1
|
McKenna MC, Sonnewald U, Waageptersen HS, White HS. A tribute to Arne Schousboe's contributions to neurochemistry and his innovative and enduring research in GABA, glutamate, and brain energy metabolism. J Neurochem 2025; 169:e16207. [PMID: 39183580 DOI: 10.1111/jnc.16207] [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: 07/19/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
This is a tribute to Arne Schousboe, Professor Emeritus at the University of Copenhagen, an eminent neurochemist and neuroscientist who was a leader in the fields of GABA, glutamate, and brain energy metabolism. Arne was known for his keen intellect, his wide-ranging expertise in neurochemistry and neuropharmacology of GABA and glutamate and brain energy metabolism. Arne was also known for his strong leadership, his warm and engaging personality and his enjoyment of fine wine and great food shared with friends, family, and colleagues. Sadly, Arne passed away on February 27, 2024, after a short illness. He is survived by his wife Inger Schousboe, his two children, and three wonderful grandchildren. His death is a tremendous loss to the neuroscience community. He will be greatly missed by his friends, family, and colleagues. Some of the highlights of Arne's career are described in this tribute.
Collapse
Affiliation(s)
- Mary C McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ursula Sonnewald
- Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - H Steve White
- Department of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Mortensen JS, Mikkelsen ANL, Wellendorph P. Ways of modulating GABA transporters to treat neurological disease. Expert Opin Ther Targets 2024; 28:529-543. [PMID: 39068514 DOI: 10.1080/14728222.2024.2383611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION The main inhibitory neurotransmitter in the central nervous system (CNS), γ-aminobutyric acid (GABA), is involved in a multitude of neurological and psychiatric disorders characterized by an imbalance in excitatory and inhibitory signaling. Regulation of extracellular levels of GABA is maintained by the four GABA transporters (GATs; GAT1, GAT2, GAT3, and BGT1), Na+/Cl--coupled transporters of the solute carrier 6 (SLC6) family. Despite mounting evidence for the involvement of the non-GAT1 GABA transporters in diseases, only GAT1 has successfully been translated into clinical practice via the drug tiagabine. AREAS COVERED In this review, all four GATs will be described in terms of their involvement in disease, and the most recent data on structure, function, expression, and localization discussed in relation to their potential role as drug targets. This includes an overview of various ways to modulate the GATs in relation to treatment of diseases caused by imbalances in the GABAergic system. EXPERT OPINION The recent publication of various GAT1 structures is an important milestone for future development of compounds targeting the GATs. Such information can provide much needed insight into mechanistic aspects of all GAT subtypes and be utilized to design improved ligands for this highly interesting drug target class.
Collapse
Affiliation(s)
- Jonas S Mortensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amalie N L Mikkelsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Singh GK, Kumari B, Das N, Zaman K, Prasad P, Singh RB. Design, synthesis, molecular docking and pharmacological evaluation of some thiadiazole based nipecotic acid derivatives as a potential anticonvulsant and antidepressant agents. 3 Biotech 2024; 14:71. [PMID: 38362592 PMCID: PMC10864245 DOI: 10.1007/s13205-023-03897-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 12/18/2023] [Indexed: 02/17/2024] Open
Abstract
In our continuous effort to develop novel antiepileptic drug, a new series of nipecotic acid derivatives having1,3,4-thiadiazole nucleus were designed and synthesized. This study aims to improve the lipophilicity of nipecotic acid by attaching some lipophilic anchors like thiadiazole and substituted aryl acid derivatives. In our previous study, we noticed that the N-substituted oxadiazole derivative of nipecotic acid exhibited significant antiepileptic activity in the rodent model. The synthesized compounds were characterized by FT-IR, 1H-NMR, 13C-NMR, Mass, and elemental analysis. The anticonvulsant activity was evaluated by using the maximal electroshock-induced seizure model in rats (MES) and the subcutaneous pentylenetetrazol (scPTZ) test in mice. None of the compounds were found to be active in the MES model whereas compounds (TN2, TN9, TN12, TN13, and TN15) produced significant protection against the scPTZ-induced seizures model. The compounds showing antiepileptic activity were additionally evaluated for antidepressant activity by using the forced swim test, 5-hydroxytryptophan (5-HTP)-induced head twitch test, and learned helplessness test. All the molecules that showed anticonvulsant activity (TN2, TN9, TN12, TN13, and TN15), also exerted significant antidepressant effects in the animal models. The selected compounds were subjected to different toxicity studies. Compounds were found to have no neurotoxicity in the rota-rod test and devoid of hepatic and renal toxicity in 30 days repeated oral toxicity test. Further, a homology model was developed to perform the in-silico molecular docking and dynamics studies which revealed the similar binding of compound TN9 within the active binding pocket and were found to be the most potent anti-epileptic agent. The market expectation for newly developed antiepileptic thiadiazole-based nipecotic acid derivatives is significant, driven by their potential to offer improved therapeutic outcomes and reduced side effects, addressing a critical need in epilepsy treatment. These innovative compounds hold promise for meeting the demand for more effective and safer antiepileptic medications. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03897-1.
Collapse
Affiliation(s)
- Gireesh Kumar Singh
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, Bihar 824236 India
| | - Bindu Kumari
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya, Bihar 824236 India
| | - Nirupam Das
- Department of Pharmaceutical Science, SSMPS, Assam University, Silchar, Assam 788151 India
| | - Kamaruz Zaman
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| | - Pratibha Prasad
- Department of Neurology, All India Institute of Medical Sciences, Deoghar, Jharkhand 814142 India
| | - Ravi Bhushan Singh
- Institute of Pharmacy, Harischandra P.G. College, Bawanbeegha, Azamgarh Road, Varanasi, 221002 India
| |
Collapse
|
4
|
Golub V, Ramakrishnan S, Reddy DS. Isobolographic analysis of adjunct antiseizure activity of the FDA-approved cannabidiol with neurosteroids and benzodiazepines in adult refractory focal onset epilepsy. Exp Neurol 2023; 360:114294. [PMID: 36493860 PMCID: PMC9884179 DOI: 10.1016/j.expneurol.2022.114294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/27/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Epilepsy is a serious neurological disorder associated with recurrent and unpredictable seizures and extensive neuropsychiatric comorbidities. There is no cure for epilepsy, and over one third of epileptic patients have been diagnosed with drug-refractory epilepsy, indicating the critical need for novel antiseizure medications (ASMs). Cannabidiol (CBD) has been shown to decrease seizures in pediatric epilepsies, such as Dravet and Lennox-Gastaut syndromes; however, it has not been rigorously tested for adult seizures or in models of refractory focal epilepsy. Although the exact mechanism is unknown, it is likely to act in a way that is unique to certain GABA-A receptor-modulating drugs, such as neurosteroids and benzodiazepines. In this study, we sought to determine the adjunct antiseizure activity of a clinical CBD product in an adult 6-Hz model of focal refractory epilepsy. CBD was evaluated alone in both a dose-response and time-course manner and in an adjunct combination with two ASMs ganaxolone (neurosteroid) and midazolam (benzodiazepine) against 6-Hz-induced refractory focal onset, generalized seizures. In pharmacological studies, CBD produced dose-dependent protection against seizures (ED50, 53 mg/kg, i.p.) without any side effects. CBD significantly reduced both electrographic activity and behavioral ictal responses with no apparent sex differences. CBD was evaluated in an isobologram design in conjunction with ganaxolone or midazolam at three standard ratios (1:1, 1:3, 3:1). Isobolographic analysis shows the combination regimens of CBD + ganaxolone and CBD + midazolam exerted combination index of 0.313 and 0.164, indicating strong synergism for seizure protection, with little to no toxicity. Together, these results demonstrate the therapeutic potential of CBD monotherapy and as an adjunct therapy for adult focal refractory epilepsy in combination with GABAergic ASMs.
Collapse
Affiliation(s)
- Victoria Golub
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA; Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University, Bryan, TX, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA; Texas A&M Health Institute of Pharmacology and Neurotherapeutics, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
5
|
Metcalf CS, Gagangras S, Bulaj G, White HS. Synergistic effects of the galanin analog 810-2 with the antiseizure medication levetiracetam in rodent seizure models. Epilepsia 2022; 63:3090-3099. [PMID: 36177529 DOI: 10.1111/epi.17420] [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: 07/27/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The use of many antiseizure medications (ASMs) is limited due to pharmacoresistance and dose-limiting side effects, suggesting an unmet need for novel therapeutic approaches. The neuropeptide galanin reduces seizures in several preclinical seizure and epilepsy models, but its clinical utility is limited due to rapid metabolism and poor blood-brain barrier penetration. The lead galanin analog 810-2 is systemically bioavailable and reduces seizures when administered alone. Further development of this analog, with the potential for use as an add-on therapy in patients with epilepsy, requires a better understanding of the use of this analog in combination with approved ASMs. We sought to evaluate 810-2 in combination with commonly used ASMs in rodent models of seizures. METHODS The mouse 6-Hz seizure assay was used to test efficacy of 810-2 in combination with levetiracetam (LEV), valproic acid (VPA), or lacosamide (LCM) using a 1:1 dose ratio in isobolographic studies. Further characterization was performed for the combination of 810-2 and LEV in the mouse corneal kindling and rat 6-Hz assays. RESULTS Whereas the combination of 810-2 with VPA and LCM yielded additive interactions, the combination of 810-2 with LEV demonstrated a synergistic interaction in the mouse 6-Hz assay. Supra-additive effects were also observed in the mouse corneal kindling and rat 6-Hz assays for this combination. SIGNIFICANCE The combination of 810-2 with LEV suggests the potential for this galanin analog to be further developed as an add-on therapy for patients with epilepsy, particularly when coadministered with LEV.
Collapse
Affiliation(s)
- Cameron S Metcalf
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Saurabh Gagangras
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Grzegorz Bulaj
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Maurer-Morelli CV, de Vasconcellos JF, Bruxel EM, Rocha CS, do Canto AM, Tedeschi H, Yasuda CL, Cendes F, Lopes-Cendes I. Gene expression profile suggests different mechanisms underlying sporadic and familial mesial temporal lobe epilepsy. Exp Biol Med (Maywood) 2022; 247:2233-2250. [PMID: 36259630 PMCID: PMC9899983 DOI: 10.1177/15353702221126666] [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] [Indexed: 02/04/2023] Open
Abstract
Most patients with pharmacoresistant mesial temporal lobe epilepsy (MTLE) have hippocampal sclerosis on the postoperative histopathological examination. Although most patients with MTLE do not refer to a family history of the disease, familial forms of MTLE have been reported. We studied surgical specimens from patients with MTLE who had epilepsy surgery for medically intractable seizures. We assessed and compared gene expression profiles of the tissue lesion found in patients with familial MTLE (n = 3) and sporadic MTLE (n = 5). In addition, we used data from control hippocampi obtained from a public database (n = 7). We obtained expression profiles using the Human Genome U133 Plus 2.0 (Affymetrix) microarray platform. Overall, the molecular profile identified in familial MTLE differed from that in sporadic MTLE. In the tissue of patients with familial MTLE, we found an over-representation of the biological pathways related to protein response, mRNA processing, and synaptic plasticity and function. In sporadic MTLE, the gene expression profile suggests that the inflammatory response is highly activated. In addition, we found enrichment of gene sets involved in inflammatory cytokines and mediators and chemokine receptor pathways in both groups. However, in sporadic MTLE, we also found enrichment of epidermal growth factor signaling, prostaglandin synthesis and regulation, and microglia pathogen phagocytosis pathways. Furthermore, based on the gene expression signatures, we identified different potential compounds to treat patients with familial and sporadic MTLE. To our knowledge, this is the first study assessing the mRNA profile in surgical tissue obtained from patients with familial MTLE and comparing it with sporadic MTLE. Our results clearly show that, despite phenotypic similarities, both forms of MTLE present distinct molecular signatures, thus suggesting different underlying molecular mechanisms that may require distinct therapeutic approaches.
Collapse
Affiliation(s)
- Claudia V Maurer-Morelli
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Jaira F de Vasconcellos
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Department of Biology, James Madison
University, Harrisonburg, VA 22807, USA
| | - Estela M Bruxel
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Cristiane S Rocha
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Amanda M do Canto
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Helder Tedeschi
- Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Clarissa L Yasuda
- Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Fernando Cendes
- Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Iscia Lopes-Cendes.
| |
Collapse
|
7
|
Rubal S, Abhishek M, Rupa J, Phulen S, Kumar R, Kaur G, AmitRaj S, Jain A, Prakash A, Alka B, Bikash M. Homotaurine ameriolates the core ASD symptomatology in VPA rats through GABAergic signalling: Role of GAD67. Brain Res Bull 2022; 190:122-133. [PMID: 36113682 DOI: 10.1016/j.brainresbull.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/02/2022]
Abstract
Dysregulated GABAergic signaling is reported in Autism Spectrum disorder (ASD). In the present study, we evaluated a GABA structural mimicker homotaurine (HT) via in-silico docking and investigated the therapeutic efficacy of this drug to ameliorate ASD symptoms in the valproic acid (VPA) rat model of ASD. For the in-vivo study, animals were divided into two groups [Normal control (NC, 0.9% saline; i.p) and disease control (VPA 600mg/kg; i.p)] on gestational day (GD) 12.5. Male pups from VPA-exposed mothers were further divided into five groups (n=6 in each group): disease control (DC, no-further treatment), standard treatment (risperidone (RES) 2.5mg/kg; i.p, consecutively from PND 23-43), HT (10, 25 and 50mg/kg; i.p, consecutively from PND 23-43). In in-silico studies, the binding pattern of homotaurine to GABA-A receptor was found similar to GABA with Tyr205, Glu155, Tyr157, Arg6, and Thr 130 as shared residues. In the in-vivo phase, the early developmental parameters (from PND 7-23) and behavioral parameters (from PND 43-54) were assessed. The offspring of the VPA exposed group exhibited significant (p<0.05) developmental delays, behavioral deficits [decreased sociability and social novelty (three-chamber sociability test), spatial memory (Morris water maze), increased stereotypy (self-grooming)], increased oxidative stress (decreased GSH, SOD, Catalase, and increased MDA), increased pro-inflammatory (IL-1β, 6, TNF-α) and decreased anti-inflammatory (IL-10) cytokines, Purkinje cell loss in the cerebellum and pyknosis in PFC (H/E, Nissil staining) and decreased GAD67 expression in the cerebellum (RT-PCR & immunohistochemistry). Compared to the DC, HT treatment (50mg/kg) was able to ameliorate the aberrant core behavioral deficits, decreased oxidative stress, decreased pro-inflammatory and increased anti-inflammatory cytokine profile with preservation of the Purkinje cell density in the cerebellum, decreased pyknosis in the prefrontal cortex and normalised the expression of GAD67. Thus, HT can be a useful therapeutic agent in ASD and requires further clinical evaluation.
Collapse
Affiliation(s)
- Singla Rubal
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Mishra Abhishek
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Joshi Rupa
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Sarma Phulen
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Rajput Kumar
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Gurjeet Kaur
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Sarma AmitRaj
- Dept. of Neurology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Ashish Jain
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Ajay Prakash
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Bhatia Alka
- Dept. of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| | - Medhi Bikash
- Dept. of Pharmacology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India.
| |
Collapse
|
8
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
9
|
Shaw AD, Chandler HL, Hamandi K, Muthukumaraswamy SD, Hammers A, Singh KD. Tiagabine induced modulation of oscillatory connectivity and activity match PET-derived, canonical GABA-A receptor distributions. Eur Neuropsychopharmacol 2021; 50:34-45. [PMID: 33957336 PMCID: PMC8415204 DOI: 10.1016/j.euroneuro.2021.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 12/04/2022]
Abstract
As the most abundant inhibitory neurotransmitter in the mammalian brain, γ-aminobutyric acid (GABA) plays a crucial role in shaping the frequency and amplitude of oscillations, which suggests a role for GABA in shaping the topography of functional connectivity and activity. This study explored the effects of pharmacologically blocking the reuptake of GABA (increasing local concentrations) using the GABA transporter 1 (GAT1) blocker, tiagabine (15 mg). In a placebo-controlled crossover design, we collected resting magnetoencephalography (MEG) recordings from 15 healthy individuals prior to, and at 1-, 3- and 5- hours post, administration of tiagabine and placebo. We quantified whole brain activity and functional connectivity in discrete frequency bands. Drug-by-session (2 × 4) analysis of variance in connectivity revealed interaction and main effects. Post-hoc permutation testing of each post-drug recording vs. respective pre-drug baseline revealed consistent reductions of a bilateral occipital network spanning theta, alpha and beta frequencies, across 1- 3- and 5- hour recordings following tiagabine only. The same analysis applied to activity revealed significant increases across frontal regions, coupled with reductions in posterior regions, across delta, theta, alpha and beta frequencies. Crucially, the spatial distribution of tiagabine-induced changes overlap with group-averaged maps of the distribution of GABAA receptors, from flumazenil (FMZ-VT) PET, demonstrating a link between GABA availability, GABAA receptor distribution, and low-frequency network oscillations. Our results indicate that the relationship between PET receptor distributions and MEG effects warrants further exploration, since elucidating the nature of this relationship may uncover electrophysiologically-derived maps of oscillatory activity as sensitive, time-resolved, and targeted receptor-mapping tools for pharmacological imaging.
Collapse
Affiliation(s)
- Alexander D Shaw
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, CF24 4HQ, Wales.
| | - Hannah L Chandler
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, CF24 4HQ, Wales
| | - Khalid Hamandi
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, CF24 4HQ, Wales
| | - Suresh D Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Alexander Hammers
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, London SE1 7EH, United States
| | - Krish D Singh
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, CF24 4HQ, Wales
| |
Collapse
|
10
|
Exploring the molecular determinants for subtype-selectivity of 2-amino-1,4,5,6-tetrahydropyrimidine-5-carboxylic acid analogs as betaine/GABA transporter 1 (BGT1) substrate-inhibitors. Sci Rep 2020; 10:12992. [PMID: 32747622 PMCID: PMC7400577 DOI: 10.1038/s41598-020-69908-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
We have previously identified 2-amino-1,4,5,6-tetrahydropyrimidine-5-carboxylic acid (ATPCA) as the most potent substrate-inhibitor of the betaine/GABA transporter 1 (BGT1) (IC50 2.5 µM) reported to date. Herein, we characterize the binding mode of 20 novel analogs and propose the molecular determinants driving BGT1-selectivity. A series of N1-, exocyclic-N-, and C4-substituted analogs was synthesized and pharmacologically characterized in radioligand-based uptake assays at the four human GABA transporters (hGATs) recombinantly expressed in mammalian cells. Overall, the analogs retained subtype-selectivity for hBGT1, though with lower inhibitory activities (mid to high micromolar IC50 values) compared to ATPCA. Further characterization of five of these BGT1-active analogs in a fluorescence-based FMP assay revealed that the compounds are substrates for hBGT1, suggesting they interact with the orthosteric site of the transporter. In silico-guided mutagenesis experiments showed that the non-conserved residues Q299 and E52 in hBGT1 as well as the conformational flexibility of the compounds potentially contribute to the subtype-selectivity of ATPCA and its analogs. Overall, this study provides new insights into the molecular interactions governing the subtype-selectivity of BGT1 substrate-inhibitors. The findings may guide the rational design of BGT1-selective pharmacological tool compounds for future drug discovery.
Collapse
|
11
|
Łątka K, Jończyk J, Bajda M. γ-Aminobutyric acid transporters as relevant biological target: Their function, structure, inhibitors and role in the therapy of different diseases. Int J Biol Macromol 2020; 158:S0141-8130(20)32987-1. [PMID: 32360967 DOI: 10.1016/j.ijbiomac.2020.04.126] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/13/2022]
Abstract
γ-Aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the nervous system. It plays a crucial role in many physiological processes. Upon release from the presynaptic element, it is removed from the synaptic cleft by reuptake due to the action of GABA transporters (GATs). GATs belong to a large SLC6 protein family whose characteristic feature is sodium-dependent relocation of neurotransmitters through the cell membrane. GABA transporters are characterized in many contexts, but their spatial structure is not fully known. They are divided into four types, which differ in occurrence and role. Herein, the special attention was paid to these transporting proteins. This comprehensive review presents the current knowledge about GABA transporters. Their distribution in the body, physiological functions and possible utilization in the therapy of different diseases were fully discussed. The important structural features were described based on published data, including sequence analysis, mutagenesis studies, and comparison with known SLC6 transporters for leucine (LeuT), dopamine (DAT) and serotonin (SERT). Moreover, the most important inhibitors of GABA transporters of various basic scaffolds, diverse selectivity and potency were presented.
Collapse
Affiliation(s)
- Kamil Łątka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland
| | - Jakub Jończyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland
| | - Marek Bajda
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Physicochemical Drug Analysis, 30-688 Cracow, Medyczna 9, Poland.
| |
Collapse
|
12
|
Lie MEK, Kickinger S, Skovgaard-Petersen J, Ecker GF, Clausen RP, Schousboe A, White HS, Wellendorph P. Pharmacological Characterization of a Betaine/GABA Transporter 1 (BGT1) Inhibitor Displaying an Unusual Biphasic Inhibition Profile and Anti-seizure Effects. Neurochem Res 2020; 45:1551-1565. [PMID: 32248400 PMCID: PMC7297817 DOI: 10.1007/s11064-020-03017-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/09/2020] [Accepted: 03/18/2020] [Indexed: 12/01/2022]
Abstract
Focal epileptic seizures can in some patients be managed by inhibiting γ-aminobutyric acid (GABA) uptake via the GABA transporter 1 (GAT1) using tiagabine (Gabitril®). Synergistic anti-seizure effects achieved by inhibition of both GAT1 and the betaine/GABA transporter (BGT1) by tiagabine and EF1502, compared to tiagabine alone, suggest BGT1 as a target in epilepsy. Yet, selective BGT1 inhibitors are needed for validation of this hypothesis. In that search, a series of BGT1 inhibitors typified by (1R,2S)-2-((4,4-bis(3-methylthiophen-2-yl)but-3-en-yl)(methyl)amino)cyclohexanecarboxylic acid (SBV2-114) was developed. A thorough pharmacological characterization of SBV2-114 using a cell-based [3H]GABA uptake assay at heterologously expressed BGT1, revealed an elusive biphasic inhibition profile with two IC50 values (4.7 and 556 μM). The biphasic profile was common for this structural class of compounds, including EF1502, and was confirmed in the MDCK II cell line endogenously expressing BGT1. The possibility of two binding sites for SBV2-114 at BGT1 was assessed by computational docking studies and examined by mutational studies. These investigations confirmed that the conserved residue Q299 in BGT1 is involved in, but not solely responsible for the biphasic inhibition profile of SBV2-114. Animal studies revealed anti-seizure effects of SBV2-114 in two mouse models, supporting a function of BGT1 in epilepsy. However, as SBV2-114 is apparent to be rather non-selective for BGT1, the translational relevance of this observation is unknown. Nevertheless, SBV2-114 constitutes a valuable tool compound to study the molecular mechanism of an emerging biphasic profile of BGT1-mediated GABA transport and the putative involvement of two binding sites for this class of compounds.
Collapse
Affiliation(s)
- Maria E K Lie
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Stefanie Kickinger
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | | | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Rasmus P Clausen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - H Steve White
- Department of Pharmacy, University of Washington, Washington, USA
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Kickinger S, Hellsberg E, Frølund B, Schousboe A, Ecker GF, Wellendorph P. Structural and molecular aspects of betaine-GABA transporter 1 (BGT1) and its relation to brain function. Neuropharmacology 2019; 161:107644. [PMID: 31108110 DOI: 10.1016/j.neuropharm.2019.05.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/14/2019] [Accepted: 05/16/2019] [Indexed: 01/09/2023]
Abstract
ɣ-aminobutyric-acid (GABA) functions as the principal inhibitory neurotransmitter in the central nervous system. Imbalances in GABAergic neurotransmission are involved in the pathophysiology of various neurological diseases such as epilepsy, Alzheimer's disease and stroke. GABA transporters (GATs) facilitate the termination of GABAergic signaling by transporting GABA together with sodium and chloride from the synaptic cleft into presynaptic neurons and surrounding glial cells. Four different GATs have been identified that all belong to the solute carrier 6 (SLC6) transporter family: GAT1-3 (SLC6A1, SLC6A13, SLC6A11) and betaine/GABA transporter 1 (BGT1, SLC6A12). BGT1 has emerged as an interesting target for treating epilepsy due to animal studies that reported anticonvulsant effects for the GAT1/BGT1 selective inhibitor EF1502 and the BGT1 selective inhibitor RPC-425. However, the precise involvement of BGT1 in epilepsy remains elusive because of its controversial expression levels in the brain and the lack of highly selective and potent tool compounds. This review gathers the current structural and functional knowledge on BGT1 with emphasis on brain relevance, discusses all available compounds, and tries to shed light on the molecular determinants driving BGT1 selectivity. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Stefanie Kickinger
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090, Vienna, Austria
| | - Eva Hellsberg
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090, Vienna, Austria
| | - Bente Frølund
- University of Copenhagen, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 2 Universitetsparken, 2100, Copenhagen, Denmark
| | - Arne Schousboe
- University of Copenhagen, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 2 Universitetsparken, 2100, Copenhagen, Denmark
| | - Gerhard F Ecker
- University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090, Vienna, Austria
| | - Petrine Wellendorph
- University of Copenhagen, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 2 Universitetsparken, 2100, Copenhagen, Denmark.
| |
Collapse
|
14
|
Héja L, Simon Á, Szabó Z, Kardos J. Feedback adaptation of synaptic excitability via Glu:Na + symport driven astrocytic GABA and Gln release. Neuropharmacology 2019; 161:107629. [PMID: 31103619 DOI: 10.1016/j.neuropharm.2019.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/30/2019] [Accepted: 05/07/2019] [Indexed: 02/08/2023]
Abstract
Glutamatergic transmission composed of the arriving of action potential at the axon terminal, fast vesicular Glu release, postsynaptic Glu receptor activation, astrocytic Glu clearance and Glu→Gln shuttle is an abundantly investigated phenomenon. Despite its essential role, however, much less is known about the consequences of the mechanistic connotations of Glu:Na+ symport. Due to the coupled Na+ transport, Glu uptake results in significantly elevated intracellular astrocytic [Na+] that markedly alters the driving force of other Na+-coupled astrocytic transporters. The resulting GABA and Gln release by reverse transport through the respective GAT-3 and SNAT3 transporters help to re-establish the physiological Na+ homeostasis without ATP dissipation and consequently leads to enhanced tonic inhibition and replenishment of axonal glutamate pool. Here, we place this emerging astrocytic adjustment of synaptic excitability into the centre of future perspectives. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Zsolt Szabó
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary.
| |
Collapse
|
15
|
Zafar S, Jabeen I. GRID-independent molecular descriptor analysis and molecular docking studies to mimic the binding hypothesis of γ-aminobutyric acid transporter 1 (GAT1) inhibitors. PeerJ 2019; 7:e6283. [PMID: 30723616 PMCID: PMC6360079 DOI: 10.7717/peerj.6283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
Abstract
Background The γ-aminobutyric acid (GABA) transporter GAT1 is involved in GABA transport across the biological membrane in and out of the synaptic cleft. The efficiency of this Na+ coupled GABA transport is regulated by an electrochemical gradient, which is directed inward under normal conditions. However, in certain pathophysiological situations, including strong depolarization or an imbalance in ion homeostasis, the GABA influx into the cytoplasm is increased by re-uptake transport mechanism. This mechanism may lead to extra removal of extracellular GABA which results in numerous neurological disorders such as epilepsy. Thus, small molecule inhibitors of GABA re-uptake may enhance GABA activity at the synaptic clefts. Methods In the present study, various GRID-independent molecular descriptor (GRIND) models have been developed to shed light on the 3D structural features of human GAT1 (hGAT1) inhibitors using nipecotic acid and N-diarylalkenyl piperidine analogs. Further, a binding hypothesis has been developed for the selected GAT1 antagonists by molecular docking inside the binding cavity of hGAT1 homology model. Results Our results indicate that two hydrogen bond acceptors, one hydrogen bond donor and one hydrophobic region at certain distances from each other play an important role in achieving high inhibitory potency against hGAT1. Our docking results elucidate the importance of the COOH group in hGAT1 antagonists by considering substitution of the COOH group with an isoxazol ring in compound 37, which subsequently leads to a three order of magnitude decrease in biological activity of 37 (IC50 = 38 µM) as compared to compound 1 (IC50 = 0.040 µM). Discussion Our docking results are strengthened by the structure activity relationship of the data series as well as by GRIND models, thus providing a significant structural basis for understanding the binding of antagonists, which may be useful for guiding the design of hGAT1 inhibitors.
Collapse
Affiliation(s)
- Sadia Zafar
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, Federal, Pakistan
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, Federal, Pakistan
| |
Collapse
|
16
|
Nowaczyk A, Fijałkowski Ł, Kowalska M, Podkowa A, Sałat K. Studies on the Activity of Selected Highly Lipophilic Compounds toward hGAT1 Inhibition. Part II. ACS Chem Neurosci 2019; 10:337-347. [PMID: 30222312 DOI: 10.1021/acschemneuro.8b00282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this paper, we describe the latest results involving molecular modeling and pharmacodynamic studies of the selected highly lipophilic compounds acting by human GABA transporter 1 (hGAT1) inhibition. The chemical interaction of 17 GABA analogues with a model of hGAT1 is described using the molecular docking method. The biological role of GAT1 is related to the regulation of GABA level in the central nervous system and GAT1 inhibition plays an important role in the control of seizure threshold. To confirm that GAT1 can be also a molecular target for drugs used to treat other neurological and psychiatric diseases (e.g., pain and anxiety), in the in vivo part of this study, potential antinociceptive and anxiolytic-like properties of tiagabine, a selective GAT1 inhibitor, are described.
Collapse
Affiliation(s)
- Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Łukasz Fijałkowski
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Magdalena Kowalska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Adrian Podkowa
- Chair of Inorganic and Analytical Chemistry, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Kraków, Poland
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland
| |
Collapse
|
17
|
Zafar S, Nguyen ME, Muthyala R, Jabeen I, Sham YY. Modeling and Simulation of hGAT1: A Mechanistic Investigation of the GABA Transport Process. Comput Struct Biotechnol J 2018; 17:61-69. [PMID: 30619541 PMCID: PMC6312766 DOI: 10.1016/j.csbj.2018.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/06/2018] [Accepted: 12/09/2018] [Indexed: 01/30/2023] Open
Abstract
Human γ-Aminobutyric acid transporter 1 (hGAT1) is a Na+/Cl- dependent co-transporter that plays a key role in the inhibitory neurotransmission of GABA in the brain. Due to the lack of structural data, the exact co-transport mechanism of GABA reuptake by hGAT1 remains unclear. To examine the roles of the co-transport ions and the nature of their interactions with GABA, homology modeling and molecular dynamics simulations of the hGAT1 in the open-to-out conformation were carried out. Our study focused on the sequential preloading of Na+ and Cl- ions, followed by GABA binding. Our simulations showed pre-loading of ions maintains the transport ready state of hGAT1 in the open-to-out conformation essential for GABA binding. Of the four putative preloaded states, GABA binding to the fully loaded state is most favored. Binding of Na+ ion to the Na1 site helps to maintain the open-to-out conformation for GABA binding as compared to the Na2 site. GABA binding to the mono-sodium or the di-sodium loaded states leads to destabilization of Na+ ions within their binding sites. The two most prominent interactions required for GABA binding include interaction between carboxylate group of GABA with the bound Na+ ion in Na1 binding site and the hydroxyl group of Y140. Overall our results support the fully loaded state as the predominate state for GABA binding. Our study further illustrates that Na+ ion within the Na1 site is crucial for GABA recognition. Therefore, a revised mechanism is proposed for the initial step of hGAT1 translocation cycle.
Collapse
Affiliation(s)
- Sadia Zafar
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Megin E. Nguyen
- Bioinformatics and Computational Biology Program, University of Minnesota, United States
| | - Ramaiah Muthyala
- Department of Experimental and Clinical Pharmacology & Center for Orphan Drug Research, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Yuk Y. Sham
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, United States
- Bioinformatics and Computational Biology Program, University of Minnesota, United States
| |
Collapse
|
18
|
Lin X, Duan X, Jacobs C, Ullmann J, Chan CY, Chen S, Cheng SH, Zhao WN, Poduri A, Wang X, Haggarty SJ, Shi P. High-throughput brain activity mapping and machine learning as a foundation for systems neuropharmacology. Nat Commun 2018; 9:5142. [PMID: 30510233 PMCID: PMC6277389 DOI: 10.1038/s41467-018-07289-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022] Open
Abstract
Technologies for mapping the spatial and temporal patterns of neural activity have advanced our understanding of brain function in both health and disease. An important application of these technologies is the discovery of next-generation neurotherapeutics for neurological and psychiatric disorders. Here, we describe an in vivo drug screening strategy that combines high-throughput technology to generate large-scale brain activity maps (BAMs) with machine learning for predictive analysis. This platform enables evaluation of compounds’ mechanisms of action and potential therapeutic uses based on information-rich BAMs derived from drug-treated zebrafish larvae. From a screen of clinically used drugs, we found intrinsically coherent drug clusters that are associated with known therapeutic categories. Using BAM-based clusters as a functional classifier, we identify anti-seizure-like drug leads from non-clinical compounds and validate their therapeutic effects in the pentylenetetrazole zebrafish seizure model. Collectively, this study provides a framework to advance the field of systems neuropharmacology. A major goal in neuropharmacology is to develop new tools to effectively test the therapeutic potential of pharmacological agents to treat neurological and psychiatric conditions. Here, authors present an in vivo drug screening system that generates large-scale brain activity maps to be used with machine learning to predict the therapeutic potential of clinically relevant drug leads.
Collapse
Affiliation(s)
- Xudong Lin
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China
| | - Xin Duan
- Department of Biomedical Science, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China
| | - Claire Jacobs
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jeremy Ullmann
- Epilepsy Genetics Program and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Chung-Yuen Chan
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China
| | - Siya Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China
| | - Shuk-Han Cheng
- Department of Biomedical Science, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston, MA, 02114, USA
| | - Annapurna Poduri
- Epilepsy Genetics Program and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Xin Wang
- Department of Biomedical Science, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China. .,Shenzhen Research Institute, City University of Hong Kong, 518057, Shenzhen, China.
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston, MA, 02114, USA.
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Kowloon, Hong Kong SAR, China. .,Shenzhen Research Institute, City University of Hong Kong, 518057, Shenzhen, China.
| |
Collapse
|
19
|
Tóth K, Höfner G, Wanner KT. Synthesis and biological evaluation of novel N-substituted nipecotic acid derivatives with a trans-alkene spacer as potent GABA uptake inhibitors. Bioorg Med Chem 2018; 26:5944-5961. [DOI: 10.1016/j.bmc.2018.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 12/13/2022]
|
20
|
Zafar S, Jabeen I. Structure, Function, and Modulation of γ-Aminobutyric Acid Transporter 1 (GAT1) in Neurological Disorders: A Pharmacoinformatic Prospective. Front Chem 2018; 6:397. [PMID: 30255012 PMCID: PMC6141625 DOI: 10.3389/fchem.2018.00397] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/20/2018] [Indexed: 02/03/2023] Open
Abstract
γ-Aminobutyric acid (GABA) Transporters (GATs) belong to sodium and chloride dependent-transporter family and are widely expressed throughout the brain. Notably, GAT1 is accountable for sustaining 75% of the synaptic GABA concentration and entails its transport to the GABAA receptors to initiate the receptor-mediated inhibition of post-synaptic neurons. Imbalance in ion homeostasis has been associated with several neurological disorders related to the GABAergic system. However, inhibition of the GABA uptake by these transporters has been accepted as an effective approach to enhance GABAergic inhibitory neurotransmission in the treatment of seizures in epileptic and other neurological disorders. Here, we reviewed computational methodologies including molecular modeling, docking, and molecular dynamic simulations studies to underscore the structure and function of GAT1 in the GABAergic system. Additionally, various SAR and QSAR methodologies have been reviewed to probe the 3D structural features of inhibitors required to modulate GATs activity. Overall, present review provides an overview of crucial role of GAT1 in GABAergic system and its modulation to evade neurological disorders.
Collapse
Affiliation(s)
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
21
|
Al-Khawaja A, Haugaard AS, Marek A, Löffler R, Thiesen L, Santiveri M, Damgaard M, Bundgaard C, Frølund B, Wellendorph P. Pharmacological Characterization of [ 3H]ATPCA as a Substrate for Studying the Functional Role of the Betaine/GABA Transporter 1 and the Creatine Transporter. ACS Chem Neurosci 2018; 9:545-554. [PMID: 29131576 DOI: 10.1021/acschemneuro.7b00351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The betaine/γ-aminobutyric acid (GABA) transporter 1 (BGT1) is one of the four GABA transporters (GATs) involved in the termination of GABAergic neurotransmission. Although suggested to be implicated in seizure management, the exact functional importance of BGT1 in the brain is still elusive. This is partly owing to the lack of potent and selective pharmacological tool compounds that can be used to probe its function. We previously reported the identification of 2-amino-1,4,5,6-tetrahydropyrimidine-5-carboxylic acid (ATPCA), a selective substrate for BGT1 over GAT1/GAT3, but also an agonist for GABAA receptors. With the aim of providing new functional insight into BGT1, we here present the synthesis and pharmacological characterization of the tritiated analogue, [3H]ATPCA. Using traditional uptake assays at recombinant transporters expressed in cell lines, [3H]ATPCA displayed a striking selectivity for BGT1 among the four GATs ( Km and Vmax values of 21 μM and 3.6 nmol ATPCA/(min × mg protein), respectively), but was also found to be a substrate for the creatine transporter (CreaT). In experiments with mouse cortical cell cultures, we observed a Na+-dependent [3H]ATPCA uptake in neurons, but not in astrocytes. The neuronal uptake could be inhibited by GABA, ATPCA, and a noncompetitive BGT1-selective inhibitor, indicating functional BGT1 in neurons. In conclusion, we report [3H]ATPCA as a novel radioactive substrate for both BGT1 and CreaT. The dual activity of the radioligand makes it most suitable for use in recombinant studies.
Collapse
Affiliation(s)
- Anas Al-Khawaja
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anne S. Haugaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Ales Marek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 542/2, 16610 Prague 6, Czech Republic
| | - Rebekka Löffler
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Louise Thiesen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mònica Santiveri
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Maria Damgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
22
|
Metcalf CS, Klein BD, Smith MD, Ceusters M, Lavreysen H, Pype S, Van Osselaer N, Twyman R, White HS. Potent and selective pharmacodynamic synergy between the metabotropic glutamate receptor subtype 2-positive allosteric modulator JNJ-46356479 and levetiracetam in the mouse 6-Hz (44-mA) model. Epilepsia 2018; 59:724-735. [PMID: 29360159 DOI: 10.1111/epi.14005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE We previously demonstrated that positive allosteric modulators (PAMs) of metabotropic glutamate subtype 2 (mGlu2 ) receptors have potential synergistic interactions with the antiseizure drug levetiracetam (LEV). The present study utilizes isobolographic analysis to evaluate the combined administration of JNJ-46356479, a selective and potent mGlu2 PAM, with LEV as well as sodium valproate (VPA) and lamotrigine (LTG). METHODS The anticonvulsant efficacy of JNJ-46356479 was evaluated in the 6-Hz model of psychomotor seizures in mice. JNJ-46356479 was administered in combination with LEV using 3 fixed dose-ratio treatment groups in the mouse 6-Hz (44-mA) seizure test. The combination of JNJ-46356479 with LEV was also evaluated in the mouse corneal kindling model. The potential interactions of JNJ-46356479 with the antiseizure drugs VPA and LTG were also evaluated using fixed dose-ratio combinations. Plasma levels were obtained for analysis of potential pharmacokinetic interactions for each combination studied in the mouse 6-Hz model. RESULTS JNJ-46356479 was active in the 6-Hz model at both 32-mA and 44-mA stimulus intensities (median effective dose = 2.8 and 10.2 mg/kg, respectively). Using 1:1, 1:3, and 3:1 fixed dose-ratio combinations (LEV:JNJ-46356479), coadministration was significantly more potent than predicted for additive effects, and plasma levels suggest this synergism was not due to pharmacokinetic interactions. Studies in kindled mice further demonstrate the positive pharmacodynamic interaction of LEV with JNJ-46356479. Using 1:1 dose-ratio combinations of JNJ-46356479 with either VPA or LTG, there were no significant differences observed for coadministration. SIGNIFICANCE These studies demonstrate a synergistic interaction of JNJ-46356479 with LEV, whereas no such effect occurred for JNJ-46356479 with either VPA or LTG. The synergy seems therefore to be specific to LEV, and the combination LEV/mGlu2 PAM has the potential to result in a rational polypharmacy approach to treat patients with refractory epilepsy, once it has been confirmed in clinical studies.
Collapse
Affiliation(s)
- Cameron S Metcalf
- NeuroAdjuvants, Salt Lake City, UT, USA.,Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Brian D Klein
- NeuroAdjuvants, Salt Lake City, UT, USA.,Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Misty D Smith
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | | | | | | | - Nancy Van Osselaer
- Janssen Research and Development, Beerse, Belgium.,UCB, Brussels, Belgium
| | - Roy Twyman
- Janssen Research and Development, Titusville, FL, USA
| | - H Steve White
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA.,Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Lie ME, Gowing EK, Clausen RP, Wellendorph P, Clarkson AN. Inhibition of GABA transporters fails to afford significant protection following focal cerebral ischemia. J Cereb Blood Flow Metab 2018; 38:166-173. [PMID: 29148909 PMCID: PMC5757447 DOI: 10.1177/0271678x17743669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Brain ischemia triggers excitotoxicity and cell death, yet no neuroprotective drugs have made it to the clinic. While enhancing GABAergic signaling to counterbalance excitotoxicity has shown promise in animal models, clinical studies have failed. Blockade of GABA transporters (GATs) offers an indirect approach to increase GABA inhibition to lower the excitation threshold of neurons. Among the GATs, GAT1 is known to promote neuroprotection, while the protective role of the extrasynaptic transporters GAT3 and BGT1 is elusive. A focal lesion was induced in the motor cortex in two to four-month-old C57BL/6 J male mice by photothrombosis. The GAT1 inhibitor, tiagabine (1 and 10 mg/kg), the GAT2/3 inhibitor, ( S)-SNAP-5114 (5 and 30 mg/kg) and the GAT1/BGT1 inhibitor, EF-1502 (1 and 10 mg/kg) were given i.p. 1 and 6 h post-stroke to assess their impact on infarct volume and motor performance seven days post-stroke. One mg/kg tiagabine improved motor performance, while 10 mg/kg tiagabine, ( S)-SNAP-5114 and EF-1502 had no effect. None of the compounds affected infarct volume. Interestingly, treatment with tiagabine induced seizures and ( S)-SNAP-5114 led to increased mortality. Although we show that tiagabine can promote protection, our findings indicate that caution should be had when using GAT1 and GAT3 inhibitors for conditions of brain ischemia.
Collapse
Affiliation(s)
- Maria Ek Lie
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Emma K Gowing
- 2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Rasmus P Clausen
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- 1 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Andrew N Clarkson
- 2 Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand.,3 Faculty of Pharmacy, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
An automated compound screening for anti-aging effects on the function of C. elegans sensory neurons. Sci Rep 2017; 7:9403. [PMID: 28839194 PMCID: PMC5570957 DOI: 10.1038/s41598-017-09651-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/27/2017] [Indexed: 12/11/2022] Open
Abstract
Discovery of molecular targets or compounds that alter neuronal function can lead to therapeutic advances that ameliorate age-related neurodegenerative pathologies. Currently, there is a lack of in vivo screening technologies for the discovery of compounds that affect the age-dependent neuronal physiology. Here, we present a high-throughput, microfluidic-based assay for automated manipulation and on-chip monitoring and analysis of stimulus-evoked calcium responses of intact C. elegans at various life stages. First, we successfully applied our technology to quantify the effects of aging and age-related genetic and chemical factors in the calcium transients of the ASH sensory neuron. We then performed a large-scale screen of a library of 107 FDA-approved compounds to identify hits that prevented the age-dependent functional deterioration of ASH. The robust performance of our assay makes it a valuable tool for future high-throughput applications based on in vivo functional imaging.
Collapse
|
25
|
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
|
26
|
Simonsen C, Boddum K, von Schoubye NL, Kloppenburg A, Sønderskov K, Hansen SL, Kristiansen U. Anticonvulsive evaluation of THIP in the murine pentylenetetrazole kindling model: lack of anticonvulsive effect of THIP despite functional δ-subunit-containing GABA A receptors in dentate gyrus granule cells. Pharmacol Res Perspect 2017; 5. [PMID: 28805971 PMCID: PMC5684853 DOI: 10.1002/prp2.322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/29/2022] Open
Abstract
THIP (4,5,6,7‐tetrahydroisoxazolo[5,4‐c]pyridin‐3‐ol) is a GABAA receptor agonist with varying potencies and efficacies at γ‐subunit‐containing receptors. More importantly, THIP acts as a selective superagonist at δ‐subunit‐containing receptors (δ‐GABAARs) at clinically relevant concentrations. Evaluation of THIP as a potential anticonvulsant has given contradictory results in different animal models and for this reason, we reevaluated the anticonvulsive properties of THIP in the murine pentylenetetrazole (PTZ) kindling model. As loss of δ‐GABAAR in the dentate gyrus has been associated with several animal models of epilepsy, we first investigated the presence of functional δ‐GABAA receptors. Both immunohistochemistry and Western blot data demonstrated that δ‐GABAAR expression is not only present in the dentate gyrus, but also the expression level was enhanced in the early phase after PTZ kindling. Whole‐cell patch‐clamp studies in acute hippocampal brain slices revealed that THIP was indeed able to induce a tonic inhibition in dentate gyrus granule cells. However, THIP induced a tonic current of similar magnitude in the PTZ‐kindled mice compared to saline‐treated animals despite the observed upregulation of δ‐GABAARs. Even in the demonstrated presence of functional δ‐GABAARs, THIP (0.5–4 mg/kg) showed no anticonvulsive effect in the PTZ kindling model using a comprehensive in vivo evaluation of the anticonvulsive properties.
Collapse
Affiliation(s)
- Charlotte Simonsen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Boddum
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadia L von Schoubye
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alissa Kloppenburg
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Sønderskov
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne L Hansen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Uffe Kristiansen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Wellendorph P, Jacobsen J, Skovgaard-Petersen J, Jurik A, Vogensen SB, Ecker G, Schousboe A, Krogsgaard-Larsen P, Clausen RP. γ-Aminobutyric Acid and Glycine Neurotransmitter Transporters. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527679430.ch4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Petrine Wellendorph
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Julie Jacobsen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Jonas Skovgaard-Petersen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Andreas Jurik
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14, A-1090 Vienna Austria
| | - Stine B. Vogensen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Gerhard Ecker
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14, A-1090 Vienna Austria
| | - Arne Schousboe
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Povl Krogsgaard-Larsen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| | - Rasmus P. Clausen
- University of Copenhagen; Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology; Universitetsparken 2, DK-2100 Copenhagen Denmark
| |
Collapse
|
28
|
Delineation of the Role of Astroglial GABA Transporters in Seizure Control. Neurochem Res 2017; 42:2019-2023. [PMID: 28190226 DOI: 10.1007/s11064-017-2188-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 02/06/2023]
Abstract
Studies of GABA transport in neurons and astrocytes have provided evidence that termination of GABA as neurotransmitter is brought about primarily by active transport into the presynaptic, GABAergic nerve endings. There is, however, a considerable transport capacity in the astrocytes surrounding the synaptic terminals, a transport which may limit the availability of transmitter GABA leading to a higher probability of seizure activity governed by the balance of excitatory and inhibitory neurotransmission. Based on this it was hypothesized that selective inhibition of astrocytic GABA transport might prevent such seizure activity. A series of GABA analogs of restricted conformation were synthesized and in a number of collaborative investigations between Prof. Steve White at the University of Utah and medicinal chemists and pharmacologists at the School of Pharmacy and the University of Copenhagen, Denmark, GABA analogs with exactly this pharmacological property were identified. The most important analogs identified were N-methyl-exo-THPO (N-methyl-3-hydroxy-4-amino-4,5,6,7-tetrahydro-1,2-benzisoxazole) and its lipophilic analog EF-1502 ((RS)-4-[N-[1,1-bis(3-methyl-2-thienyl)but-1-en-4-yl]-N-methylamino]-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol) both of which turned out to be potent anticonvulsants in animal models of epilepsy.
Collapse
|
29
|
Eskandari S, Willford SL, Anderson CM. Revised Ion/Substrate Coupling Stoichiometry of GABA Transporters. ADVANCES IN NEUROBIOLOGY 2017; 16:85-116. [PMID: 28828607 DOI: 10.1007/978-3-319-55769-4_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose of this review is to highlight recent evidence in support of a 3 Na+: 1 Cl-: 1 GABA coupling stoichiometry for plasma membrane GABA transporters (SLC6A1 , SLC6A11 , SLC6A12 , SLC6A13 ) and how the revised stoichiometry impacts our understanding of the contribution of GABA transporters to GABA homeostasis in synaptic and extrasynaptic regions in the brain under physiological and pathophysiological states. Recently, our laboratory probed the GABA transporter stoichiometry by analyzing the results of six independent measurements, which included the shifts in the thermodynamic transporter reversal potential caused by changes in the extracellular Na+, Cl-, and GABA concentrations, as well as the ratio of charge flux to substrate flux for Na+, Cl-, and GABA under voltage-clamp conditions. The shifts in the transporter reversal potential for a tenfold change in the external concentration of Na+, Cl-, and GABA were 84 ± 4, 30 ± 1, and 29 ± 1 mV, respectively. Charge flux to substrate flux ratios were 0.7 ± 0.1 charges/Na+, 2.0 ± 0.2 charges/Cl-, and 2.1 ± 0.1 charges/GABA. We then compared these experimental results with the predictions of 150 different transporter stoichiometry models, which included 1-5 Na+, 0-5 Cl-, and 1-5 GABA per transport cycle. Only the 3 Na+: 1 Cl-: 1 GABA stoichiometry model correctly predicts the results of all six experimental measurements. Using the revised 3 Na+: 1 Cl-: 1 GABA stoichiometry, we propose that the GABA transporters mediate GABA uptake under most physiological conditions. Transporter-mediated GABA release likely takes place under pathophysiological or extreme physiological conditions.
Collapse
Affiliation(s)
- Sepehr Eskandari
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA.
| | - Samantha L Willford
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| | - Cynthia M Anderson
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| |
Collapse
|
30
|
Astrocytic GABA Transporters: Pharmacological Properties and Targets for Antiepileptic Drugs. ADVANCES IN NEUROBIOLOGY 2017; 16:283-296. [PMID: 28828616 DOI: 10.1007/978-3-319-55769-4_14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inactivation of GABA-mediated neurotransmission is achieved by high-affinity transporters located at both GABAergic neurons and the surrounding astrocytes. Early studies of the pharmacological properties of neuronal and glial GABA transporters suggested that different types of transporters might be expressed in the two cell types, and such a scenario was confirmed by the cloning of four distinctly different GABA transporters from a number of different species. These GABA-transport entities have been extensively characterized using a large number of GABA analogues of restricted conformation, and several of these compounds have been shown to exhibit pronounced anticonvulsant activity in a variety of animal seizure models. As proof of concept of the validity of this drug development approach, one GABA-transport inhibitor, tiagabine, has been developed as a clinically active antiepileptic drug. This review provides a detailed account of efforts to design new subtype-selective GABA-transport inhibitors aiming at identifying novel antiepileptic drug candidates.
Collapse
|
31
|
Glial GABA Transporters as Modulators of Inhibitory Signalling in Epilepsy and Stroke. ADVANCES IN NEUROBIOLOGY 2017; 16:137-167. [PMID: 28828609 DOI: 10.1007/978-3-319-55769-4_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Imbalances in GABA-mediated tonic inhibition are involved in several pathophysiological conditions. A classical way of controlling tonic inhibition is through pharmacological intervention with extrasynaptic GABAA receptors that sense ambient GABA and mediate a persistent GABAergic conductance. An increase in tonic inhibition may, however, also be obtained indirectly by inhibiting glial GABA transporters (GATs). These are sodium-coupled membrane transport proteins that normally act to terminate GABA neurotransmitter action by taking up GABA into surrounding astrocytes. The aim of the review is to provide an overview of glial GATs in regulating tonic inhibition, especially in epilepsy and stroke. This entails a comprehensive summary of changes known to occur in GAT expression levels and signalling following epileptic and ischemic insults. Further, we discuss the accumulating pharmacological evidence for targeting GATs in these diseases.
Collapse
|
32
|
Wang TC, Ngampramuan S, Kotchabhakdi N. Tiagabine treatment in kainic acid induced cerebellar lesion of dystonia rat model. EXCLI JOURNAL 2016; 15:716-729. [PMID: 28337103 PMCID: PMC5318686 DOI: 10.17179/excli2016-482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/28/2016] [Indexed: 01/13/2023]
Abstract
Dystonia is a neurological disorder characterized by excessive involuntary muscle contractions that lead to twisting movements. The exaggerated movements have been studied and have implicated basal ganglia as the point of origin. In more recent studies, the cerebellum has also been identified as the possible target of dystonia, in the search for alternative treatments. Tiagabine is a selective GABA transporter inhibitor, which blocks the reuptake and recycling of GABA. The study of GABAergic drugs as an alternative treatment for cerebellar induced dystonia has not been reported. In our study, tiagabine was i.p. injected into kainic acid induced, cerebellar dystonic adult rats, and the effects were compared with non-tiagabine injected and sham-operated groups. Beam walking apparatus, telemetric electromyography (EMG) recording, and histological verification were performed to confirm dystonic symptoms in the rats on post-surgery treatment. Involuntary dystonic spasm was observed with repetitive rigidity, and twisting movements in the rats were also confirmed by a high score on the dystonic scoring and a high amplitude on the EMG data. The rats with tiagabine treatment were scored based on motor amelioration assessed via beam walking. The result of this study suggests and confirms that low dose of kainic acid microinjection is sufficient to induce dystonia from the cerebellar vermis. In addition, from the results of the EMG recording and the behavioral assessment through beam walking, tiagabine is demonstrated as being effective in reducing dystonic spasm and may be a possible alternative therapeutic drug in the treatment of dystonia.
Collapse
Affiliation(s)
- Tsui-Chin Wang
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya campus, Nakhon Pathom 73170, Thailand
| | - Sukonthar Ngampramuan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya campus, Nakhon Pathom 73170, Thailand
| | - Naiphinich Kotchabhakdi
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya campus, Nakhon Pathom 73170, Thailand
| |
Collapse
|
33
|
Singh D, Devi N, Kumar V, Malakar CC, Mehra S, Rawal RK, Kaith BS, Singh V. Metal-free 1,3-dipolar cycloaddition approach towards the regioselective synthesis of β-carboline and isoxazole based molecular hybrids. RSC Adv 2016. [DOI: 10.1039/c6ra15875g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nature has nourished β-carboline and isoxazole derivatives as privileged scaffolds and consequently they are ubiquitously found in alkaloids isolated from various sources.
Collapse
Affiliation(s)
- Dharmender Singh
- Department of Chemistry
- Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar
- India
| | - Nisha Devi
- Department of Chemistry
- Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar
- India
| | - Vipin Kumar
- Department of Chemistry
- Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar
- India
| | - Chandi C. Malakar
- Department of Chemistry
- National Institute of Technology (NIT) Manipur
- Imphal 795004
- India
| | - Saloni Mehra
- Amity Institute of Applied Sciences
- Amity University
- Noida
- India
| | - Ravindra K. Rawal
- Department of Pharmaceutical Chemistry
- Indo-Soviet Friendship College of Pharmacy
- Moga 142001
- India
| | - B. S. Kaith
- Department of Chemistry
- Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar
- India
| | - Virender Singh
- Department of Chemistry
- Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar
- India
| |
Collapse
|
34
|
Damgaard M, Al-Khawaja A, Vogensen SB, Jurik A, Sijm M, Lie MEK, Bæk MI, Rosenthal E, Jensen AA, Ecker GF, Frølund B, Wellendorph P, Clausen RP. Identification of the First Highly Subtype-Selective Inhibitor of Human GABA Transporter GAT3. ACS Chem Neurosci 2015; 6:1591-9. [PMID: 26154082 DOI: 10.1021/acschemneuro.5b00150] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Screening a library of small-molecule compounds using a cell line expressing human GABA transporter 3 (hGAT3) in a [(3)H]GABA uptake assay identified isatin derivatives as a new class of hGAT3 inhibitors. A subsequent structure-activity relationship (SAR) study led to the identification of hGAT3-selective inhibitors (i.e., compounds 20 and 34) that were superior to the reference hGAT3 inhibitor, (S)-SNAP-5114, in terms of potency (low micromolar IC50 values) and selectivity (>30-fold selective for hGAT3 over hGAT1/hGAT2/hBGT1). Further pharmacological characterization of compound 20 (5-(thiophen-2-yl)indoline-2,3-dione) revealed a noncompetitive mode of inhibition at hGAT3. This suggests that this compound class, which has no structural resemblance to GABA, has a binding site different from the substrate, GABA. This was supported by a molecular modeling study that suggested a unique binding site that matched the observed selectivity, inhibition kinetics, and SAR of the compound series. These compounds are the most potent GAT3 inhibitors reported to date that provide selectivity for GAT3 over other GABA transporter subtypes.
Collapse
Affiliation(s)
| | | | | | - Andreas Jurik
- Department
of Pharmaceutical Chemistry, University of Vienna, Althanstrasse
14, A-1090 Vienna, Austria
| | | | | | | | | | | | - Gerhard F. Ecker
- Department
of Pharmaceutical Chemistry, University of Vienna, Althanstrasse
14, A-1090 Vienna, Austria
| | | | | | | |
Collapse
|
35
|
Vogensen SB, Jørgensen L, Madsen KK, Jurik A, Borkar N, Rosatelli E, Nielsen B, Ecker GF, Schousboe A, Clausen RP. Structure activity relationship of selective GABA uptake inhibitors. Bioorg Med Chem 2015; 23:2480-8. [DOI: 10.1016/j.bmc.2015.03.060] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/12/2015] [Accepted: 03/19/2015] [Indexed: 10/23/2022]
|
36
|
Synthesis, biological evaluation and molecular docking studies of thiazole-based pyrrolidinones and isoindolinediones as anticonvulsant agents. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1371-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
37
|
Madsen KK, Hansen GH, Danielsen EM, Schousboe A. The subcellular localization of GABA transporters and its implication for seizure management. Neurochem Res 2014; 40:410-9. [PMID: 25519681 DOI: 10.1007/s11064-014-1494-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/27/2014] [Accepted: 12/01/2014] [Indexed: 10/24/2022]
Abstract
The ability to modulate the synaptic GABA levels has been demonstrated by using the clinically effective and selective GAT1 inhibitor tiagabine [(R)-N-[4,4-bis(3-methyl-2-thienyl)-3-butenyl]nipecotic acid]. N-[4,4-bis(3-methyl-2-thienyl)-3-butenyl]-3-hydroxy-4-(methylamino)-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol (EF1502) which not only inhibits GAT1 like tiagabine but also BGT1 has been shown to modulate extrasynaptic GABA levels. The simultaneous inhibition of synaptic and extrasynaptic GABA transporters using tiagabine and EF1502, respectively has been demonstrated to exert a synergistic anticonvulsant effect in several seizure models in mice. The pharmacological profile of these and similar compounds has been thoroughly investigated in in vitro systems, comparing the GAT subtype selectivity with the ability to inhibit GABA uptake in primary cultures of neurons and astrocytes. However, an exact explanation has not yet been found. In the present study, the ability of GATs to form homo and/or heterodimers was investigated as well as to which membrane micro environment the GATs reside. To investigate dimerization of GATs, fusion proteins of GATs tagged with either yellow fluorescent protein or cerulean fluorescent protein were made and fluorescence resonance energy transfer (FRET) was measured. It was found that GATs form both homo- and hetero-dimers in N2A and HEK-293 cells. Microdomain localization of GATs as investigated by detergent resistant membrane fractions after treatment of tissue with Brij-98 or Triton X-100 revealed that BGT1 and GAT1 mostly localize to non-membrane rafts independent of the detergent used. However, GAT3 localizes to membrane rafts when using Brij-98. Taken together, these results suggest that the observed hetero dimerization of GATs in the FRET study is unlikely to have functional implications since the GATs are located to very different cellular compartments and cell types.
Collapse
Affiliation(s)
- Karsten K Madsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen Ø, Denmark,
| | | | | | | |
Collapse
|
38
|
Pharmacological identification of a guanidine-containing β-alanine analogue with low micromolar potency and selectivity for the betaine/GABA transporter 1 (BGT1). Neurochem Res 2014; 39:1988-96. [PMID: 24852577 DOI: 10.1007/s11064-014-1336-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
Abstract
The γ-aminobutyric acid (GABA) transporters (GATs) are key membrane transporter proteins involved in the termination of GABAergic signaling at synapses in the mammalian brain and proposed drug targets in neurological disorders such as epilepsy. To date, four different GAT subtypes have been identified: GAT1, GAT2, GAT3 and the betaine/GABA transporter 1 (BGT1). Owing to the lack of potent and subtype selective inhibitors of the non-GAT1 GABA transporters, the physiological role and therapeutic potential of these transporters remain to be fully understood. Based on bioisosteric replacement of the amino group in β-alanine or GABA, a series of compounds was generated, and their pharmacological activity assessed at human GAT subtypes. Using a cell-based [(3)H]GABA uptake assay, several selective inhibitors at human BGT1 were identified. The guanidine-containing compound 9 (2-amino-1,4,5,6-tetrahydropyrimidine-5-carboxylic acid hydrochloride) displayed more than 250 times greater potency than the parent compound β-alanine at BGT1 and is thus the most potent inhibitor reported to date for this subtype (IC50 value of 2.5 µM). In addition, compound 9 displayed about 400, 16 and 40 times lower inhibitory potency at GAT1, GAT2 and GAT3, respectively. Compound 9 was shown to be a substrate for BGT1 and to have an overall similar pharmacological profile at the mouse orthologue. Compound 9 constitutes an interesting pharmacological tool for specifically investigating the cellular pharmacology of BGT1 and is the first small-molecule substrate identified with such a high selectivity for BGT1 over the three other GAT subtypes.
Collapse
|
39
|
Hiramatsu M. [Functional role for GABA transporters in the CNS]. Nihon Yakurigaku Zasshi 2014; 143:187-192. [PMID: 24717607 DOI: 10.1254/fpj.143.187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
|
40
|
Schousboe A, Madsen KK, Barker-Haliski ML, White HS. The GABA Synapse as a Target for Antiepileptic Drugs: A Historical Overview Focused on GABA Transporters. Neurochem Res 2014; 39:1980-7. [DOI: 10.1007/s11064-014-1263-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/10/2014] [Accepted: 02/12/2014] [Indexed: 01/18/2023]
|
41
|
Carver CM, Reddy DS. Neurosteroid interactions with synaptic and extrasynaptic GABA(A) receptors: regulation of subunit plasticity, phasic and tonic inhibition, and neuronal network excitability. Psychopharmacology (Berl) 2013; 230:151-88. [PMID: 24071826 PMCID: PMC3832254 DOI: 10.1007/s00213-013-3276-5] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 08/29/2013] [Indexed: 12/25/2022]
Abstract
RATIONALE Neurosteroids are steroids synthesized within the brain with rapid effects on neuronal excitability. Allopregnanolone, allotetrahydrodeoxycorticosterone, and androstanediol are three widely explored prototype endogenous neurosteroids. They have very different targets and functions compared to conventional steroid hormones. Neuronal γ-aminobutyric acid (GABA) type A (GABA(A)) receptors are one of the prime molecular targets of neurosteroids. OBJECTIVE This review provides a critical appraisal of recent advances in the pharmacology of endogenous neurosteroids that interact with GABA(A) receptors in the brain. Neurosteroids possess distinct, characteristic effects on the membrane potential and current conductance of the neuron, mainly via potentiation of GABA(A) receptors at low concentrations and direct activation of receptor chloride channel at higher concentrations. The GABA(A) receptor mediates two types of inhibition, now characterized as synaptic (phasic) and extrasynaptic (tonic) inhibition. Synaptic release of GABA results in the activation of low-affinity γ2-containing synaptic receptors, while high-affinity δ-containing extrasynaptic receptors are persistently activated by the ambient GABA present in the extracellular fluid. Neurosteroids are potent positive allosteric modulators of synaptic and extrasynaptic GABA(A) receptors and therefore enhance both phasic and tonic inhibition. Tonic inhibition is specifically more sensitive to neurosteroids. The resulting tonic conductance generates a form of shunting inhibition that controls neuronal network excitability, seizure susceptibility, and behavior. CONCLUSION The growing understanding of the mechanisms of neurosteroid regulation of the structure and function of the synaptic and extrasynaptic GABA(A) receptors provides many opportunities to create improved therapies for sleep, anxiety, stress, epilepsy, and other neuropsychiatric conditions.
Collapse
Affiliation(s)
- Chase Matthew Carver
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, 2008 Medical Research and Education Building, 8447 State Highway 47, Bryan, TX, 77807-3260, USA
| | | |
Collapse
|
42
|
Discovery of a subtype selective inhibitor of the human betaine/GABA transporter 1 (BGT-1) with a non-competitive pharmacological profile. Biochem Pharmacol 2013; 86:521-8. [DOI: 10.1016/j.bcp.2013.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 06/07/2013] [Accepted: 06/11/2013] [Indexed: 12/21/2022]
|
43
|
Jurik A, Reicherstorfer R, Zdrazil B, Ecker GF. Classification of High-Activity Tiagabine Analogs by Binary QSAR Modeling. Mol Inform 2013; 32:415-419. [PMID: 23956803 PMCID: PMC3743161 DOI: 10.1002/minf.201300020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/28/2013] [Indexed: 12/01/2022]
Affiliation(s)
- Andreas Jurik
- University of Vienna, Department of Medicinal Chemistry Althanstraße 14, A-1090 Vienna, Austria phone/fax: +431-4277-55110/-9551
| | | | | | | |
Collapse
|
44
|
Vogensen SB, Jørgensen L, Madsen KK, Borkar N, Wellendorph P, Skovgaard-Petersen J, Schousboe A, White HS, Krogsgaard-Larsen P, Clausen RP. Selective mGAT2 (BGT-1) GABA uptake inhibitors: design, synthesis, and pharmacological characterization. J Med Chem 2013; 56:2160-4. [PMID: 23398473 DOI: 10.1021/jm301872x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
β-Amino acids sharing a lipophilic diaromatic side chain were synthesized and characterized pharmacologically on mouse GABA transporter subtypes mGAT1-4. The parent amino acids were also characterized. Compounds 13a, 13b, and 17b displayed more than 6-fold selectivity for mGAT2 over mGAT1. Compound 17b displayed anticonvulsive properties inferring a role of mGAT2 in epileptic disorders. These results provide new neuropharmacological tools and a strategy for designing subtype selective GABA transport inhibitors.
Collapse
Affiliation(s)
- Stine B Vogensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schousboe A, Bak LK, Waagepetersen HS. Astrocytic Control of Biosynthesis and Turnover of the Neurotransmitters Glutamate and GABA. Front Endocrinol (Lausanne) 2013; 4:102. [PMID: 23966981 PMCID: PMC3744088 DOI: 10.3389/fendo.2013.00102] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/31/2013] [Indexed: 01/20/2023] Open
Abstract
Glutamate and GABA are the quantitatively major neurotransmitters in the brain mediating excitatory and inhibitory signaling, respectively. These amino acids are metabolically interrelated and at the same time they are tightly coupled to the intermediary metabolism including energy homeostasis. Astrocytes play a pivotal role in the maintenance of the neurotransmitter pools of glutamate and GABA since only these cells express pyruvate carboxylase, the enzyme required for de novo synthesis of the two amino acids. Such de novo synthesis is obligatory to compensate for catabolism of glutamate and GABA related to oxidative metabolism when the amino acids are used as energy substrates. This, in turn, is influenced by the extent to which the cycling of the amino acids between neurons and astrocytes may occur. This cycling is brought about by the glutamate/GABA - glutamine cycle the operation of which involves the enzymes glutamine synthetase (GS) and phosphate-activated glutaminase together with the plasma membrane transporters for glutamate, GABA, and glutamine. The distribution of these proteins between neurons and astrocytes determines the efficacy of the cycle and it is of particular importance that GS is exclusively expressed in astrocytes. It should be kept in mind that the operation of the cycle is associated with movement of ammonia nitrogen between the two cell types and different mechanisms which can mediate this have been proposed. This review is intended to delineate the above mentioned processes and to discuss quantitatively their relative importance in the homeostatic mechanisms responsible for the maintenance of optimal conditions for the respective neurotransmission processes to operate.
Collapse
Affiliation(s)
- Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Arne Schousboe, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark e-mail:
| | - Lasse K. Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helle S. Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
46
|
Ramaker MJ, Strong MN, Ford MM, Finn DA. Effect of ganaxolone and THIP on operant and limited-access ethanol self-administration. Neuropharmacology 2012; 63:555-64. [PMID: 22613838 DOI: 10.1016/j.neuropharm.2012.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/04/2012] [Accepted: 05/06/2012] [Indexed: 11/18/2022]
Abstract
Recent evidence suggests that GABA(A) receptor ligands may regulate ethanol intake via effects at both synaptic and extrasynaptic receptors. For example, the endogenous neurosteroid, allopregnanolone (ALLO) has a similar pharmacological profile as ethanol, and it alters ethanol intake in rodent models. Additionally, recent evidence suggests that δ-subunit-containing extrasynaptic GABA(A) receptors may confer high sensitivity to both ethanol and neurosteroids. The purpose of the present study was to determine the effects of ganaxolone (GAN; an ALLO analog) and gaboxadol (THIP; a GABA(A) receptor agonist with selectivity for the extrasynaptic δ-subunit) on ethanol intake, drinking patterns, and bout characteristics in operant and limited-access self-administration procedures. In separate studies, the effects of GAN (0-10 mg/kg) and THIP (2-16 mg/kg) were tested in C57BL/6J male mice provided with 2-h access to a two-bottle choice of water or 10% ethanol or trained to respond for 30 min of access to 10% ethanol. GAN had no overall significant effect on operant ethanol self-administration, but tended to decrease the latency to consume the first bout. In the limited-access procedure, GAN dose-dependently decreased ethanol intake. THIP dose-dependently decreased ethanol intake in both paradigms, altering both the consummatory and appetitive processes of operant self-administration as well as shifting the drinking patterns in both procedures. These results add to literature suggesting time-dependent effects of neurosteroids to promote the onset, and to subsequently decrease, ethanol drinking behavior, and they support a role for extrasynaptic GABA(A) receptor activation in ethanol reinforcement.
Collapse
Affiliation(s)
- Marcia J Ramaker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | |
Collapse
|
47
|
Héja L, Nyitrai G, Kékesi O, Dobolyi A, Szabó P, Fiáth R, Ulbert I, Pál-Szenthe B, Palkovits M, Kardos J. Astrocytes convert network excitation to tonic inhibition of neurons. BMC Biol 2012; 10:26. [PMID: 22420899 PMCID: PMC3342137 DOI: 10.1186/1741-7007-10-26] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/15/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Glutamate and γ-aminobutyric acid (GABA) transporters play important roles in balancing excitatory and inhibitory signals in the brain. Increasing evidence suggest that they may act concertedly to regulate extracellular levels of the neurotransmitters. RESULTS Here we present evidence that glutamate uptake-induced release of GABA from astrocytes has a direct impact on the excitability of pyramidal neurons in the hippocampus. We demonstrate that GABA, synthesized from the polyamine putrescine, is released from astrocytes by the reverse action of glial GABA transporter (GAT) subtypes GAT-2 or GAT-3. GABA release can be prevented by blocking glutamate uptake with the non-transportable inhibitor DHK, confirming that it is the glutamate transporter activity that triggers the reversal of GABA transporters, conceivably by elevating the intracellular Na+ concentration in astrocytes. The released GABA significantly contributes to the tonic inhibition of neurons in a network activity-dependent manner. Blockade of the Glu/GABA exchange mechanism increases the duration of seizure-like events in the low-[Mg2+] in vitro model of epilepsy. Under in vivo conditions the increased GABA release modulates the power of gamma range oscillation in the CA1 region, suggesting that the Glu/GABA exchange mechanism is also functioning in the intact hippocampus under physiological conditions. CONCLUSIONS The results suggest the existence of a novel molecular mechanism by which astrocytes transform glutamatergic excitation into GABAergic inhibition providing an adjustable, in situ negative feedback on the excitability of neurons.
Collapse
Affiliation(s)
- László Héja
- Department of Functional Pharmacology, Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Pusztaszeri 59-67, 1025 Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|