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
|
Fedorov NS, Sibgatullina GV, Malomouzh AI. Impairment of Skeletal Muscle Contraction by Inhibitors of GABA Transporters. Int J Mol Sci 2024; 25:12510. [PMID: 39684222 DOI: 10.3390/ijms252312510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
γ-Aminobutyric acid (GABA) has a significant impact on the functioning of not only the central but also the peripheral part of the nervous system. Recently, various elements of the GABAergic signaling system have been discovered in the area of the neuromuscular junction of mammals. At the same time, the functional activity of membrane-bound GABA transporters (GATs) and their role in neuromuscular transmission have not been identified. In the present study, performed on a neuromuscular preparation of the mouse diaphragm, the effect of GABA transporter inhibitors (nipecotic acid and β-alanine) on the force of muscle contraction was assessed. It was found that in the presence of both compounds in the bathing solution, the force of contractions caused by stimulation of the motor nerve dropped by 30-50%. However, when the muscle was stimulated directly, no effect of GABA transporter inhibitors on the contractile force was observed. The depressant effect of β-alanine induced by nerve stimulation was completely abolished by the GABAB receptor blocker CGP 55845. GABA transporters were detected at the neuromuscular junction using immunohistochemistry. Thus, our results indicate that GABA transporters are localized in the area of the neuromuscular junction, and their activity affects the muscle contraction force. This influence is most likely due to the removal of GABA released during nerve stimulation and activating GABA receptors, which leads to a decrease in the contraction force of the striated muscles.
Collapse
Affiliation(s)
- Nikita S Fedorov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
| | - Guzel V Sibgatullina
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
| | - Artem I Malomouzh
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
- Department of Radiophotonics and Microwave Technologies, Kazan National Research Technical University, 10 K. Marx St., Kazan 420111, Russia
| |
Collapse
|
3
|
Baron M, Devor M. Neurosteroids foster sedation by engaging tonic GABA A-Rs within the mesopontine tegmental anesthesia area (MPTA). Neurosci Lett 2024; 843:138030. [PMID: 39490574 DOI: 10.1016/j.neulet.2024.138030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Neurosteroids are endogenous molecules with anxiolytic, anticonvulsant, sleep-promoting and sedative effects. They are biosynthesized de novo within the brain, among other tissues, and are thought to act primarily as positive allosteric modulators of high-affinity extrasynaptic GABAAδ-receptors. The location of action of neurosteroids in the brain, however, remains unknown. We have demonstrated that GABAergic anesthetics act within the brainstem mesopontine tegmental anesthesia area (MPTA) to induce and maintain anesthetic loss-of-consciousness. Here we asked whether endogenous and synthetic neurosteroids might also act in the MPTA to induce their suppressive effects. Direct exposure of the MPTA to the endogenous neurosteroids pregnenolone and progesterone, their metabolites testosterone, allopregnanolone and 3α5α-THDOC, and the synthetic neurosteroids ganaxolone and alphaxalone, was found to be pro-anesthetic. Although we cannot rule out additional sites of action, results of this study suggest that the suppressive effects of neurosteroids are due, at least in part, to actions within the MPTA, presumably by recruitment of dedicated neuronal circuitry. This undermines the usual presumption that neurosteroids, like other sedatives, endogenous somnogens and anesthetics, act by nonspecific global distribution.
Collapse
Affiliation(s)
- Mark Baron
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Marshall Devor
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
4
|
Sebastião AM, Ribeiro JA. Adjusting the brakes to adjust neuronal activity: Adenosinergic modulation of GABAergic transmission. Neuropharmacology 2023; 236:109600. [PMID: 37225084 DOI: 10.1016/j.neuropharm.2023.109600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/20/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
About 50 years elapsed from the publication of the first full paper on the neuromodulatory action of adenosine at a 'simple' synapse model, the neuromuscular junction (Ginsborg and Hirst, 1972). In that study adenosine was used as a tool to increase cyclic AMP and for the great surprise, it decreased rather than increased neurotransmitter release, and for a further surprise, its action was prevented by theophylline, at the time only known as inhibitor of phosphodiesterases. These intriguing observations opened the curiosity for immediate studies relating the action of adenine nucleotides, known to be released together with neurotransmitters, to that of adenosine (Ribeiro and Walker, 1973, 1975). Our understanding on the ways adenosine uses to modulate synapses, circuits, and brain activity, vastly expanded since then. However, except for A2A receptors, whose actions upon GABAergic neurons of the striatum are well known, most of the attention given to the neuromodulatory action of adenosine has been focusing upon excitatory synapses. Evidence is growing that GABAergic transmission is also a target for adenosinergic neuromodulation through A1 and A2A receptors. Some o these actions have specific time windows during brain development, and others are selective for specific GABAergic neurons. Both tonic and phasic GABAergic transmission can be affected, and either neurons or astrocytes can be targeted. In some cases, those effects result from a concerted action with other neuromodulators. Implications of these actions in the control of neuronal function/dysfunction will be the focus of this review.
Collapse
Affiliation(s)
- Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| | - Joaquim Alexandre Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| |
Collapse
|
5
|
Goodspeed K, Mosca LR, Weitzel NC, Horning K, Simon EW, Pfalzer AC, Xia M, Langer K, Freed A, Bone M, Picone M, Bichell TJV. A draft conceptual model of SLC6A1 neurodevelopmental disorder. Front Neurosci 2023; 16:1026065. [PMID: 36741059 PMCID: PMC9893116 DOI: 10.3389/fnins.2022.1026065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/05/2022] [Indexed: 01/21/2023] Open
Abstract
Introduction SLC6A1 Neurodevelopmental Disorder (SLC6A1-NDD), first described in 2015, is a rare syndrome caused by a mutation in the SLC6A1 gene which encodes for the GABA Transporter 1 (GAT-1) protein. Epilepsy is one of the most common symptoms in patients and is often the primary treatment target, though the severity of epilepsy is variable. The impact of seizures and other symptoms of SLC6A1-NDD on patients and caregivers is wide-ranging and has not been described in a formal disease concept study. Methods A literature search was performed using the simple search term, "SLC6A1." Papers published before 2015, and those which did not describe the human neurodevelopmental disorder were removed from analysis. Open-ended interviews on lived experiences were conducted with two patient advocate key opinion leaders. An analysis of de-identified conversations between families of people with SLC6A1-NDD on social media was performed to quantify topics of concern. Results Published literature described symptoms in all of the following domains: neurological, visual, motor, cognitive, communication, behavior, gastrointestinal, sleep, musculo-skeletal, and emotional in addition to epilepsy. Key opinion leaders noted two unpublished features: altered hand use in infants, and developmental regression with onset of epilepsy. Analysis of social media interactions confirmed that the core symptoms of epilepsy and autistic traits were prominent concerns, but also demonstrated that other symptoms have a large impact on family life. Discussion For rare diseases, analysis of published literature is important, but may not be as comprehensive as that which can be gleaned from spontaneous interactions between families and through qualitative interviews. This report reflects our current understanding of the lived experience of SLC6A1-NDD. The discrepancy between the domains of disease reported in the literature and those discussed in patient conversations suggests that a formal qualitative interview-based disease concept study of SLC6A1-NDD is warranted.
Collapse
Affiliation(s)
- Kimberly Goodspeed
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lindsay R. Mosca
- College of Arts and Sciences, Vanderbilt University, Nashville, TN, United States
| | - Nicole C. Weitzel
- College of Arts and Sciences, Vanderbilt University, Nashville, TN, United States
| | | | - Elijah W. Simon
- College of Arts and Sciences, Vanderbilt University, Nashville, TN, United States
| | | | - Maya Xia
- COMBINEDBrain, Brentwood, TN, United States
| | - Katherine Langer
- College of Arts and Sciences, Vanderbilt University, Nashville, TN, United States
| | | | - Megan Bone
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Maria Picone
- TREND Community, Philadelphia, PA, United States
| | | |
Collapse
|
6
|
Bhatt M, Gauthier-Manuel L, Lazzarin E, Zerlotti R, Ziegler C, Bazzone A, Stockner T, Bossi E. A comparative review on the well-studied GAT1 and the understudied BGT-1 in the brain. Front Physiol 2023; 14:1145973. [PMID: 37123280 PMCID: PMC10137170 DOI: 10.3389/fphys.2023.1145973] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system (CNS). Its homeostasis is maintained by neuronal and glial GABA transporters (GATs). The four GATs identified in humans are GAT1 (SLC6A1), GAT2 (SLC6A13), GAT3 (SLC6A11), and betaine/GABA transporter-1 BGT-1 (SLC6A12) which are all members of the solute carrier 6 (SLC6) family of sodium-dependent transporters. While GAT1 has been investigated extensively, the other GABA transporters are less studied and their role in CNS is not clearly defined. Altered GABAergic neurotransmission is involved in different diseases, but the importance of the different transporters remained understudied and limits drug targeting. In this review, the well-studied GABA transporter GAT1 is compared with the less-studied BGT-1 with the aim to leverage the knowledge on GAT1 to shed new light on the open questions concerning BGT-1. The most recent knowledge on transporter structure, functions, expression, and localization is discussed along with their specific role as drug targets for neurological and neurodegenerative disorders. We review and discuss data on the binding sites for Na+, Cl-, substrates, and inhibitors by building on the recent cryo-EM structure of GAT1 to highlight specific molecular determinants of transporter functions. The role of the two proteins in GABA homeostasis is investigated by looking at the transport coupling mechanism, as well as structural and kinetic transport models. Furthermore, we review information on selective inhibitors together with the pharmacophore hypothesis of transporter substrates.
Collapse
Affiliation(s)
- Manan Bhatt
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Centre for Neuroscience—University of Insubria, Varese, Italy
| | - Laure Gauthier-Manuel
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
| | - Erika Lazzarin
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstr, Vienna
| | - Rocco Zerlotti
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
- Nanion Technologies GmbH, Munich, Germany
| | - Christine Ziegler
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
| | | | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstr, Vienna
- *Correspondence: Thomas Stockner, ; Elena Bossi,
| | - Elena Bossi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Centre for Neuroscience—University of Insubria, Varese, Italy
- *Correspondence: Thomas Stockner, ; Elena Bossi,
| |
Collapse
|
7
|
Singh K, Kumar P, Bhatia R, Mehta V, Kumar B, Akhtar MJ. Nipecotic acid as potential lead molecule for the development of GABA uptake inhibitors; structural insights and design strategies. Eur J Med Chem 2022; 234:114269. [DOI: 10.1016/j.ejmech.2022.114269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/05/2022] [Accepted: 03/05/2022] [Indexed: 11/04/2022]
|
8
|
Gabriel J, Höfner G, Wanner KT. Combination of MS Binding Assays and affinity selection mass spectrometry for screening of structurally homogenous libraries as exemplified for a focused oxime library addressing the neuronal GABA transporter 1. Eur J Med Chem 2020; 206:112598. [PMID: 32896797 DOI: 10.1016/j.ejmech.2020.112598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 11/30/2022]
Abstract
This study presents an efficient screening approach based on combination of mass spectrometry (MS) based binding assays (MS Binding Assays) and affinity selection mass spectrometry (ASMS) customized for screening of structurally homogeneous libraries sharing a common mass spectrometric fragmentation pattern. After reaction of a nipecotic acid derivative possessing a hydroxylamine functionality with aldehydes, the resulting oxime library was screened accordingly toward the GABA transporter subtype 1 (GAT1), a drug target for several neurological disorders. After assessing sublibraries' activities for inhibition of reporter ligand binding, hits in active ones were directly identified. This could be achieved by recording mass transitions for the reporter ligand as well as those predicted for the library components in a single LC-MS/MS run with a triple quadrupole mass spectrometer in the multiple reaction monitoring mode. Identification of hits with a predefined affinity could be reliably accomplished by calculation of IC50-values from specific binding concentrations of library constituents and reporter ligand. Application of this strategy revealed six hits, from which two of them were resynthesized for further biological evaluation. Thereby, the best one displayed a pKi of 7.38 in MS Binding Assays and a pIC50 of 6.82 in [3H]GABA uptake assays for GAT1.
Collapse
Affiliation(s)
- Jürgen Gabriel
- Faculty of Chemistry and Pharmacy, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Georg Höfner
- Faculty of Chemistry and Pharmacy, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Klaus T Wanner
- Faculty of Chemistry and Pharmacy, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
9
|
Gatta E, Guidotti A, Saudagar V, Grayson DR, Aspesi D, Pandey SC, Pinna G. Epigenetic Regulation of GABAergic Neurotransmission and Neurosteroid Biosynthesis in Alcohol Use Disorder. Int J Neuropsychopharmacol 2020; 24:130-141. [PMID: 32968808 PMCID: PMC7883893 DOI: 10.1093/ijnp/pyaa073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alcohol use disorder (AUD) is a chronic relapsing brain disorder. GABAA receptor (GABAAR) subunits are a target for the pharmacological effects of alcohol. Neurosteroids play an important role in the fine-tuning of GABAAR function in the brain. Recently, we have shown that AUD is associated with changes in DNA methylation mechanisms. However, the role of DNA methylation in the regulation of neurosteroid biosynthesis and GABAergic neurotransmission in AUD patients remains under-investigated. METHODS In a cohort of postmortem brains from 20 male controls and AUD patients, we investigated the expression of GABAAR subunits and neurosteroid biosynthetic enzymes and their regulation by DNA methylation mechanisms. Neurosteroid levels were quantified by gas chromatography-mass spectrometry. RESULTS The α 2 subunit expression was reduced due to increased DNA methylation at the gene promoter region in the cerebellum of AUD patients, a brain area particularly sensitive to the effects of alcohol. Alcohol-induced alteration in GABAAR subunits was also observed in the prefrontal cortex. Neurosteroid biosynthesis was also affected with reduced cerebellar expression of the 18kDa translocator protein and 3α-hydroxysteroid dehydrogenase mRNAs. Notably, increased DNA methylation levels were observed at the promoter region of 3α-hydroxysteroid dehydrogenase. These changes were associated with markedly reduced levels of allopregnanolone and pregnanolone in the cerebellum. CONCLUSION Given the key role of neurosteroids in modulating the strength of GABAAR-mediated inhibition, our data suggest that alcohol-induced impairments in GABAergic neurotransmission might be profoundly impacted by reduced neurosteroid biosynthesis most likely via DNA hypermethylation.
Collapse
Affiliation(s)
- Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois,Correspondence: Dr Alessandro Guidotti, Center for Alcohol Research in Epigenetics, Psychiatric Institute - Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612 ()
| | - Vikram Saudagar
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Dennis R Grayson
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Dario Aspesi
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Graziano Pinna
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, Chicago, Illinois
| |
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
|
Wang JY, Bai WZ, Gao YH, Zhang JL, Duanmu CL, Liu JL. GABAergic Inhibition of Spinal Cord Dorsal Horns Contributes to Analgesic Effect of Electroacupuncture in Incisional Neck Pain Rats. J Pain Res 2020; 13:1629-1645. [PMID: 32694919 PMCID: PMC7340366 DOI: 10.2147/jpr.s242330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/23/2020] [Indexed: 11/30/2022] Open
Abstract
Background Acupuncture has shown to be effective in relieving post-surgical pain. Nonetheless, its underlying mechanisms remain largely unknown. In the present study, we investigated the effect of electroacupuncture (EA) on the expression of GABA, GABA-A receptor (R) and GABA-BR in the spinal cord dorsal horns (DHs), and the involved neural cells in rats with incisional neck pain. Materials and Methods Male SD rats were randomly divided into control, model, Futu (LI18), Hegu-Neiguan (LI4-PC6), and Zusanli-Yanglingquan (ST36-GB34) groups. The incisional neck pain model was established by making a longitudinal incision and repeated mechanical separation along the thyroid gland region. EA (2Hz/100Hz, 1mA) was applied to LI18, LI4-PC6, ST36-GB34 separately for 30min, once at 4, 24 and 48h after incision. The local thermal pain threshold (TPT) of the focus was measured and the expression of GABA, and GABAR proteins and mRNAs detected by immunofluorescence stain and quantitative RT-PCR, respectively. Results The analgesic effect of LI18 and LI4-PC6 was superior to that of ST36-GB34 in incisional neck pain rats. Moreover, the EA stimulation of LI18 or LI4-PC6 increased the expression of GABA and GABA-Aα2 and GABA-Aβ3, GABA-B1, and GABA-B2 mRNAs in spinal DHs 4h after surgery, while GABA-A and GABA-B antagonists inhibited the analgesic effect of LI18. Immunofluorescence double staining showed that GABA was expressed on astrocytes and neurons, and GABA-B expressed only on neurons. Conclusion EA of both LI18 and LI4-PC6 has a good analgesic effect in incisional neck pain rats, which is closely related to their effects in upregulating the expression of GABA and its receptors in spinal DHs. The effects of LI18 and LI4-PC6 EA are obviously better that those of ST36-GB34 EA, and GABA is expressed on neurons and astrocytes.
Collapse
Affiliation(s)
- Jun-Ying Wang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Wan-Zhu Bai
- Laboratory for Architecture of Acupoints, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Yong-Hui Gao
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Jian-Liang Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Cheng-Lin Duanmu
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Jun-Ling Liu
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| |
Collapse
|
12
|
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
|
13
|
Liberato JL, Godoy LD, Cunha AOS, Mortari MR, de Oliveira Beleboni R, Fontana ACK, Lopes NP, Dos Santos WF. Parawixin2 Protects Hippocampal Cells in Experimental Temporal Lobe Epilepsy. Toxins (Basel) 2018; 10:toxins10120486. [PMID: 30469496 PMCID: PMC6316435 DOI: 10.3390/toxins10120486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/27/2018] [Accepted: 11/13/2018] [Indexed: 12/20/2022] Open
Abstract
Epilepsy is considered as one of the major disabling neuropathologies. Almost one third of adult patients with temporal lobe epilepsy (TLE) do not respond to current antiepileptic drugs (AEDs). Additionally, most AEDs do not have neuroprotective effects against the inherent neurodegenerative process underlying the hippocampal sclerosis on TLE. Dysfunctions in the GABAergic neurotransmission may contribute not only to the onset of epileptic activity but also constitute an important system for therapeutic approaches. Therefore, molecules that enhance GABA inhibitory effects could open novel avenues for the understanding of epileptic plasticity and for drug development. Parawixin2, a compound isolated from Parawixia bistriata spider venom, inhibits both GABA and glycine uptake and has an anticonvulsant effect against a wide range of chemoconvulsants. The neuroprotective potential of Parawixin2 was analyzed in a model of TLE induced by a long-lasting Status Epilepticus (SE), and its efficiency was compared to well-known neuroprotective drugs, such as riluzole and nipecotic acid. Neuroprotection was assessed through histological markers for cell density (Nissl), astrocytic reactivity (GFAP) and cell death labeling (TUNEL), which were performed 24 h and 72 h after SE. Parawixin2 treatment resulted in neuroprotective effects in a dose dependent manner at 24 h and 72 h after SE, as well as reduced reactive astrocytes and apoptotic cell death. Based on these findings, Parawixin2 has a great potential to be used as a tool for neuroscience research and as a probe to the development of novel GABAergic neuroprotective agents.
Collapse
Affiliation(s)
- José Luiz Liberato
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| | - Lívea Dornela Godoy
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| | - Alexandra Olimpio Siqueira Cunha
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
| | - Marcia Renata Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, DF 70910-900 Brasília, Brazil.
| | - Rene de Oliveira Beleboni
- Department of Biotechnology/School of Medicine, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, 14096-900 São Paulo, Brazil.
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102, USA.
| | - Norberto Peporine Lopes
- NPPNS, Department of Physics and Chemistry, College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil, Av. do Cafe s/n, Ribeirão Preto, 14040-903 São Paulo, Brazil.
| | - Wagner Ferreira Dos Santos
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| |
Collapse
|
14
|
Impaired Autophagy of GABAergic Interneurons in Neuropathic Pain. Pain Res Manag 2018; 2018:9185368. [PMID: 30356379 PMCID: PMC6176324 DOI: 10.1155/2018/9185368] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022]
Abstract
Neuropathic pain (NP) is caused by lesions of the peripheral fibers and central neurons in the somatosensory nervous system and affects 7-10% of the general population. Although the distinct cause of neuropathic pain has been investigated in primary afferent neurons over the years, pain modulation by central sensitization remains controversial. NP is believed to be driven by cell type-specific spinal synaptic plasticity in the dorsal horn. Upon intense afferent stimulation, spinothalamic tract neurons are potentiated, whereas GABAergic interneurons are inhibited leading to long-term depression. Growing evidences suggest that the inhibition of GABAergic neurons plays pivotal roles in the manifestation of neuropathic and inflammatory pain states. Downregulation of GABA transmission and impairment of GABAergic interneurons in the dorsal horn are critical consequences after spinal cord and peripheral nerve injuries. These impairments in GABAergic interneurons may be associated with dysfunctional autophagy, resulting in neuropathic pain. Here, we review an emerging number of investigations that suggest a pivotal role of impaired autophagy of GABAergic interneurons in NP. We discuss relevant research spurring the development of new targets and therapeutic agents of NP and emphasize the need for a multidisciplinary approach to manage NP in the future.
Collapse
|
15
|
Nowaczyk A, Fijałkowski Ł, Zaręba P, Sałat K. Docking and pharmacodynamic studies on hGAT1 inhibition activity in the presence of selected neuronal and astrocytic inhibitors. Part I. J Mol Graph Model 2018; 85:171-181. [PMID: 30219588 DOI: 10.1016/j.jmgm.2018.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/11/2018] [Accepted: 09/02/2018] [Indexed: 12/30/2022]
Abstract
Inhibition of 4-aminobutanoic acid (GABA) uptake is a strategy for enhancing GABA transmission. The utility of this approach is demonstrated by the successful development of such agents for the treatment of epilepsy and pain. Existing reports on acute brain slice preparations indicate the intersecting of complementary channels and receptors sets between astrocytes and neurons cells. Thorough analysis of astroglial cells by means of molecular and functional studies demonstrated their active modulatory role in intercellular communication. The chemical interactions between sixteen GABA analogues and isoform of hGAT1 is outlined in the light of molecular docking results. In the in vivo part antinociceptive properties of racemic nipecotic acid, its R and S enantiomers and isonipecotic acid, each administered intraperitoneally at 3 fixed doses (10, 30 and 100 mg/kg), were assessed in a thermally-induced acute pain model i.e. the mouse hot plate test. Docking analyses provided complex binding energies, specific h-bond components, and h-bond properties, such as energies, distances and angles. In vivo tests revealed statistically significant antinociceptive properties of isonipecotic acid (10 and 30 mg/kg), R-nipecotic acid (30 and 100 mg/kg) and S-nipecotic acid (100 mg/kg) in mice. The docking data endorse the hypothesis of correlation between the strength of their chemical interactions with hGAT1 and analgesic action of studied compounds.
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.
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Chair of Pharmaceutical Chemistry, Jagiellonian University Medical College, 9 Medyczna St., 30 - 688, Krakow, Poland.
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30 - 688, Krakow, Poland.
| |
Collapse
|
16
|
Astrocytic transporters in Alzheimer's disease. Biochem J 2017; 474:333-355. [DOI: 10.1042/bcj20160505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/16/2016] [Accepted: 11/29/2016] [Indexed: 12/26/2022]
Abstract
Astrocytes play a fundamental role in maintaining the health and function of the central nervous system. Increasing evidence indicates that astrocytes undergo both cellular and molecular changes at an early stage in neurological diseases, including Alzheimer's disease (AD). These changes may reflect a change from a neuroprotective to a neurotoxic phenotype. Given the lack of current disease-modifying therapies for AD, astrocytes have become an interesting and viable target for therapeutic intervention. The astrocyte transport system covers a diverse array of proteins involved in metabolic support, neurotransmission and synaptic architecture. Therefore, specific targeting of individual transporter families has the potential to suppress neurodegeneration, a characteristic hallmark of AD. A small number of the 400 transporter superfamilies are expressed in astrocytes, with evidence highlighting a fraction of these are implicated in AD. Here, we review the current evidence for six astrocytic transporter subfamilies involved in AD, as reported in both animal and human studies. This review confirms that astrocytes are indeed a viable target, highlights the complexities of studying astrocytes and provides future directives to exploit the potential of astrocytes in tackling AD.
Collapse
|
17
|
Rombo DM, Ribeiro JA, Sebastião AM. Hippocampal GABAergic transmission: a new target for adenosine control of excitability. J Neurochem 2016; 139:1056-1070. [PMID: 27778347 DOI: 10.1111/jnc.13872] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 01/01/2023]
Abstract
Physiological network functioning in the hippocampus is dependent on a balance between glutamatergic cell excitability and the activity of diverse local circuit neurons that release the inhibitory neurotransmitter γ-aminobutyric acid (GABA). Tuners of neuronal communication such as adenosine, an endogenous modulator of synapses, control hippocampal network operations by regulating excitability. Evidence has been recently accumulating on the influence of adenosine on different aspects of GABAergic transmission to shape hippocampal function. This review addresses how adenosine, through its high-affinity A1 (A1 R) and A2A receptors (A2A R), interferes with different GABA-mediated forms of inhibition in the hippocampus to regulate neuronal excitability. Adenosine-mediated modulation of phasic/tonic inhibitory transmission, of GABA transport mechanisms and its interference with other modulatory systems are discussed together with the putative implications for neuronal function in physiological and pathological conditions. This article is part of a mini review series: 'Synaptic Function and Dysfunction in Brain Diseases'.
Collapse
Affiliation(s)
- Diogo M Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
18
|
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
|
19
|
[3+2] Redox-Neutral Cycloaddition of Nitrocyclopropanes with Styrenes by Visible-Light Photocatalysis. Chemistry 2015; 21:9676-80. [DOI: 10.1002/chem.201500873] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 12/22/2022]
|
20
|
The effect of GABA transporter 1 (GAT1) inhibitor, tiagabine, on scopolamine-induced memory impairments in mice. Pharmacol Rep 2015; 67:1155-62. [PMID: 26481535 DOI: 10.1016/j.pharep.2015.04.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND GABAergic neurotransmission is involved in long-term potentiation, a neurophysiological basis for learning and memory. On the other hand, GABA-enhancing drugs may impair memory and learning in humans and animals. The present study aims at investigating the effect of GAT1 inhibitor tiagabine on memory and learning. METHODS Albino Swiss (CD-1) and C57BL/6J mice were used in the passive avoidance (PA), Morris water maze (MWM) and radial arm water maze (RAWM) tasks. Scopolamine (1mg/kg ip) was applied to induce cognitive deficits. RESULTS In the retention trial of PA scopolamine reduced step-through latency as compared to vehicle-treated mice, and pretreatment with tiagabine did not have any influence on this effect. In MWM the results obtained for vehicle-treated mice, scopolamine-treated group and combined scopolamine+tiagabine-treated mice revealed variable learning abilities in these groups. Tiagabine did not impair learning in the acquisition trial. In RAWM on day 1 scopolamine-treated group made nearly two-fold more errors than vehicle-treated mice and mice that received combined scopolamine and tiagabine. Learning abilities in the latter group were similar to those of vehicle-treated mice in the corresponding trial block on day 1, except for the last trial block, during which tiagabine+scopolamine-injected mice made more errors than control mice and the scopolamine-treated group. In all groups a complete reversal of memory deficits was observed in the last trial block of day 2. CONCLUSIONS The lack of negative influence of tiagabine on cognitive functions in animals with scopolamine-induced memory impairments may be relevant for patients treated with this drug.
Collapse
|
21
|
Ford A, Castonguay A, Cottet M, Little JW, Chen Z, Symons-Liguori AM, Doyle T, Egan TM, Vanderah TW, De Koninck Y, Tosh DK, Jacobson KA, Salvemini D. Engagement of the GABA to KCC2 signaling pathway contributes to the analgesic effects of A3AR agonists in neuropathic pain. J Neurosci 2015; 35:6057-67. [PMID: 25878279 PMCID: PMC4397603 DOI: 10.1523/jneurosci.4495-14.2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 01/18/2023] Open
Abstract
More than 1.5 billion people worldwide suffer from chronic pain, yet current treatment strategies often lack efficacy or have deleterious side effects in patients. Adenosine is an inhibitory neuromodulator that was previously thought to mediate antinociception through the A1 and A2A receptor subtypes. We have since demonstrated that A3AR agonists have potent analgesic actions in preclinical rodent models of neuropathic pain and that A3AR analgesia is independent of adenosine A1 or A2A unwanted effects. Herein, we explored the contribution of the GABA inhibitory system to A3AR-mediated analgesia using well-characterized mouse and rat models of chronic constriction injury (CCI)-induced neuropathic pain. The deregulation of GABA signaling in pathophysiological pain states is well established: GABA signaling can be hampered by a reduction in extracellular GABA synthesis by GAD65 and enhanced extracellular GABA reuptake via the GABA transporter, GAT-1. In neuropathic pain, GABAAR-mediated signaling can be further disrupted by the loss of the KCC2 chloride anion gradient. Here, we demonstrate that A3AR agonists (IB-MECA and MRS5698) reverse neuropathic pain via a spinal mechanism of action that modulates GABA activity. Spinal administration of the GABAA antagonist, bicuculline, disrupted A3AR-mediated analgesia. Furthermore, A3AR-mediated analgesia was associated with reductions in CCI-related GAD65 and GAT-1 serine dephosphorylation as well as an enhancement of KCC2 serine phosphorylation and activity. Our results suggest that A3AR-mediated reversal of neuropathic pain increases modulation of GABA inhibitory neurotransmission both directly and indirectly through protection of KCC2 function, underscoring the unique utility of A3AR agonists in chronic pain.
Collapse
Affiliation(s)
- Amanda Ford
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Annie Castonguay
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Martin Cottet
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Joshua W Little
- Department of Surgery, Center for Anatomical Science and Education, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Zhoumou Chen
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Ashley M Symons-Liguori
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Timothy Doyle
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Terrance M Egan
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Todd W Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Daniela Salvemini
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104,
| |
Collapse
|
22
|
Kobayashi T, Suemasa A, Igawa A, Ide S, Fukuda H, Abe H, Arisawa M, Minami M, Shuto S. Conformationally Restricted GABA with Bicyclo[3.1.0]hexane Backbone as the First Highly Selective BGT-1 Inhibitor. ACS Med Chem Lett 2014; 5:889-93. [PMID: 25147609 DOI: 10.1021/ml500134k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/16/2014] [Indexed: 11/29/2022] Open
Abstract
On the basis of the three-dimensional diversity-oriented conformational restriction strategy using key chiral cyclopropane units, we previously identified 3 ((2S,3R)-4-amino-3,4-methanobutyric acid) with a chiral trans-cyclopropane structure as a γ-aminobutyric acid (GABA) transporter inhibitor selective for GABA transporter (GAT) subtypes GAT-3 and BGT-1 (betaine/GABA transporter-1). Further conformational restriction of 3 with the rigid bicyclo[3.1.0]hexane backbone led to the successful development of the first highly potent and selective BGT-1 inhibitor 4 (IC50 = 0.59 μM). The bioactive conformation of 3 for BGT-1 was also identified.
Collapse
Affiliation(s)
- Takaaki Kobayashi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Akihiro Suemasa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Arisa Igawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Soichiro Ide
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hayato Fukuda
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroshi Abe
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mitsuhiro Arisawa
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Masabumi Minami
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Satoshi Shuto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12,
Nishi-6, Kita-ku, Sapporo 060-0812, Japan
- Center for Research
and Education on Drug Discovery, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
23
|
Kowalczyk P, Sałat K, Höfner GC, Mucha M, Rapacz A, Podkowa A, Filipek B, Wanner KT, Kulig K. Synthesis, biological evaluation and structure–activity relationship of new GABA uptake inhibitors, derivatives of 4-aminobutanamides. Eur J Med Chem 2014; 83:256-73. [DOI: 10.1016/j.ejmech.2014.06.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/04/2014] [Accepted: 06/12/2014] [Indexed: 11/17/2022]
|
24
|
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]
|
25
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: transporters. Br J Pharmacol 2013; 170:1706-96. [PMID: 24528242 PMCID: PMC3892292 DOI: 10.1111/bph.12450] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Transporters are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
26
|
Timple JMV, Magalhães LG, Souza Rezende KC, Pereira AC, Cunha WR, Andrade e Silva ML, Mortensen OV, Fontana ACK. The lignan (-)-hinokinin displays modulatory effects on human monoamine and GABA transporter activities. JOURNAL OF NATURAL PRODUCTS 2013; 76:1889-95. [PMID: 24112084 DOI: 10.1021/np400452n] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The neurotransmitter transporters of the SLC6 family play critical roles in the regulation of neurotransmission and are the primary targets of therapeutic agents used to treat clinical disorders involving compromised neurotransmitter signaling. The dopamine and norepinephrine transporters have been implicated in clinical disorders such as attention deficit hyperactivity disorder (ADHD) and substance abuse. The GABA transporters (GATs) serve as a target for anxiolytic, antidepressant, and antiepileptic therapies. In this work, the interaction with neurotransmitter transporters was characterized for a derivative of the lignan (-)-cubebin (1), namely, (-)-hinokinin (2). Using in vitro pharmacological assays, 2 selectively inhibited the human dopamine and norepinephrine transporters, in a noncompetitive manner possibly mediated by binding to a novel site within the transporters, and displayed low affinity for the serotonin transporter. Compound 2 also specifically inhibited the GAT-1 GABA transporter subtype. Compound 2 is not a substrate of the carriers as it had no effect on the efflux of either of the neurotransmitters investigated. This compound is inactive toward glutamate and glycine transporters. These results suggest that 2 may serve as a tool to develop new therapeutic drugs for ADHD and anxiety that target the DAT, NET, and GAT-1 transporters.
Collapse
Affiliation(s)
- Julie Marie V Timple
- Department of Pharmacology and Physiology, Drexel University College of Medicine , Philadelphia, Pennsylvania 19102, United States
| | | | | | | | | | | | | | | |
Collapse
|
27
|
2-Substituted 4-hydroxybutanamides as potential inhibitors of γ-aminobutyric acid transporters mGAT1–mGAT4: Synthesis and biological evaluation. Bioorg Med Chem 2013; 21:5154-67. [DOI: 10.1016/j.bmc.2013.06.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/12/2013] [Accepted: 06/16/2013] [Indexed: 11/22/2022]
|
28
|
Cyclopropane-based conformational restriction of GABA by a stereochemical diversity-oriented strategy: identification of an efficient lead for potent inhibitors of GABA transports. Bioorg Med Chem 2013; 21:4938-50. [PMID: 23886812 DOI: 10.1016/j.bmc.2013.06.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/26/2013] [Accepted: 06/26/2013] [Indexed: 12/22/2022]
Abstract
A series of cyclopropane-based conformationally restricted γ-aminobutyric acid (GABA) analogs with stereochemical diversity, that is, the trans- and cis-2,3-methano analogs Ia and Ib and their enantiomers ent-Ia and ent-Ib, and also the trans- and cis-3,4-methano analogs IIa and IIb and their enantiomers ent-IIa and ent-Iib, were synthesized from the chiral cyclopropane units Type-a and Type-b that we developed. These analogs were systematically evaluated with four GABA transporter (GAT) subtypes. The trans-3,4-methano analog IIa had inhibitory effects on GAT3 (IC50=23.9μM) and betaine-GABA transporter1 (5.48μM), indicating its potential as an effective lead compound for the development of potent GAT inhibitors due to its hydrophilic and low molecular weight properties and excellent ligand efficiency.
Collapse
|
29
|
Gelfuso EA, Liberato JL, Cunha AOS, Mortari MR, Beleboni RO, Lopes NP, Dos Santos WF. Parawixin2, a novel non-selective GABA uptake inhibitor from Parawixia bistriata spider venom, inhibits pentylenetetrazole-induced chemical kindling in rats. Neurosci Lett 2013; 543:12-6. [PMID: 23562887 DOI: 10.1016/j.neulet.2013.02.074] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/05/2013] [Accepted: 02/24/2013] [Indexed: 12/20/2022]
Abstract
The aims of the present work were to investigate the effects of the repeated administration of Parawixin2 (2-amino-5-ureidopentanamide; formerly FrPbAII), a novel GABA and glycine uptake inhibitor, in rats submitted to PTZ-induced kindling. Wistar rats were randomly divided in groups (n=6-8) for different treatments. Systemic injections of PTZ were administered every 48 h in the dose of 33 mg/kg; i.p., that is sufficient to induce fully kindled seizures in saline i.c.v. treated rats in a short period of time (28 days). Treatments in two types of positive controls (diazepam - DZP and nipecotic acid - NA groups) consisted in daily systemic injections of DZP (2mg/kg; i.p.) or i.c.v. injections of NA (12 μg/μL), while in experimental groups in daily i.c.v. injections of different doses of Parawixin2 (0.15; 0.075; 0.015 μg/μL). Seizures were analyzed using the Lamberty & Klitgaard score and kindling was considered as established after at least three consecutive seizures of score 4 or 5. Cumulative seizure scores for each group were analyzed using repeated measures of ANOVA followed by Tukey test. PTZ induced 4 and 5-score seizures after 12 injections in saline treated rats, whereas daily injection of Parawixin2 inhibited the onset of seizures in a dose dependent manner. Also, the challenging administration of PTZ did not raise seizure score in animals treated with the highest dose of Parawixin2 or those treated with DZP or NA. These findings together with previous data from our laboratory show that Parawixin2 could be a useful probe to design new antiepileptic drugs.
Collapse
Affiliation(s)
- Erica A Gelfuso
- Neurobiology and Venoms Laboratory, Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
30
|
Polley M, Höfner G, Wanner KT. Development and validation of an LC-ESI-MS/MS quantification method for a potential γ-aminobutyric acid transporter 3 (GAT3) marker and its application in preliminary MS binding assays. Biomed Chromatogr 2012; 27:641-54. [PMID: 23225341 DOI: 10.1002/bmc.2841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/17/2012] [Accepted: 10/17/2012] [Indexed: 11/10/2022]
Abstract
Binding assays for the γ-aminobutyric acid (GABA) transporter GAT3 can be assumed to significantly facilitate screening for respective inhibitors. As appropriate labeled ligands for this promising drug target are not available so far, we started efforts to set up mass spectrometry-based binding assays (MS binding assays), for which labeled markers are not required. Therefore, we developed a sensitive and rapid LC-ESI-MS/MS quantification method for DDPM-1007 {(RS)-1-[4,4,4-Tris(4-methoxyphenyl)but-2-en-1-yl]piperidine-3-carboxylic acid}, one of the most potent GAT3 inhibitors yet known, as a potential GAT3 marker. Using a 50 × 2 mm C(8) column in combination with a mobile phase composed of 10 mM ammonium bicarbonate buffer pH 8.0 and acetonitrile (60:40, v/v) at a flow rate of 450 μL/min DDPM-1007 could be analyzed in the positive multiple reaction monitoring mode [(m/z) 502.5 → 265.4] within a chromatographic cycle time of 3 min. Deuterated DDPM-1007 [((2)H(9))DDPM-1007] was synthesized and employed as internal standard. This way DDPM-1007 could be quantified in a range from 100 pM to 10 nM in the matrix resulting from respective binding experiments without any sample preparation. The established quantification method met the requirements of the FDA guidance for bioanalytical method validation concerning linearity and intra- and inter-batch accuracy. Based on this LC-ESI-MS/MS quantification preliminary MS binding assays employing membrane preparations obtained from a stably GAT3 expressing HEK293 cell line and DDPM-1007 as nonlabeled GAT3 marker could be performed. In these experiments specific binding of DDPM-1007 at GAT3 could be unambiguously detected. Additionally, the established LC-MS method provides a suitable analytical tool for further pharmacokinetic characterization of DDPM-1007, as exemplified for its logD determination.
Collapse
Affiliation(s)
- Maria Polley
- Department Pharmazie - Zentrum für Pharmaforschung, Ludwig-Maximilians-Universität München, Butenandtstr. 7, 81377 München, Germany
| | | | | |
Collapse
|
31
|
Eipper-Mains JE, Kiraly DD, Duff MO, Horowitz MJ, McManus CJ, Eipper BA, Graveley BR, Mains RE. Effects of cocaine and withdrawal on the mouse nucleus accumbens transcriptome. GENES BRAIN AND BEHAVIOR 2012; 12:21-33. [PMID: 23094851 DOI: 10.1111/j.1601-183x.2012.00873.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/24/2012] [Accepted: 10/17/2012] [Indexed: 12/13/2022]
Abstract
Genetic association studies, pharmacological investigations and analysis of mice-lacking individual genes have made it clear that Cocaine administration and Withdrawal have a profound impact on multiple neurotransmitter systems. The GABAergic medium spiny neurons of the nucleus accumbens (NAc) exhibit changes in the expression of genes encoding receptors for glutamate and in the signaling pathways triggered by dopamine binding to G-protein-coupled dopamine receptors. Deep sequence analysis provides a sensitive, quantitative and global analysis of the effects of Cocaine on the NAc transcriptome. RNA prepared from the NAc of adult male mice receiving daily injections of Saline or Cocaine, or Cocaine followed by a period of Withdrawal, was used for high-throughput sequence analysis. Changes were validated by quantitative polymerase chain reaction or Western blot. On the basis of pathway analysis, a preponderance of the genes affected by Cocaine and Withdrawal was involved in the cadherin, heterotrimeric G-protein and Wnt signaling pathways. Distinct subsets of cadherins and protocadherins exhibited a sustained increase or decrease in expression. Sustained down-regulation of several heterotrimeric G-protein β- and γ-subunits was observed. In addition to altered expression of receptors for small molecule neurotransmitters, neuropeptides and endocannabinoids, changes in the expression of plasma membrane transporters and vesicular neurotransmitter transporters were also observed. The effects of chronic Cocaine and Withdrawal on the expression of genes essential to cholinergic, glutamatergic, GABAergic, peptidergic and endocannabinoid signaling are as profound as their effects on dopaminergic transmission. Simultaneous targeting of multiple Withdrawal-specific changes in gene expression may facilitate development of new therapeutic approaches that are better able to prevent relapse.
Collapse
Affiliation(s)
- J E Eipper-Mains
- Department of Genetics & Developmental Biology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
A steered molecular dynamics study of binding and translocation processes in the GABA transporter. PLoS One 2012; 7:e39360. [PMID: 22737235 PMCID: PMC3380839 DOI: 10.1371/journal.pone.0039360] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 05/21/2012] [Indexed: 12/03/2022] Open
Abstract
The entire substrate translocation pathway in the human GABA transporter (GAT-1) was explored for the endogenous substrate GABA and the anti-convulsive drug tiagabine. Following a steered molecular dynamics (SMD) approach, in which a harmonic restraining potential is applied to the ligand, dissociation and re-association of ligands were simulated revealing events leading to substrate (GABA) translocation and inhibitor (tiagabine) mechanism of action. We succeeded in turning the transporter from the outward facing occluded to the open-to-out conformation, and also to reorient the transporter to the open-to-in conformation. The simulations are validated by literature data and provide a substrate pathway fingerprint in terms of which, how, and in which sequence specific residues are interacted with. They reveal the essential functional roles of specific residues, e.g. the role of charged residues in the extracellular vestibule including two lysines (K76 (TM1) and K448 (TM10)) and a TM6-triad (D281, E283, and D287) in attracting and relocating substrates towards the secondary/interim substrate-binding site (S2). Likewise, E101 is highlighted as essential for the relocation of the substrate from the primary substrate-binding site (S1) towards the cytoplasm.
Collapse
|
33
|
Daniele S, Da Pozzo E, Abelli M, Panighini A, Pini S, Gesi C, Lari L, Cardini A, Cassano GB, Martini C. Platelet uptake of GABA and glutamate in patients with bipolar disorder. Bipolar Disord 2012; 14:301-8. [PMID: 22548903 DOI: 10.1111/j.1399-5618.2012.01005.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Gamma aminobutyric acid (GABA) and glutamate (Glu) are the major neurotransmitters of the human central nervous system, and their actions are determined by specific transporters. Several studies suggest that GABA- and Glu-uptake mechanisms are modified in patients with bipolar disorder (BD). We explored the functionality of the GABA and Glu transporters in three groups of patients with BD, each with a different polarity of index episode (manic, depressive, or euthymic) at the time of blood draw. METHODS Forty patients with a diagnosis of BD, according to DSM-IV-TR criteria, and 15 healthy subjects were enrolled in the study. GABA and Glu uptake were evaluated in freshly prepared platelets using [(3) H]GABA or [(3) H]glutamate. RESULTS Compared to controls, GABA uptake was significantly increased in patients with depressive episodes and significantly decreased in subjects with manic episodes. Glu uptake was significantly increased in patients with index manic episodes and in euthymic patients compared to healthy controls. Moreover, a positive correlation was found between GABA platelet uptake and Hamilton Depression Rating Scale scores and between Glu platelet uptake and Young Mania Rating Scale scores in patients with manic episodes. CONCLUSIONS We found a relationship between GABA- and Glu-uptake levels and the polarity of episodes in patients with BD. Our data suggest that the functionality of both GABA and Glu transporters could represent a useful neurobiological marker to characterize the real polarity of an index episode of illness in patients with BD.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mechanisms Underlying Tolerance after Long-Term Benzodiazepine Use: A Future for Subtype-Selective GABA(A) Receptor Modulators? Adv Pharmacol Sci 2012; 2012:416864. [PMID: 22536226 PMCID: PMC3321276 DOI: 10.1155/2012/416864] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 10/10/2011] [Accepted: 11/02/2011] [Indexed: 01/01/2023] Open
Abstract
Despite decades of basic and clinical research, our understanding of how benzodiazepines tend to lose their efficacy over time (tolerance) is at least incomplete. In appears that tolerance develops relatively quickly for the sedative and anticonvulsant actions of benzodiazepines, whereas tolerance to anxiolytic and amnesic effects probably does not develop at all. In light of this evidence, we review the current evidence for the neuroadaptive mechanisms underlying benzodiazepine tolerance, including changes of (i) the GABA(A) receptor (subunit expression and receptor coupling), (ii) intracellular changes stemming from transcriptional and neurotrophic factors, (iii) ionotropic glutamate receptors, (iv) other neurotransmitters (serotonin, dopamine, and acetylcholine systems), and (v) the neurosteroid system. From the large variance in the studies, it appears that either different (simultaneous) tolerance mechanisms occur depending on the benzodiazepine effect, or that the tolerance-inducing mechanism depends on the activated GABA(A) receptor subtypes. Importantly, there is no convincing evidence that tolerance occurs with α subunit subtype-selective compounds acting at the benzodiazepine site.
Collapse
|
35
|
Kakinohana O, Hefferan MP, Miyanohara A, Nejime T, Marsala S, Juhas S, Juhasova J, Motlik J, Kucharova K, Strnadel J, Platoshyn O, Lazar P, Galik J, Vinay L, Marsala M. Combinational spinal GAD65 gene delivery and systemic GABA-mimetic treatment for modulation of spasticity. PLoS One 2012; 7:e30561. [PMID: 22291989 PMCID: PMC3264568 DOI: 10.1371/journal.pone.0030561] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 12/19/2011] [Indexed: 01/08/2023] Open
Abstract
Background Loss of GABA-mediated pre-synaptic inhibition after spinal injury plays a key role in the progressive increase in spinal reflexes and the appearance of spasticity. Clinical studies show that the use of baclofen (GABAB receptor agonist), while effective in modulating spasticity is associated with major side effects such as general sedation and progressive tolerance development. The goal of the present study was to assess if a combined therapy composed of spinal segment-specific upregulation of GAD65 (glutamate decarboxylase) gene once combined with systemic treatment with tiagabine (GABA uptake inhibitor) will lead to an antispasticity effect and whether such an effect will only be present in GAD65 gene over-expressing spinal segments. Methods/Principal Findings Adult Sprague-Dawley (SD) rats were exposed to transient spinal ischemia (10 min) to induce muscle spasticity. Animals then received lumbar injection of HIV1-CMV-GAD65 lentivirus (LVs) targeting ventral α-motoneuronal pools. At 2–3 weeks after lentivirus delivery animals were treated systemically with tiagabine (4, 10, 20 or 40 mg/kg or vehicle) and the degree of spasticity response measured. In a separate experiment the expression of GAD65 gene after spinal parenchymal delivery of GAD65-lentivirus in naive minipigs was studied. Spastic SD rats receiving spinal injections of the GAD65 gene and treated with systemic tiagabine showed potent and tiagabine-dose-dependent alleviation of spasticity. Neither treatment alone (i.e., GAD65-LVs injection only or tiagabine treatment only) had any significant antispasticity effect nor had any detectable side effect. Measured antispasticity effect correlated with increase in spinal parenchymal GABA synthesis and was restricted to spinal segments overexpressing GAD65 gene. Conclusions/Significance These data show that treatment with orally bioavailable GABA-mimetic drugs if combined with spinal-segment-specific GAD65 gene overexpression can represent a novel and highly effective anti-spasticity treatment which is associated with minimal side effects and is restricted to GAD65-gene over-expressing spinal segments.
Collapse
Affiliation(s)
- Osamu Kakinohana
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Michael P. Hefferan
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Atsushi Miyanohara
- Gene Therapy Program and Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Tetsuya Nejime
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Silvia Marsala
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, AS CR, Liběchov, Czech Republic
| | - Jana Juhasova
- Institute of Animal Physiology and Genetics, AS CR, Liběchov, Czech Republic
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, AS CR, Liběchov, Czech Republic
| | - Karolina Kucharova
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Jan Strnadel
- Institute of Animal Physiology and Genetics, AS CR, Liběchov, Czech Republic
| | - Oleksandr Platoshyn
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Peter Lazar
- Department of Breeding and Diseases of Game and Fish, University of Veterinary Medicine and Pharmacy, Komenskeho, Košice, Slovakia
| | - Jan Galik
- Institute of Neurobiology, Slovak Academy of Sciences, Košice, Slovakia
- Faculty of Science, Institute of Biology and Ecology, Pavol Jozef Safarik University, Košice, Slovakia
| | - Laurent Vinay
- Laboratoire Plasticité et Physio-Pathologie de la Motricité (UMR6196), Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, Marseille, France
| | - Martin Marsala
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- Institute of Neurobiology, Slovak Academy of Sciences, Košice, Slovakia
- * E-mail:
| |
Collapse
|
36
|
Analgesic and anticonvulsant activity of new derivatives of 2-substituted 4-hydroxybutanamides in mice. Pharmacol Rep 2012; 64:102-12. [DOI: 10.1016/s1734-1140(12)70736-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 09/15/2011] [Indexed: 01/18/2023]
|
37
|
Tellez R, Gómez-Víquez L, Meneses A. GABA, glutamate, dopamine and serotonin transporters expression on memory formation and amnesia. Neurobiol Learn Mem 2011; 97:189-201. [PMID: 22183017 DOI: 10.1016/j.nlm.2011.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 11/11/2011] [Accepted: 12/01/2011] [Indexed: 10/14/2022]
Abstract
Notwithstanding several neurotransmission systems are frequently related to memory formation, amnesia and/or therapeutic targets for memory alterations, the role of transporters γ-aminobutyric acid (GABA, GAT1), glutamate (neuronal glutamate transporter excitatory amino acid carrier; EACC1), dopamine (DAT) and serotonin (SERT) is poorly understood. Hence, in this paper Western-blot analysis was used to evaluate expression changes on them during memory formation in trained and untrained rats treated with the selective serotonin transporter inhibitor fluoxetine, the amnesic drug d-methamphetamine (METH) and fluoxetine plus METH. Transporters expression was evaluated in the hippocampus, prefrontal cortex and striatum. Data indicated that in addition of memory performance other behavioral parameters (e.g., explorative behavior, food-intake, etc.) that memory formation was recorded. Thus, memory formation in a Pavlovian/instrumental autoshaping was associated to up-regulation of prefrontal cortex GAT1 and EAAC1, striatal SERT, DAT and EACC1; while, hippocampal EACC1, GAT1 and SERT were down-regulated. METH impaired short (STM) and long-term memory (LTM), at 24 or 48h. The METH-induced amnesia down-regulated SERT, DAT, EACC1 and GAT1 in hippocampus and the GAT1 in striatum; no-changes were observed in prefrontal cortex. Post-training administration of fluoxetine improved LTM (48h), which was associated to DAT, GAT1 (prefrontal cortex) up-regulation, but GAT1 (striatum) and SERT (hippocampus) down-regulation. Fluoxetine plus METH administration was able to prevent amnesia, which was associated to DAT, EACC1 and GAT1 (prefrontal cortex), SERT and DAT (hippocampus) and EACC1 or DAT (striatal) up-regulation. Together these data show that memory formation, amnesia and anti-amnesic effects are associated to specific patters of transporters expression.
Collapse
Affiliation(s)
- Ruth Tellez
- Depto. de Farmacobiología, CINVESTAV-IPN, Tenorios 235, Granjas Coapa, Mexico City 14330, Mexico
| | | | | |
Collapse
|
38
|
Kristensen AS, Andersen J, Jørgensen TN, Sørensen L, Eriksen J, Loland CJ, Strømgaard K, Gether U. SLC6 neurotransmitter transporters: structure, function, and regulation. Pharmacol Rev 2011; 63:585-640. [PMID: 21752877 DOI: 10.1124/pr.108.000869] [Citation(s) in RCA: 628] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The neurotransmitter transporters (NTTs) belonging to the solute carrier 6 (SLC6) gene family (also referred to as the neurotransmitter-sodium-symporter family or Na(+)/Cl(-)-dependent transporters) comprise a group of nine sodium- and chloride-dependent plasma membrane transporters for the monoamine neurotransmitters serotonin (5-hydroxytryptamine), dopamine, and norepinephrine, and the amino acid neurotransmitters GABA and glycine. The SLC6 NTTs are widely expressed in the mammalian brain and play an essential role in regulating neurotransmitter signaling and homeostasis by mediating uptake of released neurotransmitters from the extracellular space into neurons and glial cells. The transporters are targets for a wide range of therapeutic drugs used in treatment of psychiatric diseases, including major depression, anxiety disorders, attention deficit hyperactivity disorder and epilepsy. Furthermore, psychostimulants such as cocaine and amphetamines have the SLC6 NTTs as primary targets. Beginning with the determination of a high-resolution structure of a prokaryotic homolog of the mammalian SLC6 transporters in 2005, the understanding of the molecular structure, function, and pharmacology of these proteins has advanced rapidly. Furthermore, intensive efforts have been directed toward understanding the molecular and cellular mechanisms involved in regulation of the activity of this important class of transporters, leading to new methodological developments and important insights. This review provides an update of these advances and their implications for the current understanding of the SLC6 NTTs.
Collapse
Affiliation(s)
- Anders S Kristensen
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
GABA, the principal inhibitory neurotransmitter in the cerebral cortex, maintains the inhibitory tone that counterbalances neuronal excitation. The identification and subsequent development of GABA-transport inhibitors has shown the important role that GABA transporters play in the control of the CNS. To date, four GABA transporters have been cloned (GAT1-4). Compounds that inhibit GABA uptake are targets for epilepsy treatment. Currently, they are also being investigated for other possible indications such as the treatment of psychosis, general anxiety and sleep disorders, drug addiction, acute and chronic pain. These and other issues are discussed in this article.
Collapse
|
40
|
Lehre A, Rowley N, Zhou Y, Holmseth S, Guo C, Holen T, Hua R, Laake P, Olofsson A, Poblete-Naredo I, Rusakov D, Madsen K, Clausen R, Schousboe A, White H, Danbolt N. Deletion of the betaine-GABA transporter (BGT1; slc6a12) gene does not affect seizure thresholds of adult mice. Epilepsy Res 2011; 95:70-81. [PMID: 21459558 PMCID: PMC3376448 DOI: 10.1016/j.eplepsyres.2011.02.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 02/02/2011] [Accepted: 02/27/2011] [Indexed: 10/18/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mammalian brain. Once released, it is removed from the extracellular space by cellular uptake catalyzed by GABA transporter proteins. Four GABA transporters (GAT1, GAT2, GAT3 and BGT1) have been identified. Inhibition of the GAT1 by the clinically available anti-epileptic drug tiagabine has been an effective strategy for the treatment of some patients with partial seizures. Recently, the investigational drug EF1502, which inhibits both GAT1 and BGT1, was found to exert an anti-convulsant action synergistic to that of tiagabine, supposedly due to inhibition of BGT1. The present study addresses the role of BGT1 in seizure control and the effect of EF1502 by developing and exploring a new mouse line lacking exons 3-5 of the BGT1 (slc6a12) gene. The deletion of this sequence abolishes the expression of BGT1 mRNA. However, homozygous BGT1-deficient mice have normal development and show seizure susceptibility indistinguishable from that in wild-type mice in a variety of seizure threshold models including: corneal kindling, the minimal clonic and minimal tonic extension seizure threshold tests, the 6Hz seizure threshold test, and the i.v. pentylenetetrazol threshold test. We confirm that BGT1 mRNA is present in the brain, but find that the levels are several hundred times lower than those of GAT1 mRNA; possibly explaining the apparent lack of phenotype. In conclusion, the present results do not support a role for BGT1 in the control of seizure susceptibility and cannot provide a mechanistic understanding of the synergism that has been previously reported with tiagabine and EF1502.
Collapse
Affiliation(s)
- A.C. Lehre
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - N.M. Rowley
- Department of Pharmacology and Toxicology, Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT, USA
| | - Y. Zhou
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - S. Holmseth
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - C. Guo
- HHMI, Janelia Farm Research Campus, Ashburn, VA, USA
| | - T. Holen
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - R. Hua
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - P. Laake
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - A.M. Olofsson
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| | - I. Poblete-Naredo
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, México D.F., Mexico
| | - D.A. Rusakov
- UCL Institute of Neurology, University College London, UK
| | - K.K. Madsen
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmaceutical sciences, University of Copenhagen, Denmark
| | - R.P. Clausen
- Department of medicinal chemistry, Faculty of Pharmaceutical sciences, University of Copenhagen, Denmark
| | - A. Schousboe
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmaceutical sciences, University of Copenhagen, Denmark
| | - H.S. White
- Department of Pharmacology and Toxicology, Anticonvulsant Drug Development Program, University of Utah, Salt Lake City, UT, USA
| | - N.C. Danbolt
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, N-0317 Oslo, Norway
| |
Collapse
|
41
|
Gillies SG, Haddley K, Vasiliou SA, Jacobson GM, von Mentzer B, Bubb VJ, Quinn JP. Distinct gene expression profiles directed by the isoforms of the transcription factor neuron-restrictive silencer factor in human SK-N-AS neuroblastoma cells. J Mol Neurosci 2011; 44:77-90. [PMID: 20652837 DOI: 10.1007/s12031-010-9420-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 06/25/2010] [Indexed: 11/28/2022]
Abstract
Neuron-restrictive silencer factor (NRSF) and its isoforms are differentially regulated in rodent models of self-sustaining status epilepticus (SSSE). NRSF isoforms regulate genes associated with SSSE, including the proconvulsant tachykinins, brain-derived neurotrophic factor and multiple ion channels. NRSF isoforms may direct distinct gene expression patterns during SSSE, and the ratio of each isoform may be a causative factor in traumatic damage to the central nervous system. Here, we analysed global gene expression changes by microarray in human SK-N-AS neuroblastoma cells following the over-expression of NRSF and a truncated isoform, HZ4. We used bioinformatics software to analyse the microarray dataset and correlated these data with epilepsy candidate gene pathways. Findings were validated by reverse transcriptase-polymerase chain reaction. We demonstrated that NRSF and HZ4 direct overlapping as well as distinct gene expression patterns, and that they differentially modulated gene expression patterns associated with epilepsy. Finally, we revealed that NRSF gene expression may be modulated by the anticonvulsant, phenytoin. We have interpreted our data to reflect altered gene expression directed by NRSF that might be relevant for SSSE.
Collapse
Affiliation(s)
- Stuart G Gillies
- Division of Human Anatomy & Cell Biology and Division of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Gwak YS, Hulsebosch CE. GABA and central neuropathic pain following spinal cord injury. Neuropharmacology 2011; 60:799-808. [PMID: 21216257 PMCID: PMC3285561 DOI: 10.1016/j.neuropharm.2010.12.030] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 12/02/2010] [Accepted: 12/22/2010] [Indexed: 12/31/2022]
Abstract
Spinal cord injury induces maladaptive synaptic transmission in the somatosensory system that results in chronic central neuropathic pain. Recent literature suggests that glial-neuronal interactions are important modulators in synaptic transmission following spinal cord injury. Neuronal hyperexcitability is one of the predominant phenomenon caused by maladaptive synaptic transmission via altered glial-neuronal interactions after spinal cord injury. In the somatosensory system, spinal inhibitory neurons counter balance the enhanced synaptic transmission from peripheral input. For a decade, the literature suggests that hypofunction of GABAergic inhibitory tone is an important factor in the enhanced synaptic transmission that often results in neuronal hyperexcitability in dorsal horn neurons following spinal cord injury. Neurons and glial cells synergistically control intracellular chloride ion gradients via modulation of chloride transporters, extracellular glutamate and GABA concentrations via uptake mechanisms. Thus, the intracellular "GABA-glutamate-glutamine cycle" is maintained for normal physiological homeostasis. However, hyperexcitable neurons and glial activation after spinal cord injury disrupts the balance of chloride ions, glutamate and GABA distribution in the spinal dorsal horn and results in chronic neuropathic pain. In this review, we address spinal cord injury induced mechanisms in hypofunction of GABAergic tone that results in chronic central neuropathic pain. This article is part of a Special Issue entitled 'Synaptic Plasticity & Interneurons'.
Collapse
Affiliation(s)
- Young S Gwak
- Department of Neuroscience & Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA.
| | | |
Collapse
|
43
|
Vinkers CH, Mirza NR, Olivier B, Kahn RS. The inhibitory GABA system as a therapeutic target for cognitive symptoms in schizophrenia: investigational agents in the pipeline. Expert Opin Investig Drugs 2011; 19:1217-33. [PMID: 20812877 DOI: 10.1517/13543784.2010.513382] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Cognitive impairments associated with schizophrenia include neuropsychological deficits in attention, working memory, learning and executive function. Because these cognitive deficits precede the onset of psychosis, are present in non-affected relatives and constitute the best predictor of functional outcome, they are a cardinal clinical feature in schizophrenia. Currently, no effective treatment for the cognitive symptoms in schizophrenia exists. AREAS COVERED IN THIS REVIEW There is evidence that the inhibitory GABA system is affected in schizophrenia, suggesting that cognitive impairments associated with schizophrenia may be effectively treated by drugs that modulate the GABA(A) receptor. However, classical benzodiazepines produce cognitive impairments and are associated with numerous side effects. The recent development of compounds with selective efficacy for different α subunits at the benzodiazepine site of the GABA(A) receptor has renewed interest for the therapeutic potential of GABAergic drugs. WHAT THE READER WILL GAIN This review summarizes the involvement of the inhibitory GABA system in the cognitive abnormalities of schizophrenia and discusses putative (selective) GABAergic cognition-enhancing drugs for schizophrenia. TAKE HOME MESSAGE If cognitive abnormalities in schizophrenic individuals are the result of GABAergic dysfunction, selectively modulating the GABA system could comprise a promising therapeutic intervention for cognitive symptoms in schizophrenia.
Collapse
Affiliation(s)
- Christiaan H Vinkers
- Department of Psychiatry, University Medical Center Utrecht, Rudolf Magnus Institute of Neuroscience, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
44
|
Kulig K, Więckowski K, Więckowska A, Gajda J, Pochwat B, Höfner GC, Wanner KT, Malawska B. Synthesis and biological evaluation of new derivatives of 2-substituted 4-hydroxybutanamides as GABA uptake inhibitors. Eur J Med Chem 2011; 46:183-90. [DOI: 10.1016/j.ejmech.2010.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 10/19/2010] [Accepted: 11/01/2010] [Indexed: 11/15/2022]
|
45
|
Zhuang W, Zhao X, Zhao G, Guo L, Lian Y, Zhou J, Fang D. Synthesis and biological evaluation of 4-fluoroproline and 4-fluoropyrrolidine-2-acetic acid derivatives as new GABA uptake inhibitors. Bioorg Med Chem 2009; 17:6540-6. [PMID: 19703775 DOI: 10.1016/j.bmc.2009.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 08/03/2009] [Accepted: 08/04/2009] [Indexed: 11/26/2022]
Abstract
Preparation for the N-alkylated derivatives of enantiomerically pure (2S)-4-fluoroproline and (2S)-4-fluoropyrrolidine-2-acetic acid is described. The final compounds were evaluated as potential GAT-1 uptake inhibitors via cultured cell lines expressing mouse GAT-1. Compared with their corresponding 4-hydroxy compounds, these derivatives exhibited slight improvement on their inhibitory potency, but still much weaker than their corresponding compounds with no substituents at the C-4 of the pyrrolidine moiety, with the most potent affinity being about 1/15 fold as that of Tiagabine. The drastic decrease of their affinity may arise from sharp reduction of their basicity due to strong inductive effect of the 4-fluorine. However the configuration of the C-4 linking fluorine did not have much influence on their affinity for GAT-1.
Collapse
Affiliation(s)
- Weiping Zhuang
- Fujian Institute of Microbiology, No. 25, Jinbu road, Fuzhou 350007, China
| | | | | | | | | | | | | |
Collapse
|
46
|
β-hydroxybutyrate alters GABA-transaminase activity in cultured astrocytes. Brain Res 2009; 1268:17-23. [DOI: 10.1016/j.brainres.2009.02.074] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 02/06/2009] [Accepted: 02/28/2009] [Indexed: 11/21/2022]
|
47
|
Baliova M, Knab A, Franekova V, Jursky F. Modification of the cytosolic regions of GABA transporter GAT1 by calpain. Neurochem Int 2009; 55:288-94. [PMID: 19576516 DOI: 10.1016/j.neuint.2009.03.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 02/20/2009] [Accepted: 03/18/2009] [Indexed: 11/18/2022]
Abstract
Cytosolic regions of sodium dependent neurotransmitter transporters regulate their surface density and transporting function by interconnecting themselves with intracellular signaling pathways. Here we show that calpain activation in rat brain synaptosomes leads to cleavage of both N- and C-terminal regions of GABA transporter GAT1. In the C-terminal region, calpain removes a short segment of amino acids involved in binding of GAT1 to a high-density PDZ anchoring matrix. Using a protein pull-down assay, we found that C-terminal truncation of GAT1 results in modification of its interacting proteome in vitro. Results indicate that calpain activation/inhibition in GABAergic terminals may influence the scaffolding and surface expression of GABA transporter GAT1 under normal conditions or imbalance GAT1-mediated GABAergic transmission under pathological states.
Collapse
Affiliation(s)
- Martina Baliova
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Dubravska cesta 21, 84251 Bratislava, Slovakia
| | | | | | | |
Collapse
|
48
|
Kwakowsky A, Schwirtlich M, Kooy F, Ábrahám I, Máté Z, Katarova Z, Szabó G. GABA neurotransmitter signaling in the developing mouse lens: Dynamic regulation of components and functionality. Dev Dyn 2008; 237:3830-41. [DOI: 10.1002/dvdy.21768] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
49
|
Gwak YS, Crown ED, Unabia GC, Hulsebosch CE. Propentofylline attenuates allodynia, glial activation and modulates GABAergic tone after spinal cord injury in the rat. Pain 2008; 138:410-422. [PMID: 18353556 PMCID: PMC2676790 DOI: 10.1016/j.pain.2008.01.021] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 12/19/2007] [Accepted: 01/22/2008] [Indexed: 01/05/2023]
Abstract
In this study, we evaluated whether propentofylline, a methylxanthine derivative, modulates spinal glial activation and GABAergic inhibitory tone by modulation of glutamic acid decarboxylase (GAD)(65), the GABA synthase enzyme, in the spinal dorsal horn following spinal cord injury (SCI). Sprague-Dawley rats (225-250 g) were given a unilateral spinal transverse injury, from dorsal to ventral, at the T13 spinal segment. Unilateral spinal injured rats developed robust bilateral hindlimb mechanical allodynia and hyperexcitability of spinal wide dynamic range (WDR) neurons in the lumbar enlargement (L4-L5) compared to sham controls, which was attenuated by intrathecal (i.t.) administration of GABA, dose-dependently (0.01, 0.1, 0.5 microg). Western blotting and immunohistochemical data demonstrated that the expression level of GAD(65) protein significantly decreased on both sides of the lumbar dorsal horn (L4/5) after SCI (p<0.05). In addition, astrocytes and microglia showed soma hypertrophy as determined by increased soma area and increased GFAP and CD11b on both sides of the lumbar dorsal horn compared to sham controls, respectively (p<0.05). Intrathecal treatment with propentofylline (PPF 10 mM) significantly attenuated the astrocytic and microglial soma hypertrophy and mechanical allodynia (p<0.05). Additionally, the Western blotting and immunohistochemistry data demonstrated that i.t. treatment of PPF significantly prevented the decrease of GAD(65) expression in both sides of the lumbar dorsal horn following SCI (p<0.05). In conclusion, our present data demonstrate that propentofylline modulates glia activation and GABAergic inhibitory tone by modulation of GAD(65) protein expression following spinal cord injury.
Collapse
Affiliation(s)
| | | | | | - Claire E. Hulsebosch
- Department of Neuroscience & Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA
| |
Collapse
|
50
|
Fast homeostatic plasticity of inhibition via activity-dependent vesicular filling. PLoS One 2008; 3:e2979. [PMID: 18714334 PMCID: PMC2495031 DOI: 10.1371/journal.pone.0002979] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/14/2008] [Indexed: 11/30/2022] Open
Abstract
Synaptic activity in the central nervous system undergoes rapid state-dependent changes, requiring constant adaptation of the homeostasis between excitation and inhibition. The underlying mechanisms are, however, largely unclear. Chronic changes in network activity result in enhanced production of the inhibitory transmitter GABA, indicating that presynaptic GABA content is a variable parameter for homeostatic plasticity. Here we tested whether such changes in inhibitory transmitter content do also occur at the fast time scale required to ensure inhibition-excitation-homeostasis in dynamic cortical networks. We found that intense stimulation of afferent fibers in the CA1 region of mouse hippocampal slices yielded a rapid and lasting increase in quantal size of miniature inhibitory postsynaptic currents. This potentiation was mediated by the uptake of GABA and glutamate into presynaptic endings of inhibitory interneurons (the latter serving as precursor for the synthesis of GABA). Thus, enhanced release of inhibitory and excitatory transmitters from active networks leads to enhanced presynaptic GABA content. Thereby, inhibitory efficacy follows local neuronal activity, constituting a negative feedback loop and providing a mechanism for rapid homeostatic scaling in cortical circuits.
Collapse
|