiety disorders. This review's first aim is to summarize current pharmacological treatments (both approved and off-label) for panic disorder (PD), generalized anxiety disorder (GAD), social anxiety disorder (SAD), and specific phobias (SP), including selective serotonin reuptake inhibitors (SSRIs), serotonin norepinephrine reuptake inhibitors (SNRIs), azapirones (e.g., buspirone), mixed antidepressants (e.g., mirtazapine), antipsychotics, antihistamines (e.g., hydroxyzine), alpha- and beta-adrenergic medications (e.g., propranolol, clonidine), and GABAergic medications (benzodiazepines, pregabalin, and gabapentin). Posttraumatic stress disorder and obsessive-compulsive disorder are excluded from this review. Second, we will review novel pharmacotherapeutic agents under investigation for the treatment of anxiety disorders in adults. The pathways and neurotransmitters reviewed include serotonergic agents, glutamate modulators, GABAergic medications, neuropeptides, neurosteroids, alpha- and beta-adrenergic agents, cannabinoids, and natural remedies. The outcome of the review reveals a lack of randomized double-blind placebo- controlled trials for anxiety disorders and few studies comparing novel treatments to existing anxiolytic agents. Although there are some recent randomized controlled trials for novel agents including neuropeptides, glutamatergic agents (such as ketamine and d-cycloserine), and cannabinoids (including cannabidiol) primarily in GAD or SAD, these trials have largely been negative, with only some promise for kava and PH94B (an inhaled neurosteroid). Overall, the progression of current and future psychopharmacology research in anxiety disorders suggests that there needs to be further expansion in research of these novel pathways and larger-scale studies of promising agents with positive results from smaller trials.
Collapse
Affiliation(s)
- Amir Garakani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Silver Hill Hospital, New Canaan, CT, United States.,Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - James W Murrough
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rafael C Freire
- Department of Psychiatry and Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Robyn P Thom
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kaitlyn Larkin
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Frank D Buono
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dan V Iosifescu
- Clinical Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.,Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Jacob TC. Neurobiology and Therapeutic Potential of α5-GABA Type A Receptors. Front Mol Neurosci 2019; 12:179. [PMID: 31396049 PMCID: PMC6668551 DOI: 10.3389/fnmol.2019.00179] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/08/2019] [Indexed: 01/11/2023] Open
Abstract
α5 subunit containing GABA type A receptors (GABAARs) have long been an enigmatic receptor subtype of interest due to their specific brain distribution, unusual surface localization and key role in synaptic plasticity, cognition and memory. These receptors are uniquely positioned to sculpt both the developing and mature hippocampal circuitry due to high overall expression and a distinct peak within the critical synapse formation period during the second postnatal week. Unlike the majority of other GABAARs, they exhibit both receptor clustering at extrasynaptic sites via interactions with the radixin scaffold as well as synaptic sites via gephyrin, thus contributing respectively to tonic currents and synaptic GABAergic neurotransmission. α5 GABAAR signaling can be altered in neurodevelopmental disorders including autism and mental retardation and by inflammation in CNS injury and disease. Due to the unique physiology and pharmacology of α5 GABAARs, drugs targeting these receptors are being developed and tested as treatments for neurodevelopmental disorders, depression, schizophrenia, and mild cognitive impairment. This review article focuses on advances in understanding how the α5 subunit contributes to GABAAR neurobiology. In particular, I discuss both recent insights and remaining knowledge gaps for the functional role of these receptors, pathologies associated with α5 GABAAR dysfunction, and the effects and potential therapeutic uses of α5 receptor subtype targeted drugs.
Collapse
Affiliation(s)
- Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
31
|
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]
|
32
|
Chandler CM, Reeves-Darby J, Jones SA, McDonald JA, Li G, Rahman MT, Cook JM, Platt DM. α5GABA A subunit-containing receptors and sweetened alcohol cue-induced reinstatement and active sweetened alcohol self-administration in male rats. Psychopharmacology (Berl) 2019; 236:1797-1806. [PMID: 30637435 PMCID: PMC6606346 DOI: 10.1007/s00213-018-5163-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022]
Abstract
RATIONALE GABAA receptors containing the α5 subunit (i.e., α5GABAA receptors) appear to be critically involved in the reinforcing and subjective effects of alcohol. Their role in alcohol relapse remains unknown. OBJECTIVES Pharmacological approaches were used to probe the role of α5GABAA receptors in alcohol seeking induced by re-exposure to a sweetened alcohol-paired cue, as well as in alcohol + sucrose vs. sucrose self-administration. METHODS For reinstatement studies, rats were trained to self-administer alcohol under a fixed-ratio schedule in which responding was maintained by alcohol + sucrose deliveries and an alcohol-paired stimulus. Sweetened alcohol seeking was extinguished by eliminating solution deliveries and the sweetened alcohol-paired stimulus. During reinstatement tests, animals received pretreatments of an α5GABAA inverse agonist (L-655,708) or an agonist (QH-ii-066) prior to sessions in which presentation of the sweetened alcohol-paired stimulus was restored, but no solution was delivered. For self-administration studies, rats were trained to self-administer alcohol + sucrose or sucrose under a fixed-ratio schedule. Once stable, animals received pretreatments of QH-ii-066, L-655,708, the inverse agonist RY-023, or naltrexone. RESULTS L-655,708 attenuated reinstatement of sweetened alcohol seeking by alcohol + sucrose-paired cues; whereas sweetened alcohol-seeking behavior was augmented by QH-ii-066, albeit at different doses in different rats. Both L-655,708 and RY-023 selectively reduced alcohol + sucrose vs. sucrose self-administration. In contrast, naltrexone reduced both alcohol + sucrose and sucrose self-administration; whereas QH-ii-066 enhanced sucrose self-administration only. CONCLUSIONS α5GABAA receptors play a key role in the modulation of sweetened alcohol cue-induced reinstatement, as well as in alcohol + sucrose but not sucrose self-administration. Inverse agonist activity at α5GABAA receptors may offer a novel strategy for both the reduction of problematic drinking and the prevention of relapse.
Collapse
Affiliation(s)
- Cassie M. Chandler
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jaren Reeves-Darby
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Sherman A. Jones
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - J. Abigail McDonald
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Guanguan Li
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| | - Md T. Rahman
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| | - James M. Cook
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53211
| | - Donna M. Platt
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216,Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216,Correspondence: Donna M. Platt, Ph.D.; Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 North State St., Jackson, MS 39216, USA; phone: 601-984-5896; fax: 601-984-5899;
| |
Collapse
|
33
|
Kwakowsky A, Calvo-Flores Guzmán B, Pandya M, Turner C, Waldvogel HJ, Faull RL. GABA A receptor subunit expression changes in the human Alzheimer's disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus. J Neurochem 2019; 145:374-392. [PMID: 29485232 DOI: 10.1111/jnc.14325] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/17/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. GABA type A receptors (GABAA Rs) are severely affected in Alzheimer's disease (AD). However, the distribution and subunit composition of GABAA Rs in the AD brain are not well understood. This is the first comprehensive study to show brain region- and cell layer-specific alterations in the expression of the GABAA R subunits α1-3, α5, β1-3 and γ2 in the human AD hippocampus, entorhinal cortex and superior temporal gyrus. In late-stage AD tissue samples using immunohistochemistry we found significant alteration of all investigated GABAA Rs subunits except for α3 and β1 that were well preserved. The most prominent changes include an increase in GABAA R α1 expression associated with AD in all layers of the CA3 region, in the stratum (str.) granulare and hilus of the dentate gyrus. We found a significant increase in GABAA R α2 expression in the str. oriens of the CA1-3, str. radiatum of the CA2,3 and decrease in the str. pyramidale of the CA1 region in AD cases. In AD there was a significant increase in GABAA R α5 subunit expression in str. pyramidale, str. oriens of the CA1 region and decrease in the superior temporal gyrus. We also found a significant decrease in the GABAA R β3 subunit immunoreactivity in the str. oriens of the CA2, str. granulare and str. moleculare of the dentate gyrus. In conclusion, these findings indicate that the expression of the GABAA R subunits shows brain region- and layer-specific alterations in AD, and these changes could significantly influence and alter GABAA R function in the disease. Cover Image for this issue: doi: 10.1111/jnc.14179.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Madhavi Pandya
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Branch A, Monasterio A, Blair G, Knierim JJ, Gallagher M, Haberman RP. Aged rats with preserved memory dynamically recruit hippocampal inhibition in a local/global cue mismatch environment. Neurobiol Aging 2019; 76:151-161. [PMID: 30716540 DOI: 10.1016/j.neurobiolaging.2018.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 10/27/2022]
Abstract
Similar to elderly humans, aged outbred Long-Evans rats exhibit individual differences in memory abilities, including a subset of aged rats that maintain memory function on par with young adults. Such individuals provide a basis for investigating mechanisms of resilience to age-related decline. The present study examined hippocampal gene expression in young adults and aged rats with preserved memory function under behavioral task conditions well established for assessing information processing central to the formation of episodic memory. Although behavioral measures and hippocampal gene induction associated with neural activity and synaptic plasticity were similar across age groups, a marker for inhibitory interneuron function in the hippocampal formation was distinctively increased only in aged rats but not in young adults. Because heightened hippocampal neural activity is associated with age-related memory impairment across species, including rats, monkeys, and humans, this finding may represent an adaptive homeostatic adjustment necessary to maintain neural plasticity and memory function in aging.
Collapse
Affiliation(s)
- Audrey Branch
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Amy Monasterio
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Grace Blair
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - James J Knierim
- Department of Neuroscience and Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Rebecca P Haberman
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
35
|
Mohamad FH, Has ATC. The α5-Containing GABA A Receptors-a Brief Summary. J Mol Neurosci 2019; 67:343-351. [PMID: 30607899 DOI: 10.1007/s12031-018-1246-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
GABAA receptors are the major inhibitory neurotransmitter receptor in the human brain. The receptors are assembled from combination of protein subunits in pentameric complex which may consist of α1-6, β1-3, γ1-3, ρ1-3, δ, ε, θ, or π subunits. There are a theoretical > 150,000 possible assemblies and arrangements of GABAA subunits, although only a few combinations have been found in human with the most dominant consists of 2α1, 2β2, and 1γ2 in a counterclockwise arrangement as seen from the synaptic cleft. The receptors also possess binding sites for various unrelated substances including benzodiazepines, barbiturates, and anesthetics. The α5-containing GABAARs only make up ≤ 5% of the entire receptor population, but up to 25% of the receptor subtype is located in the crucial learning and memory-associated area of the brain-the hippocampus, which has ignited myriads of hypotheses and theories in regard to its role. As well as exhibiting synaptic phasic inhibition, the α5-containing receptors are also extrasynaptic and mediate tonic inhibition with continuously occurring smaller amplitude. Studies on negative-allosteric modulators for reducing this tonic inhibition have been shown to enhance learning and memory in neurological disorders such as schizophrenia, Down syndrome, and autism with a possible alternative benzodiazepine binding site. Therefore, a few α5 subunit-specific compounds have been developed to address these pharmacological needs. With its small population, the α5-containing receptors could be the key and also the answer for many untreated cognitive dysfunctions and disorders.
Collapse
Affiliation(s)
- Fatin H Mohamad
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kampus Kesihatan, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Tarmizi Che Has
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kampus Kesihatan, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
36
|
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
|
37
|
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]
|
38
|
Sieghart W, Savić MM. International Union of Basic and Clinical Pharmacology. CVI: GABA A Receptor Subtype- and Function-selective Ligands: Key Issues in Translation to Humans. Pharmacol Rev 2018; 70:836-878. [PMID: 30275042 DOI: 10.1124/pr.117.014449] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
GABAA receptors are the major inhibitory transmitter receptors in the brain. They are ligand-gated chloride channels and the site of action of benzodiazepines, barbiturates, neuroactive steroids, anesthetics, and convulsants. GABAA receptors are composed of five subunits that can belong to different subunit classes. The existence of 19 homologous subunits and their distinct regional, cellular, and subcellular distribution gives rise to a large number of GABAA receptor subtypes with distinct pharmacology, which modulate different functions of the brain. A variety of compounds have been identified that were claimed to modulate selectively individual GABAA receptor subtypes. However, many of these compounds have only incompletely been investigated or, in addition to a preferential modulation of a receptor subtype, also modulate other subtypes at similar concentrations. Although their differential efficacy at distinct receptor subtypes reduced side effects in behavioral experiments in rodents, the exact receptor subtypes mediating their behavioral effects cannot be unequivocally delineated. In addition, the discrepant in vivo effects of some of these compounds in rodents and man raised doubts on the applicability of the concept of receptor subtype selectivity as a guide for the development of clinically useful drugs. Here, we provide an up-to-date review on the currently available GABAA receptor subtype-selective ligands. We present data on their actual activity at GABAA receptor subtypes, discuss the translational aspect of subtype-selective drugs, and make proposals for the future development of ligands with better anxioselectivity in humans. Finally, we discuss possible ways to strengthen the conclusions of behavioral studies with the currently available drugs.
Collapse
Affiliation(s)
- Werner Sieghart
- Center for Brain Research, Medical University of Vienna, Vienna, Austria (W.S.) and Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia (M.M.S.)
| | - Miroslav M Savić
- Center for Brain Research, Medical University of Vienna, Vienna, Austria (W.S.) and Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia (M.M.S.)
| |
Collapse
|
39
|
Szodorai E, Bampali K, Romanov RA, Kasper S, Hökfelt T, Ernst M, Lubec G, Harkany T. Diversity matters: combinatorial information coding by GABA A receptor subunits during spatial learning and its allosteric modulation. Cell Signal 2018; 50:142-159. [DOI: 10.1016/j.cellsig.2018.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 01/11/2023]
|
40
|
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
|
41
|
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: 28] [Impact Index Per Article: 4.0] [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
|
42
|
Kawaharada S, Nakanishi M, Nakanishi N, Hazama K, Higashino M, Yasuhiro T, Lewis A, Clark GS, Chambers MS, Maidment SA, Katsumata S, Kaneko S. ONO-8590580, a Novel GABA Aα5 Negative Allosteric Modulator Enhances Long-Term Potentiation and Improves Cognitive Deficits in Preclinical Models. J Pharmacol Exp Ther 2018; 366:58-65. [PMID: 29674331 DOI: 10.1124/jpet.117.247627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/17/2018] [Indexed: 03/08/2025] Open
Abstract
GABAA receptors containing α5 subunits (GABAAα5) are highly expressed in the hippocampus and negatively involved in memory processing, as shown by the fact that GABAAα5-deficient mice show higher hippocampus-dependent performance than wild-type mice. Accordingly, small-molecule GABAAα5 negative allosteric modulators (NAMs) are known to enhance spatial learning and memory in rodents. Here we introduce a new, orally available GABAAα5 NAM that improves hippocampal functions. ONO-8590580 [1-(cyclopropylmethyl)-5-fluoro-4-methyl-N-[5-(1-methyl-1H-imidazol-4-yl)-2-pyridinyl]-1H-benzimidazol-6-amine] binds to the benzodiazepine binding sites on recombinant human α5-containing GABAA receptors with a Ki of 7.9 nM, and showed functionally selective GABAAα5 NAM activity for GABA-induced Cl- channel activity with a maximum 44.4% inhibition and an EC50 of 1.1 nM. In rat hippocampal slices, tetanus-induced long-term potentiation of CA1 synapse response was significantly augmented in the presence of 300 nM ONO-8590580. Orally administered ONO-8590580 (1-20 mg/kg) dose-dependently occupied hippocampal GABAAα5 in a range of 40%-90% at 1 hour after intake. In the rat passive avoidance test, ONO-8590580 (3-20 mg/kg, by mouth) significantly prevented (+)-MK-801 hydrogen maleate (MK-801)-induced memory deficit. In addition, ONO-8590580 (20 mg/kg, p.o.) was also effective in improving the cognitive deficit induced by scopolamine and MK-801 in the rat eight-arm radial maze test with equal or greater activity than 0.5 mg/kg donepezil. No anxiogenic-like or proconvulsant effect was associated with ONO-8590580 at 20 mg/kg p.o. in the elevated plus maze test or pentylenetetrazole-induced seizure test, respectively. In sum, ONO-8590580 is a novel GABAAα5 NAM that enhances hippocampal memory function without an anxiogenic or proconvulsant risk.
Collapse
Affiliation(s)
- Soichi Kawaharada
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Miki Nakanishi
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Nobuto Nakanishi
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Keisuke Hazama
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Masato Higashino
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Tetsuya Yasuhiro
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Arwel Lewis
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Gary S Clark
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Mark S Chambers
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Scott A Maidment
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Seishi Katsumata
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| | - Shuji Kaneko
- Discovery Research Laboratories I (So.K., M.N., N.N., K.H., T.Y., Se.K.) and Medicinal Chemistry Research Laboratories (M.H.), ONO Pharmaceutical Co., Ltd., Shimamoto-cho, Mishima-gun, Osaka, Japan; Charles River Laboratories International, Inc., Saffron Walden, Essex, United Kingdom (A.L., G.S.C., M.S.C., S.A.M.); and Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan (So.K., Sh.K.)
| |
Collapse
|
43
|
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: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022]
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
|
44
|
Zorrilla de San Martin J, Delabar JM, Bacci A, Potier MC. GABAergic over-inhibition, a promising hypothesis for cognitive deficits in Down syndrome. Free Radic Biol Med 2018; 114:33-39. [PMID: 28993272 DOI: 10.1016/j.freeradbiomed.2017.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/01/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022]
Abstract
Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability. It is also a model human disease for exploring consequences of gene dosage imbalance on complex phenotypes. Learning and memory impairments linked to intellectual disabilities in DS could result from synaptic plasticity deficits and excitatory-inhibitory alterations leading to changes in neuronal circuitry in the brain of affected individuals. Increasing number of studies in mouse and cellular models converge towards the assumption that excitatory-inhibitory imbalance occurs in DS, likely early during development. Thus increased inhibition appears to be a common trend that could explain synaptic and circuit disorganization. Interestingly using several potent pharmacological tools, preclinical studies strongly demonstrated that cognitive deficits could be restored in mouse models of DS. Clinical trials have not yet provided robust data for therapeutic application and additional studies are needed. Here we review the literature and our own published work emphasizing the over-inhibition hypothesis in DS and their links with gene dosage imbalance paving the way for future basic and clinical research.
Collapse
Affiliation(s)
- Javier Zorrilla de San Martin
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Jean-Maurice Delabar
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Alberto Bacci
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Marie-Claude Potier
- INSERM U1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.
| |
Collapse
|
45
|
Pandit S, Lee GS, Park JB. Developmental changes in GABA A tonic inhibition are compromised by multiple mechanisms in preadolescent dentate gyrus granule cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:695-702. [PMID: 29200913 PMCID: PMC5709487 DOI: 10.4196/kjpp.2017.21.6.695] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 01/28/2023]
Abstract
The sustained tonic currents (Itonic) generated by γ-aminobutyric acid A receptors (GABAARs) are implicated in diverse age-dependent brain functions. While various mechanisms regulating Itonic in the hippocampus are known, their combined role in Itonic regulation is not well understood in different age groups. In this study, we demonstrated that a developmental increase in GABA transporter (GAT) expression, combined with gradual decrease in GABAAR α5 subunit, resulted in various Itonic in the dentate gyrus granule cells (DGGCs) of preadolescent rats. Both GAT-1 and GAT-3 expression gradually increased at infantile (P6-8 and P13-15) and juvenile (P20-22 and P27-29) stages, with stabilization observed thereafter in adolescents (P34-36) and young adults (P41-43). Itonic facilitation of a selective GAT-1 blocker (NO-711) was significantly less at P6-8 than after P13-15. The facilitation of Itonic by SNAP-5114, a GAT-3 inhibitor, was negligible in the absence of exogenous GABA at all tested ages. In contrast, Itonic in the presence of a nonselective GAT blocker (nipecotic acid, NPA) gradually decreased with age during the preadolescent period, which was mimicked by Itonic changes in the presence of exogenous GABA. Itonic sensitivity to L-655,708, a GABAAR α5 subunit inverse agonist, gradually decreased during the preadolescent period in the presence of NPA or exogenous GABA. Finally, Western blot analysis showed that the expression of the GABAAR α5 subunit in the dentate gyrus gradually decreased with age. Collectively, our results suggested that the Itonic regulation of altered GATs is under the final tune of GABAAR α5 subunit activation in DGGCs at different ages.
Collapse
Affiliation(s)
- Sudip Pandit
- Department of Physiology, School of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Gyu Seung Lee
- Department of Physiology, School of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Jin Bong Park
- Department of Physiology, School of Medicine and Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
46
|
Towards a Better Understanding of GABAergic Remodeling in Alzheimer's Disease. Int J Mol Sci 2017; 18:ijms18081813. [PMID: 28825683 PMCID: PMC5578199 DOI: 10.3390/ijms18081813] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022] Open
Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the vertebrate brain. In the past, there has been a major research drive focused on the dysfunction of the glutamatergic and cholinergic neurotransmitter systems in Alzheimer’s disease (AD). However, there is now growing evidence in support of a GABAergic contribution to the pathogenesis of this neurodegenerative disease. Previous studies paint a complex, convoluted and often inconsistent picture of AD-associated GABAergic remodeling. Given the importance of the GABAergic system in neuronal function and homeostasis, in the maintenance of the excitatory/inhibitory balance, and in the processes of learning and memory, such changes in GABAergic function could be an important factor in both early and later stages of AD pathogenesis. Given the limited scope of currently available therapies in modifying the course of the disease, a better understanding of GABAergic remodeling in AD could open up innovative and novel therapeutic opportunities.
Collapse
|
47
|
Chen X, Keramidas A, Lynch JW. Physiological and pharmacological properties of inhibitory postsynaptic currents mediated by α5β1γ2, α5β2γ2 and α5β3γ2 GABA A receptors. Neuropharmacology 2017; 125:243-253. [PMID: 28757051 DOI: 10.1016/j.neuropharm.2017.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
α5-containing GABAARs are potential therapeutic targets for clinical conditions including age-related dementia, stroke, schizophrenia, Down syndrome, anaesthetic-induced amnesia, anxiety and pain. α5-containing GABAARs are expressed in layer 5 cortical neurons and hippocampal pyramidal neurons where they mediate both tonic currents and slow inhibitory postsynaptic currents (IPSCs). A range of drugs has been developed to specifically modulate these receptors. The main α5-containing GABAARs that are likely to exist in vivo are the α5β1γ2, α5β2γ2 and α5β3γ2 isoforms. We currently have few clues as to how these isoforms are distributed between synaptic and extrasynaptic compartments or their relative roles in controlling neuronal excitability. Accordingly, the aim of this study was to define the basic biophysical and pharmacological properties of IPSCs mediated by the three isoforms in a hippocampal neuron-HEK293 cell co-culture assay. The IPSC decay time constants were slow (α5β1γ2L: 45 ms; α5β1γ2L: 80 ms; α5β3γ2L: 184 ms) and were largely dominated by the intrinsic channel deactivation rates. By comparing IPSC rise times, we inferred that α5β1γ2L GABAARs are located postsynaptically whereas the other two are predominantly perisynaptic. α5β3γ2L GABAARs alone mediated tonic currents. We quantified the effects of four α5-specific inverse agonists (TB-21007, MRK-016, α5IA and L-655708) on IPSCs mediated by the three isoforms. All compounds selectively inhibited IPSC amplitudes and accelerated IPSC decay rates, albeit with distinct isoform specificities. MRK-016 also significantly accelerated IPSC rise times. These results provide a reference for future studies seeking to identify and characterize the properties of IPSCs mediated by α5-containing GABAAR isoforms in neurons.
Collapse
Affiliation(s)
- Xiumin Chen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Angelo Keramidas
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joseph W Lynch
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
48
|
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
|
49
|
Abstract
Learning and memory are dependent on interactive excitatory and inhibitory mechanisms. In this review, we discuss a mechanism called disinhibition, which is the release of an inhibitory constraint that effectively results in an increased activity in the target neurons (for example, principal or projection neurons). We focus on discussing the role of disinhibition in learning and memory at a basic level and in disease models with cognitive deficits and highlight a strategy to reverse cognitive deficits caused by excess inhibition, through disinhibition of α5-containing GABA
A receptors mediating tonic inhibition in the hippocampus, based on subtype-selective negative allosteric modulators as a novel class of drugs.
Collapse
Affiliation(s)
- Hanns Möhler
- Institute of Pharmacology, University of Zurich, Zurich, Switzerland; Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, characterized by the loss of memory, multiple cognitive impairments and changes in the personality and behavior. Several decades of intense research have revealed that multiple cellular changes are involved in disease process, including synaptic damage, mitochondrial abnormalities and inflammatory responses, in addition to formation and accumulation of amyloid-β (Aβ) and phosphorylated tau. Although tremendous progress has been made in understanding the impact of neurotransmitters in the progression and pathogenesis of AD, we still do not have a drug molecule associated with neurotransmitter(s) that can delay disease process in elderly individuals and/or restore cognitive functions in AD patients. The purpose of our article is to assess the latest developments in neurotransmitters research using cell and mouse models of AD. We also updated the current status of clinical trials using neurotransmitters' agonists/antagonists in AD.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P. Hemachandra Reddy
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|