1
|
Lemons AH, Murphy B, Dengler JS, Salar S, Davies PA, Smalley JL, Moss SJ. Neuroactive steroids activate membrane progesterone receptors to induce sex specific effects on protein kinase activity. iScience 2025; 28:112352. [PMID: 40292327 PMCID: PMC12032937 DOI: 10.1016/j.isci.2025.112352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/30/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Neuroactive steroids (NAS), which are synthesized in the brain from progesterone, exert potent effects on behavior and are used to treat postpartum depression, yet how these compounds induce sustained modifications in neuronal activity are ill-defined. Here, we examined the efficacy of NAS for membrane progesterone receptors (mPRs) δ and ε, members of a family of GPCRs for progestins that are expressed in the CNS. NAS increase PKC activity via the Gq activation of mPRδ with EC50s between 3 and 11nM. In contrast, they activate Gs via mPRε to potentiate PKA activity with similar potencies. NAS also induced the rapid internalization of only mPRδ. In the forebrain of female mice, mPRδ expression levels were 8-fold higher than in males. Consistent with this, the activation of PKC by NAS was evident in acute brain slices from female mice. Collectively, our results suggest that NAS may exert sex-specific effects on intracellular signaling in the brain via the activation of mPRs.
Collapse
Affiliation(s)
- Abigail H.S. Lemons
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Briana Murphy
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Jake S. Dengler
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Seda Salar
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Paul A. Davies
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Joshua L. Smalley
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Stephen J. Moss
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1 6BT, UK
| |
Collapse
|
2
|
Xu C, Wu JH, Yu H, Chun-Ge, Liu YX, Zou JJ, Li J. Effect of two novel GABAA receptor positive allosteric modulators on neuropathic and inflammatory pain in mice. Neuropharmacology 2025; 269:110317. [PMID: 39884570 DOI: 10.1016/j.neuropharm.2025.110317] [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: 11/25/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 02/01/2025]
Abstract
Loss of GABAergic inhibition in the spinal dorsal horn (SDH) is implicated in central sensitization and chronic pain. Both agonists and positive allosteric modulators (PAMs) of GABAA receptor are found to be effective in the management of chronic pain. In addition to benzodiazepines, neuroactive steroids (NASs) also act as PAMs through binding to unique sites of GABAA receptors. Thus, it is worth investigating whether these NASs can attenuate chronic pain. This study tested the antinociceptive properties of two novel NAS PAMs, ganaxolone and zuranolone, in segmental spinal nerve ligation (SNL)-induced neuropathic pain and complete Freund's adjuvant (CFA)-induced inflammation pain models. Spinally administered ganaxolone and zuranolone both exhibited dose-dependent analgesic effects but with quite different durations. This antinociceptive effect might be generated from elevated GABAergic inhibition, as the PAMs both enhanced GABA-evoked currents in SDH neurons, and the K+-Cl- cotransporter isoform 2 (KCC2) antagonist reversed the analgesic effect of the PAMs. Different from ganaxolone, zuranolone produced a durable increase in the surface expression of GABAA receptors and of the amplitude of spontaneous inhibitory currents, which may contribute to the long-lasting analgesic effect. Furthermore, the PAMs alleviated SNL-induced mechanical allodynia synergistically with diazepam or GABAA receptor activator muscimol at inactive doses, consistent with the non-competitive activity and distinct binding sites from benzodiazepines. In summary, our findings suggest that NASs may not only acutely modulate GABA receptor activity but also induce sustained metabotropic effects on GABAA receptors and thus exert long-lasting antinociceptive effects.
Collapse
Affiliation(s)
- Chu Xu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jian-Hong Wu
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, China
| | - Hui Yu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Chun-Ge
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yun-Xin Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jian-Jun Zou
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
3
|
Lemons AHS, Murphy B, Dengler JS, Salar S, Davies PA, Smalley JL, Moss SJ. Neuroactive steroids activate membrane progesterone receptors to induce sex specific effects on protein kinase activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634751. [PMID: 39896469 PMCID: PMC11785215 DOI: 10.1101/2025.01.24.634751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Neuroactive steroids (NAS), which are synthesized in the brain from progesterone, exert potent effects on behavior and are used to treat postpartum depression, yet how these compounds induce sustained modifications in neuronal activity are ill-defined. Here, we examined the efficacy of NAS for membrane progesterone receptors (mPRs) δ and ε, members of a family of GPCRs for progestins that are expressed in the CNS. NAS increase PKC activity via G q activation of mPRδ with EC50s between 3-11nM. In contrast, they activate G s via mPRε to potentiate PKA activity with similar potencies. NAS also induced rapid internalization of only mPRδ. In the forebrain of female mice, mPRδ expression levels were 8-fold higher than males. Consistent with this, activation of PKC by NAS was evident in acute brain slices from female mice. Collectively, our results suggests that NAS may exert sex-specific effects on intracellular signaling in the brain via activation of mPRs.
Collapse
|
4
|
Reddy DS. Neurosteroids as Novel Anticonvulsants for Refractory Status Epilepticus and Medical Countermeasures for Nerve Agents: A 15-Year Journey to Bring Ganaxolone from Bench to Clinic. J Pharmacol Exp Ther 2024; 388:273-300. [PMID: 37977814 PMCID: PMC10801762 DOI: 10.1124/jpet.123.001816] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
This article describes recent advances in the use of neurosteroids as novel anticonvulsants for refractory status epilepticus (RSE) and as medical countermeasures (MCs) for organophosphates and chemical nerve agents (OPNAs). We highlight a comprehensive 15-year journey to bring the synthetic neurosteroid ganaxolone (GX) from bench to clinic. RSE, including when caused by nerve agents, is associated with devastating morbidity and permanent long-term neurologic dysfunction. Although recent approval of benzodiazepines such as intranasal midazolam and intranasal midazolam offers improved control of acute seizures, novel anticonvulsants are needed to suppress RSE and improve neurologic function outcomes. Currently, few anticonvulsant MCs exist for victims of OPNA exposure and RSE. Standard-of-care MCs for postexposure treatment include benzodiazepines, which do not effectively prevent or mitigate seizures resulting from nerve agent intoxication, leaving an urgent unmet medical need for new anticonvulsants for RSE. Recently, we pioneered neurosteroids as next-generation anticonvulsants that are superior to benzodiazepines for treatment of OPNA intoxication and RSE. Because GX and related neurosteroids that activate extrasynaptic GABA-A receptors rapidly control seizures and offer robust neuroprotection by reducing neuronal damage and neuroinflammation, they effectively improve neurologic outcomes after acute OPNA exposure and RSE. GX has been selected for advanced, Biomedical Advanced Research and Development Authority-supported phase 3 trials of RSE and nerve agent seizures. In addition, in mechanistic studies of neurosteroids at extrasynaptic receptors, we identified novel synthetic analogs with features that are superior to GX for current medical needs. Development of new MCs for RSE is complex, tedious, and uncertain due to scientific and regulatory challenges. Thus, further research will be critical to fill key gaps in evaluating RSE and anticonvulsants in vulnerable (pediatric and geriatric) populations and military persons. SIGNIFICANCE STATEMENT: Following organophosphate and nerve agent intoxication, refractory status epilepticus (RSE) occurs despite benzodiazepine treatment. RSE occurs in 40% of status epilepticus patients, with a 35% mortality rate and significant neurological morbidity in survivors. To treat RSE, neurosteroids are better anticonvulsants than benzodiazepines. Our pioneering use of neurosteroids for RSE and nerve agents led us to develop ganaxolone as a novel anticonvulsant and neuroprotectant with significantly improved neurological outcomes. This article describes the bench-to-bedside journey of bringing neurosteroid therapy to patients, with ganaxolone leading the way.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
5
|
Maguire JL, Mennerick S. Neurosteroids: mechanistic considerations and clinical prospects. Neuropsychopharmacology 2024; 49:73-82. [PMID: 37369775 PMCID: PMC10700537 DOI: 10.1038/s41386-023-01626-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023]
Abstract
Like other classes of treatments described in this issue's section, neuroactive steroids have been studied for decades but have risen as a new class of rapid-acting, durable antidepressants with a distinct mechanism of action from previous antidepressant treatments and from other compounds covered in this issue. Neuroactive steroids are natural derivatives of progesterone but are proving effective as exogenous treatments. The best understood mechanism is that of positive allosteric modulation of GABAA receptors, where subunit selectivity may promote their profile of action. Mechanistically, there is some reason to think that neuroactive steroids may separate themselves from liabilities of other GABA modulators, although research is ongoing. It is also possible that intracellular targets, including inflammatory pathways, may be relevant to beneficial actions. Strengths and opportunities for further development include exploiting non-GABAergic targets, structural analogs, enzymatic production of natural steroids, precursor loading, and novel formulations. The molecular mechanisms of behavioral effects are not fully understood, but study of brain network states involved in emotional processing demonstrate a robust influence on affective states not evident with at least some other GABAergic drugs including benzodiazepines. Ongoing studies with neuroactive steroids will further elucidate the brain and behavioral effects of these compounds as well as likely underpinnings of disease.
Collapse
Affiliation(s)
- Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Steven Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
6
|
Pedraza N, Monserrat MV, Ferrezuelo F, Torres-Rosell J, Colomina N, Miguez-Cabello F, Párraga JP, Soto D, López-Merino E, García-Vilela C, Esteban JA, Egea J, Garí E. Cyclin D1-Cdk4 regulates neuronal activity through phosphorylation of GABAA receptors. Cell Mol Life Sci 2023; 80:280. [PMID: 37684532 PMCID: PMC10491536 DOI: 10.1007/s00018-023-04920-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 09/10/2023]
Abstract
Nuclear Cyclin D1 (Ccnd1) is a main regulator of cell cycle progression and cell proliferation. Interestingly, Ccnd1 moves to the cytoplasm at the onset of differentiation in neuronal precursors. However, cytoplasmic functions and targets of Ccnd1 in post-mitotic neurons are unknown. Here we identify the α4 subunit of gamma-aminobutyric acid (GABA) type A receptors (GABAARs) as an interactor and target of Ccnd1-Cdk4. Ccnd1 binds to an intracellular loop in α4 and, together with Cdk4, phosphorylates the α4 subunit at threonine 423 and serine 431. These modifications upregulate α4 surface levels, increasing the response of α4-containing GABAARs, measured in whole-cell patch-clamp recordings. In agreement with this role of Ccnd1-Cdk4 in neuronal signalling, inhibition of Cdk4 or expression of the non-phosphorylatable α4 decreases synaptic and extra-synaptic currents in the hippocampus of newborn rats. Moreover, according to α4 functions in synaptic pruning, CCND1 knockout mice display an altered pattern of dendritic spines that is rescued by the phosphomimetic α4. Overall, our findings molecularly link Ccnd1-Cdk4 to GABAARs activity in the central nervous system and highlight a novel role for this G1 cyclin in neuronal signalling.
Collapse
Affiliation(s)
- Neus Pedraza
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain.
| | - Ma Ventura Monserrat
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Francisco Ferrezuelo
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Jordi Torres-Rosell
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Neus Colomina
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Federico Miguez-Cabello
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Javier Picañol Párraga
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - David Soto
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Esperanza López-Merino
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia García-Vilela
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José A Esteban
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Joaquim Egea
- Molecular and Developmental Neurobiology, Dept. Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Rovira Roure 80, 25198, Lleida, Spain
| | - Eloi Garí
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain.
| |
Collapse
|
7
|
Luscher B, Maguire JL, Rudolph U, Sibille E. GABA A receptors as targets for treating affective and cognitive symptoms of depression. Trends Pharmacol Sci 2023; 44:586-600. [PMID: 37543478 PMCID: PMC10511219 DOI: 10.1016/j.tips.2023.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 08/07/2023]
Abstract
In the past 20 years, our understanding of the pathophysiology of depression has evolved from a focus on an imbalance of monoaminergic neurotransmitters to a multifactorial picture including an improved understanding of the role of glutamatergic excitatory and GABAergic inhibitory neurotransmission. FDA-approved treatments targeting the glutamatergic [esketamine for major depressive disorder (MDD)] and GABAergic (brexanolone for peripartum depression) systems have become available. This review focuses on the GABAA receptor (GABAAR) system as a target for novel antidepressants and discusses the mechanisms by which modulation of δ-containing GABAARs with neuroactive steroids (NASs) or of α5-containing GABAARs results in antidepressant or antidepressant-like actions and discusses clinical data on NASs. Moreover, a potential mechanism by which α5-GABAAR-positive allosteric modulators (PAMs) may improve cognitive deficits in depression is presented.
Collapse
Affiliation(s)
- Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA; Department of Psychiatry, Pennsylvania State University, University Park, PA 16802, USA; Penn State Neuroscience Institute, Pennsylvania State University, University Park, PA 16802, USA
| | - Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of the Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Lorenz-Guertin JM, Povysheva N, Chapman CA, MacDonald ML, Fazzari M, Nigam A, Nuwer JL, Das S, Brady ML, Vajn K, Bambino MJ, Weintraub ST, Johnson JW, Jacob TC. Inhibitory and excitatory synaptic neuroadaptations in the diazepam tolerant brain. Neurobiol Dis 2023; 185:106248. [PMID: 37536384 PMCID: PMC10578451 DOI: 10.1016/j.nbd.2023.106248] [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: 05/12/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Benzodiazepine (BZ) drugs treat seizures, anxiety, insomnia, and alcohol withdrawal by potentiating γ2 subunit containing GABA type A receptors (GABAARs). BZ clinical use is hampered by tolerance and withdrawal symptoms including heightened seizure susceptibility, panic, and sleep disturbances. Here, we investigated inhibitory GABAergic and excitatory glutamatergic plasticity in mice tolerant to benzodiazepine sedation. Repeated diazepam (DZP) treatment diminished sedative effects and decreased DZP potentiation of GABAAR synaptic currents without impacting overall synaptic inhibition. While DZP did not alter γ2-GABAAR subunit composition, there was a redistribution of extrasynaptic GABAARs to synapses, resulting in higher levels of synaptic BZ-insensitive α4-containing GABAARs and a concomitant reduction in tonic inhibition. Conversely, excitatory glutamatergic synaptic transmission was increased, and NMDAR subunits were upregulated at synaptic and total protein levels. Quantitative proteomics further revealed cortex neuroadaptations of key pro-excitatory mediators and synaptic plasticity pathways highlighted by Ca2+/calmodulin-dependent protein kinase II (CAMKII), MAPK, and PKC signaling. Thus, reduced inhibitory GABAergic tone and elevated glutamatergic neurotransmission contribute to disrupted excitation/inhibition balance and reduced BZ therapeutic power with benzodiazepine tolerance.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadya Povysheva
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Caitlyn A Chapman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew L MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aparna Nigam
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L Nuwer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sabyasachi Das
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Megan L Brady
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katarina Vajn
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew J Bambino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antoni, TX, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Rodríguez-Palma EJ, De la Luz-Cuellar YE, Islas-Espinoza AM, Félix-Leyva AE, Shiers SI, García G, Torres-Lopez JE, Delgado-Lezama R, Murbartián J, Price TJ, Granados-Soto V. Activation of α 6 -containing GABA A receptors induces antinociception under physiological and pathological conditions. Pain 2023; 164:948-966. [PMID: 36001074 PMCID: PMC9950299 DOI: 10.1097/j.pain.0000000000002763] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT The loss of GABAergic inhibition is a mechanism that underlies neuropathic pain. Therefore, rescuing the GABAergic inhibitory tone through the activation of GABA A receptors is a strategy to reduce neuropathic pain. This study was designed to elucidate the function of the spinal α 6 -containing GABA A receptor in physiological conditions and neuropathic pain in female and male rats. Results show that α 6 -containing GABA A receptor blockade or transient α 6 -containing GABA A receptor knockdown induces evoked hypersensitivity and spontaneous pain in naive female rats. The α 6 subunit is expressed in IB4 + and CGRP + primary afferent neurons in the rat spinal dorsal horn and dorsal root ganglia but not astrocytes. Nerve injury reduces α 6 subunit protein expression in the central terminals of the primary afferent neurons and dorsal root ganglia, whereas intrathecal administration of positive allosteric modulators of the α 6 -containing GABA A receptor reduces tactile allodynia and spontaneous nociceptive behaviors in female, but not male, neuropathic rats and mice. Overexpression of the spinal α 6 subunit reduces tactile allodynia and restores α 6 subunit expression in neuropathic rats. Positive allosteric modulators of the α 6 -containing GABA A receptor induces a greater antiallodynic effect in female rats and mice compared with male rats and mice. Finally, α 6 subunit is expressed in humans. This receptor is found in CGRP + and P2X3 + primary afferent fibers but not astrocytes in the human spinal dorsal horn. Our results suggest that the spinal α 6 -containing GABA A receptor has a sex-specific antinociceptive role in neuropathic pain, suggesting that this receptor may represent an interesting target to develop a novel treatment for neuropathic pain.
Collapse
Affiliation(s)
- Erick J. Rodríguez-Palma
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Yarim E. De la Luz-Cuellar
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Ana M. Islas-Espinoza
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Adalberto E. Félix-Leyva
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Stephanie I. Shiers
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Guadalupe García
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Jorge E. Torres-Lopez
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, Mexico
| | - Rodolfo Delgado-Lezama
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Zacatenco, Mexico City, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| |
Collapse
|
10
|
Strain MM, Espinoza L, Fedorchak S, Littlejohn EL, Andrade MA, Toney GM, Boychuk CR. Early central cardiovagal dysfunction after high fat diet in a murine model. Sci Rep 2023; 13:6550. [PMID: 37085567 PMCID: PMC10121716 DOI: 10.1038/s41598-023-32492-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/28/2023] [Indexed: 04/23/2023] Open
Abstract
High fat diet (HFD) promotes cardiovascular disease and blunted cardiac vagal regulation. Temporal onset of loss of cardiac vagal control and its underlying mechanism are presently unclear. We tested our hypothesis that reduced central vagal regulation occurs early after HFD and contributes to poor cardiac regulation using cardiovascular testing paired with pharmacology in mice, molecular biology, and a novel bi-transgenic mouse line. Results show HFD, compared to normal fat diet (NFD), significantly blunted cardio/pulmonary chemoreflex bradycardic responses after 15 days, extending as far as tested (> 30 days). HFD produced resting tachycardia by day 3, reflected significant loss of parasympathetic tone. No differences in bradycardic responses to graded electrical stimulation of the distal cut end of the cervical vagus indicated diet-induced differences in vagal activity were centrally mediated. In nucleus ambiguus (NA), surface expression of δ-subunit containing type A gamma-aminobutyric acid receptors (GABAA(δ)R) increased at day 15 of HFD. Novel mice lacking δ-subunit expression in vagal motor neurons (ChAT-δnull) failed to exhibit blunted reflex bradycardia or resting tachycardia after two weeks of HFD. Thus, reduced parasympathetic output contributes to early HFD-induced HR dysregulation, likely through increased GABAA(δ)Rs. Results underscore need for research on mechanisms of early onset increases in GABAA(δ)R expression and parasympathetic dysfunction after HFD.
Collapse
Affiliation(s)
- Misty M Strain
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Erica L Littlejohn
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, Mail Code 7746, San Antonio, TX, 78229-3901, USA.
| |
Collapse
|
11
|
Vien TN, Ackley MA, Doherty JJ, Moss SJ, Davies PA. Preventing Phosphorylation of the GABAAR β3 Subunit Compromises the Behavioral Effects of Neuroactive Steroids. Front Mol Neurosci 2022; 15:817996. [PMID: 35431797 PMCID: PMC9009507 DOI: 10.3389/fnmol.2022.817996] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/09/2022] [Indexed: 11/26/2022] Open
Abstract
Neuroactive steroids (NASs) have potent anxiolytic, anticonvulsant, sedative, and hypnotic actions, that reflect in part their efficacy as GABAAR positive allosteric modulators (PAM). In addition to this, NAS exert metabotropic effects on GABAergic inhibition via the activation of membrane progesterone receptors (mPRs), which are G-protein coupled receptors. mPR activation enhances the phosphorylation of residues serine 408 and 409 (S408/9) in the β3 subunit of GABAARs, increasing their accumulation in the plasma membrane leading to a sustained increase in tonic inhibition. To explore the significance of NAS-induced phosphorylation of GABAARs, we used mice in which S408/9 in the β3 subunit have been mutated to alanines, mutations that prevent the metabotropic actions of NASs on GABAAR function while preserving NAS allosteric potentiation of GABAergic current. While the sedative actions of NAS were comparable to WT, their anxiolytic actions were reduced in S408/9A mice. Although the induction of hypnosis by NAS were maintained in the mutant mice the duration of the loss of righting reflex was significantly shortened. Finally, ability of NAS to terminate diazepam pharmacoresistant seizures was abolished in S408/9A mice. In conclusion, our results suggest that S408/9 in the GABAAR β3 subunit contribute to the anxiolytic and anticonvulsant efficacy of NAS, in addition to their ability to regulate the loss of righting reflex.
Collapse
Affiliation(s)
- Thuy N. Vien
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Michael A. Ackley
- Research and Non-clinical Development, Sage Therapeutics, Inc., Cambridge, MA, United States
| | - James J. Doherty
- Research and Non-clinical Development, Sage Therapeutics, Inc., Cambridge, MA, United States
| | - Stephen J. Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Stephen J. Moss,
| | - Paul A. Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
- Paul A. Davies,
| |
Collapse
|
12
|
van der Lei MB, Kooy RF. Therapeutic potential of GABAA receptor subunit expression abnormalities in fragile X syndrome. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2021.2008168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
13
|
Li N, Tao W, Yang L, Spain WJ, Ransom CB. GABA-B receptors enhance GABA-A receptor currents by modulation of membrane trafficking in dentate gyrus granule cells. Neurosci Lett 2022; 773:136481. [DOI: 10.1016/j.neulet.2022.136481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 11/24/2022]
|
14
|
Morud J, Hardege I, Liu H, Wu T, Choi MK, Basu S, Zhang Y, Schafer WR. Deorphanization of novel biogenic amine-gated ion channels identifies a new serotonin receptor for learning. Curr Biol 2021; 31:4282-4292.e6. [PMID: 34388373 PMCID: PMC8536830 DOI: 10.1016/j.cub.2021.07.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/09/2021] [Accepted: 07/15/2021] [Indexed: 11/16/2022]
Abstract
Pentameric ligand-gated ion channels (LGICs) play conserved, critical roles in both excitatory and inhibitory synaptic transmission and can be activated by diverse neurochemical ligands. We have performed a characterization of orphan channels from the nematode C. elegans, identifying five new monoamine-gated LGICs with diverse functional properties and expression postsynaptic to aminergic neurons. These include polymodal anion channels activated by both dopamine and tyramine, which may mediate inhibitory transmission by both molecules in vivo. Intriguingly, we also find that a novel serotonin-gated cation channel, LGC-50, is essential for aversive olfactory learning of pathogenic bacteria, a process known to depend on serotonergic neurotransmission. Remarkably, the redistribution of LGC-50 to neuronal processes is modulated by olfactory conditioning, and lgc-50 point mutations that cause misregulation of receptor membrane expression interfere with olfactory learning. Thus, the intracellular trafficking and localization of these receptors at synapses may represent a molecular cornerstone of the learning mechanism.
Collapse
Affiliation(s)
- Julia Morud
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Iris Hardege
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - He Liu
- Department of Organismic and Evolutionary Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; Centre for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Taihong Wu
- Department of Organismic and Evolutionary Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; Centre for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Myung-Kyu Choi
- Department of Organismic and Evolutionary Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; Centre for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Swaraj Basu
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Medicinaregatan 9A, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; Centre for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - William R Schafer
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Biology, KU Leuven, Naamsestraat 59, 3000 Leuven, Belgium.
| |
Collapse
|
15
|
Wu K, Han W, Tian Q, Li Y, Lu W. Activity- and sleep-dependent regulation of tonic inhibition by Shisa7. Cell Rep 2021; 34:108899. [PMID: 33761345 PMCID: PMC8025742 DOI: 10.1016/j.celrep.2021.108899] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/22/2020] [Accepted: 03/03/2021] [Indexed: 02/08/2023] Open
Abstract
Tonic inhibition mediated by extrasynaptic γ-aminobutyric acid type A receptors (GABAARs) critically regulates neuronal excitability and brain function. However, the mechanisms regulating tonic inhibition remain poorly understood. Here, we report that Shisa7 is critical for tonic inhibition regulation in hippocampal neurons. In juvenile Shisa7 knockout (KO) mice, α5-GABAAR-mediated tonic currents are significantly reduced. Mechanistically, Shisa7 is crucial for α5-GABAAR exocytosis. Additionally, Shisa7 regulation of tonic inhibition requires protein kinase A (PKA) that phosphorylates Shisa7 serine 405 (S405). Importantly, tonic inhibition undergoes activity-dependent regulation, and Shisa7 is required for homeostatic potentiation of tonic inhibition. Interestingly, in young adult Shisa7 KOs, basal tonic inhibition in hippocampal neurons is unaltered, largely due to the diminished α5-GABAAR component of tonic inhibition. However, at this stage, tonic inhibition oscillates during the daily sleep/wake cycle, a process requiring Shisa7. Together, these data demonstrate that intricate signaling mechanisms regulate tonic inhibition at different developmental stages and reveal a molecular link between sleep and tonic inhibition.
Collapse
Affiliation(s)
- Kunwei Wu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qingjun Tian
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yan Li
- Proteomics Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Charsouei S, Jabalameli MR, Karimi-Moghadam A. Molecular insights into the role of AMPA receptors in the synaptic plasticity, pathogenesis and treatment of epilepsy: therapeutic potentials of perampanel and antisense oligonucleotide (ASO) technology. Acta Neurol Belg 2020; 120:531-544. [PMID: 32152997 DOI: 10.1007/s13760-020-01318-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Glutamate is considered as the predominant excitatory neurotransmitter in the mammalian central nervous systems (CNS). Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are the main glutamate-gated ionotropic channels that mediate the majority of fast synaptic excitation in the brain. AMPARs are highly dynamic that constitutively move into and out of the postsynaptic membrane. Changes in the postsynaptic number of AMPARs play a key role in controlling synaptic plasticity and also brain functions such as memory formation and forgetting development. Impairments in the regulation of AMPAR function, trafficking, and signaling pathway may also contribute to neuronal hyperexcitability and epileptogenesis process, which offers AMPAR as a potential target for epilepsy therapy. Over the last decade, various types of AMPAR antagonists such as perampanel and talampanel have been developed to treat epilepsy, but they usually show limited efficacy at low doses and produce unwanted cognitive and motor side effects when administered at higher doses. In the present article, the latest findings in the field of molecular mechanisms controlling AMPAR biology, as well as the role of these mechanism dysfunctions in generating epilepsy will be reviewed. Also, a comprehensive summary of recent findings from clinical trials with perampanel, in treating epilepsy, glioma-associated epilepsy and Parkinson's disease is provided. Finally, antisense oligonucleotide therapy as an alternative strategy for the efficient treatment of epilepsy is discussed.
Collapse
Affiliation(s)
- Saeid Charsouei
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, IR, Iran
| | - M Reza Jabalameli
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Amin Karimi-Moghadam
- Division of Genetics, Department of Biology, Faculty of Science, University of Isfahan, Isfahan, IR, Iran.
| |
Collapse
|
17
|
Kfir A, Awasthi R, Ghosh S, Kundu S, Paul B, Lamprecht R, Barkai E. A Cellular Mechanism of Learning-Induced Enhancement of Synaptic Inhibition: PKC-Dependent Upregulation of KCC2 Activation. Sci Rep 2020; 10:962. [PMID: 31969605 PMCID: PMC6976593 DOI: 10.1038/s41598-020-57626-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 12/18/2019] [Indexed: 01/23/2023] Open
Abstract
Long-term memory of complex olfactory learning is expressed by wide spread enhancement in excitatory and inhibitory synaptic transmission onto piriform cortex pyramidal neurons. A particularly interesting modification in synaptic inhibition is the hyperpolarization of the reversal potential of the fast post synaptic inhibitory potential (fIPSP). Here we study the mechanism underlying the maintenance of such a shift in the fIPSP. Blocking of the neuronal specific K+-Cl- co-transporter (KCC2) in neurons of trained rats significantly depolarized the averaged fIPSP reversal potential of the spontaneous miniature inhibitory post synaptic currents (mIPSCs), to the averaged pre-training level. A similar effect was obtained by blocking PKC, which was previously shown to upregulate KCC2. Accordingly, the level of PKC-dependent phosphorylation of KCC2, at the serine 940 site, was significantly increased after learning. In contrast, blocking two other key second messenger systems CaMKII and PKA, which have no phosphorylation sites on KCC2, had no effect on the fIPSP reversal potential. Importantly, the PKC inhibitor also reduced the averaged amplitude of the spontaneous miniature excitatory synaptic currents (mEPSCs) in neurons of trained rats only, to the pre-training level. We conclude that learning-induced hyper-polarization of the fIPSP reversal potential is mediated by PKC-dependent increase of KCC2 phosphorylation.
Collapse
Affiliation(s)
- Adi Kfir
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Richa Awasthi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Sourav Ghosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Sankhanava Kundu
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Blesson Paul
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
18
|
Parakala ML, Zhang Y, Modgil A, Chadchankar J, Vien TN, Ackley MA, Doherty JJ, Davies PA, Moss SJ. Metabotropic, but not allosteric, effects of neurosteroids on GABAergic inhibition depend on the phosphorylation of GABA A receptors. J Biol Chem 2019; 294:12220-12230. [PMID: 31239352 PMCID: PMC6690684 DOI: 10.1074/jbc.ra119.008875] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/28/2019] [Indexed: 11/06/2022] Open
Abstract
Neuroactive steroids (NASs) are synthesized within the brain and exert profound effects on behavior. These effects are primarily believed to arise from the activities of NASs as positive allosteric modulators (PAMs) of the GABA-type A receptor (GABAAR). NASs also activate a family of G protein-coupled receptors known as membrane progesterone receptors (mPRs). Here, using surface-biotinylation assays and electrophysiology techniques, we examined mPRs' role in mediating the effects of NAS on the efficacy of GABAergic inhibition. Selective mPR activation enhanced phosphorylation of Ser-408 and Ser-409 (Ser-408/9) within the GABAAR β3 subunit, which depended on the activity of cAMP-dependent protein kinase A (PKA) and protein kinase C (PKC). mPR activation did not directly modify GABAAR activity and had no acute effects on phasic or tonic inhibition. Instead, mPR activation induced a sustained elevation in tonic current, which was blocked by PKA and PKC inhibition. Substitution of Ser-408/9 to alanine residues also prevented the effects of mPR activation on tonic current. Furthermore, this substitution abolished the effects of sustained NAS exposure on tonic inhibition. Interestingly, the allosteric effects of NAS on GABAergic inhibition were independent of Ser-408/9 in the β3 subunit. Additionally, although allosteric effects of NAS on GABAergic inhibition were sensitive to a recently developed "NAS antagonist," the sustained effects of NAS on tonic inhibition were not. We conclude that metabotropic effects of NAS on GABAergic inhibition are mediated by mPR-dependent modulation of GABAAR phosphorylation. We propose that this mechanism may contribute to the varying behavioral effects of NAS.
Collapse
Affiliation(s)
- Manasa L Parakala
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Yihui Zhang
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Amit Modgil
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Jayashree Chadchankar
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Thuy N Vien
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | | | | | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111; Department of Neuroscience, Physiology, and Pharmacology, University College, London WC1E 6BT, United Kingdom.
| |
Collapse
|
19
|
O'Neill N, Sylantyev S. The Functional Role of Spontaneously Opening GABA A Receptors in Neural Transmission. Front Mol Neurosci 2019; 12:72. [PMID: 30983968 PMCID: PMC6447609 DOI: 10.3389/fnmol.2019.00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/08/2019] [Indexed: 12/22/2022] Open
Abstract
Ionotropic type of γ-aminobutyric acid receptors (GABAARs) produce two forms of inhibitory signaling: phasic inhibition generated by rapid efflux of neurotransmitter GABA into the synaptic cleft with subsequent binding to GABAARs, and tonic inhibition generated by persistent activation of extrasynaptic and/or perisynaptic GABAARs by GABA continuously present in the extracellular space. It is widely accepted that phasic and tonic GABAergic inhibition is mediated by receptor groups of distinct subunit composition and modulated by different cytoplasmic mechanisms. Recently, however, it has been demonstrated that spontaneously opening GABAARs (s-GABAARs), which do not need GABA binding to enter an active state, make a significant input into tonic inhibitory signaling. Due to GABA-independent action mode, s-GABAARs promise new safer options for therapy of neural disorders (such as epilepsy) devoid of side effects connected to abnormal fluctuations of GABA concentration in the brain. However, despite the potentially important role of s-GABAARs in neural signaling, they still remain out of focus of neuroscience studies, to a large extent due to technical difficulties in their experimental research. Here, we summarize present data on s-GABAARs functional properties and experimental approaches that allow isolation of s-GABAARs effects from those of conventional (GABA-dependent) GABAARs.
Collapse
Affiliation(s)
- Nathanael O'Neill
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sergiy Sylantyev
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Keating N, Zeak N, Smith SS. Pubertal hormones increase hippocampal expression of α4βδ GABA A receptors. Neurosci Lett 2019; 701:65-70. [PMID: 30742936 DOI: 10.1016/j.neulet.2019.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
CA1 hippocampal expression of α4βδ GABAA receptors (GABARs) increases at the onset of puberty in female mice, an effect dependent upon the decline in hippocampal levels of the neurosteroid THP (3α-OH-5α-pregnan-20-one) which occurs at this time. The present study further characterized the mechanisms underlying α4βδ expression, assessed in vivo. Blockade of pubertal levels of 17β-estradiol (E2) (formestane, 0.5 mg/kg, i.p. 3 d) reduced α4 and δ expression by 75-80% (P < 0.05) in CA1 hippocampus of female mice, assessed using Western blot techniques. Conversely, E2 administration increased α4 and δ expression by 50-100% in adults, an effect enhanced by more than 2-fold by concomitant administration of the 5α-reductase blocker finasteride (50 mg/kg, i.p., 3d, P < 0.05), suggesting that both declining THP levels and increasing E2 levels before puberty trigger α4βδ expression. This effect was blocked by ICI 182,780 (20 mg/kg, s.c., 3 d), a selective blocker of E2 receptor-α (ER-α). These results suggest that both the rise in circulating levels of E2 and the decline in hippocampal THP levels at the onset of puberty trigger maximal levels of α4βδ expression in the CA1 hippocampus.
Collapse
Affiliation(s)
- Nicole Keating
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY, 11203, USA
| | - Nicole Zeak
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY, 11203, USA
| | - Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY, 11203, USA.
| |
Collapse
|
21
|
Modgil A, Vien TN, Ackley MA, Doherty JJ, Moss SJ, Davies PA. Neuroactive Steroids Reverse Tonic Inhibitory Deficits in Fragile X Syndrome Mouse Model. Front Mol Neurosci 2019; 12:15. [PMID: 30804752 PMCID: PMC6371020 DOI: 10.3389/fnmol.2019.00015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. A reduction in neuronal inhibition mediated by γ-aminobutyric acid type A receptors (GABAARs) has been implicated in the pathophysiology of FXS. Neuroactive steroids (NASs) are known allosteric modulators of GABAAR channel function, but recent studies from our laboratory have revealed that NASs also exert persistent metabotropic effects on the efficacy of tonic inhibition by increasing the protein kinase C (PKC)-mediated phosphorylation of the α4 and β3 subunits which increase the membrane expression and boosts tonic inhibition. We have assessed the GABAergic signaling in the hippocampus of fragile X mental retardation protein (FMRP) knock-out (Fmr1KO) mouse. The GABAergic tonic current in dentate gyrus granule cells (DGGCs) from 3- to 5-week-old (p21–35) Fmr1KO mice was significantly reduced compared to WT mice. Additionally, spontaneous inhibitory post synaptic inhibitory current (sIPSC) amplitudes were increased in DGGCs from Fmr1 KO mice. While sIPSCs decay in both genotypes was prolonged by the prototypic benzodiazepine diazepam, those in Frm1-KO mice were selectively potentiated by RO15-4513. Consistent with this altered pharmacology, modifications in the expression levels and phosphorylation of receptor GABAAR subtypes that mediate tonic inhibition were seen in Fmr1 KO mice. Significantly, exposure to NASs induced a sustained elevation in tonic current in Fmr1 KO mice which was prevented with PKC inhibition. Likewise, exposure reduced elevated membrane excitability seen in the mutant mice. Collectively, our results suggest that NAS act to reverse the deficits of tonic inhibition seen in FXS, and thereby reduce aberrant neuronal hyperexcitability seen in this disorder.
Collapse
Affiliation(s)
- Amit Modgil
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Thuy N Vien
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | | | | | - Stephen J Moss
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States.,Department of Neuroscience, Physiology and Pharmacology, University College, London, United Kingdom
| | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
22
|
Szodorai E, Bampali K, Romanov RA, Kasper S, Hökfelt T, Ernst M, Lubec G, Harkany T. Diversity matters: combinatorial information coding by GABA A receptor subunits during spatial learning and its allosteric modulation. Cell Signal 2018; 50:142-159. [DOI: 10.1016/j.cellsig.2018.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 01/11/2023]
|
23
|
Chuang SH, Reddy DS. 3 β-Methyl-Neurosteroid Analogs Are Preferential Positive Allosteric Modulators and Direct Activators of Extrasynaptic δ-Subunit γ-Aminobutyric Acid Type A Receptors in the Hippocampus Dentate Gyrus Subfield. J Pharmacol Exp Ther 2018; 365:583-601. [PMID: 29602830 PMCID: PMC5941194 DOI: 10.1124/jpet.117.246660] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/28/2018] [Indexed: 02/06/2023] Open
Abstract
Neurosteroids are powerful modulators of γ-aminobutyric acid (GABA)-A receptors. Ganaxolone (3α-hydroxy-3β-methyl-5α-pregnan-20-one, GX) and synthetic analogs of the neurosteroid allopregnanolone (AP) are designed to treat epilepsy and related conditions. However, their precise mechanism of action in native neurons remains unclear. Here, we sought to determine the mode of action of GX and its analogs at GABA-A receptors in native hippocampal neurons by analyzing extrasynaptic receptor-mediated tonic currents and synaptic receptor-mediated phasic currents. Concentration-response profiles of GX were determined in two cell types: δ-containing dentate gyrus granule cells (DGGCs) and γ2-containing CA1 pyramidal cells (CA1PCs). GX produced significantly greater potentiation of the GABA-A receptor-activated chloride currents in DGGCs (500%) than CA1PCs (200%). In the absence of GABA, GX evoked 2-fold greater inward currents in DGGCs than CA1PCs, which were 2-fold greater than AP within DGGCs. In hippocampus slices, GX potentiated and directly activated tonic currents in DGGCs. These responses were significantly diminished in DGGCs from δ-subunit knockout (δKO) mice, confirming GX's selectivity for δGABA-A receptors. Like AP, GX potentiation of tonic currents was prevented by protein kinase C inhibition. Furthermore, GX's protection against hippocampus-kindled seizures was significantly diminished in δKO mice. GX analogs exhibited greater potency and efficacy than GX on δGABA-A receptor-mediated tonic inhibition. In summary, these results provide strong evidence that GX and its analogs are preferential allosteric modulators and direct activators of extrasynaptic δGABA-A receptors regulating network inhibition and seizures in the dentate gyrus. Therefore, these findings provide a mechanistic rationale for the clinical use of synthetic neurosteroids in epilepsy and seizure disorders.
Collapse
Affiliation(s)
- Shu-Hui Chuang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
24
|
Chen J, He Y, Wu Y, Zhou H, Su LD, Li WN, Olsen RW, Liang J, Zhou YD, Shen Y. Single Ethanol Withdrawal Regulates Extrasynaptic δ-GABA A Receptors Via PKCδ Activation. Front Mol Neurosci 2018; 11:141. [PMID: 29755316 PMCID: PMC5932167 DOI: 10.3389/fnmol.2018.00141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/09/2018] [Indexed: 11/23/2022] Open
Abstract
Alcohol (ethanol, EtOH) is one of the most widely abused drugs with profound effects on brain function and behavior. GABAA receptors (GABAARs) are one of the major targets for EtOH in the brain. Temporary plastic changes in GABAARs after withdrawal from a single EtOH exposure occur both in vivo and in vitro, which may be the basis for chronic EtOH addiction, tolerance and withdrawal symptoms. Extrasynaptic δ-GABAAR endocytosis is implicated in EtOH-induced GABAAR plasticity, but the mechanisms by which the relative abundance and localization of specific GABAARs are altered by EtOH exposure and withdrawal remain unclear. In this study, we investigated the mechanisms underlying rapid regulation of extrasynaptic δ-GABAAR by a single EtOH withdrawal in cultured rat hippocampal neurons. Thirty-minutes EtOH (60 mM) exposure increased extrasynaptic tonic current (Itonic) amplitude without affecting synaptic GABAAR function in neurons. In contrast, at 30 min after withdrawal, Itonic amplitude and responsiveness to acute EtOH were both reduced. Similar results occurred in neurons with okadaic acid (OA) or phorbol 12,13-dibutyrate (PDBu) exposure. Protein kinase C (PKC) inhibition prevented the reduction of Itonic amplitude and the tolerance to acute EtOH, as well as the reduction of GABAAR-δ subunit abundance induced by a single EtOH withdrawal. Moreover, EtOH withdrawal selectively increased PKCδ level, whereas PKCδ inhibition specifically rescued the EtOH-induced alterations in GABAAR-δ subunit level and δ-GABAAR function. Together, we provided strong evidence for the important roles of PKCδ in the rapid regulation of extrasynaptic δ-GABAAR induced by a single EtOH withdrawal.
Collapse
Affiliation(s)
- Juan Chen
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang He
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wu
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Zhou
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Nan Li
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Richard W Olsen
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jing Liang
- Titus Family Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Yu-Dong Zhou
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Shen
- Department of Neurobiology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
Tang Y, Sun Y, Xu R, Huang X, Gu S, Hong C, Liu M, Jiang H, Yang Y, Shi J. Arginine vasopressin differentially modulates
GABA
ergic synaptic transmission onto temperature‐sensitive and temperature‐insensitive neurons in the rat preoptic area. Eur J Neurosci 2018; 47:866-886. [DOI: 10.1111/ejn.13868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 11/27/2022]
Affiliation(s)
- Yu Tang
- Department of Neurobiology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei 430030 China
- Key Laboratory of Thermoregulatory and Inflammation of Sichuan Higher Education Institutes Chengdu Medical College Chengdu Sichuan China
- Department of Physiology Chengdu Medical College Chengdu Sichuan China
| | - Yan‐Ni Sun
- Department of Medical Laboratory Chengdu Medical College Chengdu Sichuan China
| | - Run Xu
- Department of Medical Laboratory Chengdu Medical College Chengdu Sichuan China
| | - Xiao Huang
- Department of Public Health Chengdu Medical College Chengdu Sichuan China
| | - Shuang Gu
- Department of Public Health Chengdu Medical College Chengdu Sichuan China
| | - Cheng‐Cheng Hong
- Department of Public Health Chengdu Medical College Chengdu Sichuan China
| | - Mi‐Jia Liu
- School of Clinical Medicine Chengdu Medical College Chengdu Sichuan China
| | - Heng Jiang
- Department of Medical Laboratory Chengdu Medical College Chengdu Sichuan China
| | - Yong‐Lu Yang
- Key Laboratory of Thermoregulatory and Inflammation of Sichuan Higher Education Institutes Chengdu Medical College Chengdu Sichuan China
| | - Jing Shi
- Department of Neurobiology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei 430030 China
| |
Collapse
|
26
|
Lorenz-Guertin JM, Jacob TC. GABA type a receptor trafficking and the architecture of synaptic inhibition. Dev Neurobiol 2018; 78:238-270. [PMID: 28901728 PMCID: PMC6589839 DOI: 10.1002/dneu.22536] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
Ubiquitous expression of GABA type A receptors (GABAA R) in the central nervous system establishes their central role in coordinating most aspects of neural function and development. Dysregulation of GABAergic neurotransmission manifests in a number of human health disorders and conditions that in certain cases can be alleviated by drugs targeting these receptors. Precise changes in the quantity or activity of GABAA Rs localized at the cell surface and at GABAergic postsynaptic sites directly impact the strength of inhibition. The molecular mechanisms constituting receptor trafficking to and from these compartments therefore dictate the efficacy of GABAA R function. Here we review the current understanding of how GABAA Rs traffic through biogenesis, plasma membrane transport, and degradation. Emphasis is placed on discussing novel GABAergic synaptic proteins, receptor and scaffolding post-translational modifications, activity-dependent changes in GABAA R confinement, and neuropeptide and neurosteroid mediated changes. We further highlight modern techniques currently advancing the knowledge of GABAA R trafficking and clinically relevant neurodevelopmental diseases connected to GABAergic dysfunction. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 238-270, 2018.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| |
Collapse
|
27
|
Chuang SH, Reddy DS. Genetic and Molecular Regulation of Extrasynaptic GABA-A Receptors in the Brain: Therapeutic Insights for Epilepsy. J Pharmacol Exp Ther 2018; 364:180-197. [PMID: 29142081 PMCID: PMC5771312 DOI: 10.1124/jpet.117.244673] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/13/2017] [Indexed: 12/18/2022] Open
Abstract
GABA-A receptors play a pivotal role in many brain diseases. Epilepsy is caused by acquired conditions and genetic defects in GABA receptor channels regulating neuronal excitability in the brain. The latter is referred to as GABA channelopathies. In the last two decades, major advances have been made in the genetics of epilepsy. The presence of specific GABAergic genetic abnormalities leading to some of the classic epileptic syndromes has been identified. Advances in molecular cloning and recombinant systems have helped characterize mutations in GABA-A receptor subunit genes in clinical neurology. GABA-A receptors are the prime targets for neurosteroids (NSs). However, GABA-A receptors are not static but undergo rapid changes in their number or composition in response to the neuroendocrine milieu. This review describes the recent advances in the genetic and neuroendocrine control of extrasynaptic and synaptic GABA-A receptors in epilepsy and its impact on neurologic conditions. It highlights the current knowledge of GABA genetics in epilepsy, with an emphasis on the neuroendocrine regulation of extrasynaptic GABA-A receptors in network excitability and seizure susceptibility. Recent advances in molecular regulation of extrasynaptic GABA-A receptor-mediated tonic inhibition are providing unique new therapeutic approaches for epilepsy, status epilepticus, and certain brain disorders. The discovery of an extrasynaptic molecular mechanism represents a milestone for developing novel therapies such as NS replacement therapy for catamenial epilepsy.
Collapse
Affiliation(s)
- Shu-Hui Chuang
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
28
|
Tuem KB, Atey TM. Neuroactive Steroids: Receptor Interactions and Responses. Front Neurol 2017; 8:442. [PMID: 28894435 PMCID: PMC5581316 DOI: 10.3389/fneur.2017.00442] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022] Open
Abstract
Neuroactive steroids (NASs) are naturally occurring steroids, which are synthesized centrally as de novo from cholesterol and are classified as pregnane, androstane, and sulfated neurosteroids (NSs). NASs modulate many processes via interacting with gamma-aminobutyric acid (GABA), N-methyl-d-aspartate, serotonin, voltage-gated calcium channels, voltage-dependent anion channels, α-adrenoreceptors, X-receptors of the liver, transient receptor potential channels, microtubule-associated protein 2, neurotrophin nerve growth factor, and σ1 receptors. Among these, NSs (especially allopregnanolone) have high potency and extensive GABA-A receptors and hence demonstrate anticonvulsant, anesthetic, central cytoprotectant, and baroreflex inhibitory effects. NSs are also involved in mood and learning via serotonin and anti-nociceptive activity via T-type voltage-gated Ca2+ channels. Moreover, they are modulators of mitochondrial function, synaptic plasticity, or regulators of apoptosis, which have a role in neuroprotective via voltage-dependent anion channels receptors. For proper functioning, NASs need to be in their normal level, whereas excess and deficiency may lead to abnormalities. When they are below the normal, NSs could have a part in development of depression, neuro-inflammation, multiple sclerosis, experimental autoimmune encephalitis, epilepsy, and schizophrenia. On the other hand, stress and attention deficit disorder could occur during excessive level. Overall, NASs are very important molecules with major neuropsychiatric activity.
Collapse
Affiliation(s)
- Kald Beshir Tuem
- Department of Pharmacology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Tesfay Mehari Atey
- Clinical Pharmacy Unit, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
29
|
Speigel I, Bichler EK, García PS. The Influence of Regional Distribution and Pharmacologic Specificity of GABA AR Subtype Expression on Anesthesia and Emergence. Front Syst Neurosci 2017; 11:58. [PMID: 28878632 PMCID: PMC5572268 DOI: 10.3389/fnsys.2017.00058] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/19/2017] [Indexed: 01/31/2023] Open
Abstract
Anesthetics produce unconsciousness by modulating ion channels that control neuronal excitability. Research has shown that specific GABAA receptor (GABAAR) subtypes in particular regions of the central nervous system contribute to different hyperpolarizing conductances, and behaviorally to distinct components of the anesthetized state. The expression of these receptors on the neuron cell surface, and thus the strength of inhibitory neurotransmission, is dynamically regulated by intracellular trafficking mechanisms. Pharmacologic or activity-based perturbations to these regulatory systems have been implicated in pathology of several neurological conditions, and can alter the individual response to anesthesia. Furthermore, studies are beginning to uncover how anesthetic exposure itself elicits enduring changes in subcellular physiology, including the processes that regulate ion channel trafficking. Here, we review the mechanisms that determine GABAAR surface expression, and elaborate on influences germane to anesthesia and emergence. We address known trafficking differences between the intrasynaptic receptors that mediate phasic current and the extra-synaptic receptors mediating tonic current. We also describe neurophysiologic consequences and network-level abnormalities in brain function that result from receptor trafficking aberrations. We hypothesize that the relationship between commonly used anesthetic agents and GABAAR surface expression has direct consequences on mature functioning neural networks and by extension ultimately influence the outcome of patients that undergo general anesthesia. Rational design of new anesthetics, anesthetic techniques, EEG-based monitoring strategies, or emergence treatments will need to take these effects into consideration.
Collapse
Affiliation(s)
- Iris Speigel
- Department of Anesthesiology, Emory University School of Medicine, AtlantaGA, United States.,Research Division, Atlanta Veteran's Affairs Medical Center, AtlantaGA, United States
| | - Edyta K Bichler
- Department of Anesthesiology, Emory University School of Medicine, AtlantaGA, United States.,Research Division, Atlanta Veteran's Affairs Medical Center, AtlantaGA, United States
| | - Paul S García
- Department of Anesthesiology, Emory University School of Medicine, AtlantaGA, United States.,Research Division, Atlanta Veteran's Affairs Medical Center, AtlantaGA, United States
| |
Collapse
|
30
|
Decreased surface expression of the δ subunit of the GABA A receptor contributes to reduced tonic inhibition in dentate granule cells in a mouse model of fragile X syndrome. Exp Neurol 2017; 297:168-178. [PMID: 28822839 DOI: 10.1016/j.expneurol.2017.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/21/2017] [Accepted: 08/15/2017] [Indexed: 11/22/2022]
Abstract
While numerous changes in the GABA system have been identified in models of Fragile X Syndrome (FXS), alterations in subunits of the GABAA receptors (GABAARs) that mediate tonic inhibition are particularly intriguing. Considering the key role of tonic inhibition in controlling neuronal excitability, reduced tonic inhibition could contribute to FXS-associated disorders such as hyperactivity, hypersensitivity, and increased seizure susceptibility. The current study has focused on the expression and function of the δ subunit of the GABAAR, a major subunit involved in tonic inhibition, in granule cells of the dentate gyrus in the Fmr1 knockout (KO) mouse model of FXS. Electrophysiological studies of dentate granule cells revealed a marked, nearly four-fold, decrease in tonic inhibition in the Fmr1 KO mice, as well as reduced effects of two δ subunit-preferring pharmacological agents, THIP and DS2, supporting the suggestion that δ subunit-containing GABAARs are compromised in the Fmr1 KO mice. Immunohistochemistry demonstrated a small but statistically significant decrease in δ subunit labeling in the molecular layer of the dentate gyrus in Fmr1 KO mice compared to wildtype (WT) littermates. The discrepancy between the large deficits in GABA-mediated tonic inhibition in granule cells in the Fmr1 KO mice and only modest reductions in immunolabeling of the δ subunit led to studies of surface expression of the δ subunit. Cross-linking experiments followed by Western blot analysis demonstrated a small, non-significant decrease in total δ subunit protein in the hippocampus of Fmr1 KO mice, but a four-fold decrease in surface expression of the δ subunit in these mice. No significant changes were observed in total or surface expression of the α4 subunit protein, a major partner of the δ subunit in the forebrain. Postembedding immunogold labeling for the δ subunit demonstrated a large, three-fold, decrease in the number of symmetric synapses with immunolabeling at perisynaptic locations in Fmr1 KO mice. While α4 immunogold particles were also reduced at perisynaptic locations in the Fmr1 KO mice, the labeling was increased at synaptic sites. Together these findings suggest that, in the dentate gyrus, altered surface expression of the δ subunit, rather than a decrease in δ subunit expression alone, could be limiting δ subunit-mediated tonic inhibition in this model of FXS. Finding ways to increase surface expression of the δ subunit of the GABAAR could be a novel approach to treatment of hyperexcitability-related alterations in FXS.
Collapse
|
31
|
Zhang T, Chen T, Chen P, Zhang B, Hong J, Chen L. MPTP-Induced Dopamine Depletion in Basolateral Amygdala via Decrease of D2R Activation Suppresses GABA A Receptors Expression and LTD Induction Leading to Anxiety-Like Behaviors. Front Mol Neurosci 2017; 10:247. [PMID: 28824377 PMCID: PMC5545577 DOI: 10.3389/fnmol.2017.00247] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/24/2017] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders commonly occur in Parkinson’s disease. Using field potential recording and patch-clamp recording, we evaluated influence of MPTP-reduced dopaminergic afferent in basolateral amygdala (BLA), a main region for affective regulation, on excitatory–inhibitory circuits and synaptic plasticity. Field excitatory post-synaptic potential (fEPSP) slopes at external capsule-BLA synapses were increased in MPTP-mice with decreases in paired-pulse facilitation and long-term potentiation amplitude, which were corrected by bath-application of D2R agonist quinpirole or cannabinoid type 1 receptors agonist WIN55,212-2, but not D1R agonist SKF38393. Compared to single waveform fEPSP in control mice, a multi-spike waveform fEPSP was observed in MPTP-mice with prolongation of duration and an increase in paired-pulse inhibition, which were recovered by BLA-injection of quinpirole for 2 days rather than bath-application. Density of GABA-evoked current (IGABA) in BLA principal neurons and GABAAR-α2 subunit expression were reduced in MPTP-mice, which were recovered by administration of quinpirole. Decline of PKC phosphorylation in BLA of MPTP-mice was corrected by bath-application of quinpirole, but not SKF38393. In MPTP-mice, BLA-injection of quinpirole or PKC activator PMA could recover GABAAR expression, which was sensitive to PKC inhibitor GF109203X. The impairment of long-term depression (LTD) in MPTP-mice was rescued by bath-application of GABAAR agonist muscimol or BLA-injection of quinpirole and PMA. Finally, BLA-injection of muscimol, quinpirole or PMA relieved anxiety-like behaviors in MPTP-mice. The results indicate that the MPTP-induced dopamine depletion in BLA principal neurons through reducing D2R-mediated PKC phosphorylation suppresses GABAAR expression and activity, which impairs GABAAR-mediated inhibition and LTD induction leading to anxiety-like behaviors.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Tingting Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Peipei Chen
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Baofeng Zhang
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Juan Hong
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| |
Collapse
|
32
|
Vien TN, Moss SJ, Davies PA. Regulating the Efficacy of Inhibition Through Trafficking of γ-Aminobutyric Acid Type A Receptors. Anesth Analg 2017; 123:1220-1227. [PMID: 27285004 DOI: 10.1213/ane.0000000000001349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Trafficking of anesthetic-sensitive receptors within the plasma membrane, or from one cellular component to another, occurs continuously. Changes in receptor trafficking have implications in altering anesthetic sensitivity. γ-Aminobutyric acid type A receptors (GABAARs) are anion-permeable ion channels and are the major class of receptor in the adult mammalian central nervous system that mediates inhibition. GABAergic signaling allows for precise synchronized firing of action potentials within brain circuits that is critical for cognition, behavior, and consciousness. This precision depends upon tightly controlled trafficking of GABAARs into the membrane. General anesthetics bind to and allosterically enhance GABAARs by prolonging the open state of the receptor and thereby altering neuronal and brain circuit activity. Subunit composition and GABAAR localization strongly influence anesthetic end points; therefore, changes in GABAAR trafficking could have significant consequences to anesthetic sensitivity. GABAARs are not static membrane structures but are in a constant state of flux between extrasynaptic and synaptic locations and are continually endocytosed and recycled from and to the membrane. Neuronal activity, posttranslational modifications, and some naturally occurring and synthetic compounds can influence the expression and trafficking of GABAARs. In this article, we review GABAARs, their trafficking, and how phosphorylation of GABAAR subunits can influence the surface expression and function of the receptor. Ultimately, alterations of GABAAR trafficking could modify anesthetic end points, both unintentionally through pathologic processes but potentially as a therapeutic target to adjust anesthetic-sensitive GABAARs.
Collapse
Affiliation(s)
- Thuy N Vien
- From the *Department of Neuroscience, Tufts University School of Medicine, Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts; and †Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | | | | |
Collapse
|
33
|
Nikaido Y, Furukawa T, Shimoyama S, Yamada J, Migita K, Koga K, Kushikata T, Hirota K, Kanematsu T, Hirata M, Ueno S. Propofol Anesthesia Is Reduced in Phospholipase C-Related Inactive Protein Type-1 Knockout Mice. J Pharmacol Exp Ther 2017; 361:367-374. [PMID: 28404686 DOI: 10.1124/jpet.116.239145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 11/22/2022] Open
Abstract
The GABA type A receptor (GABAA-R) is a major target of intravenous anesthetics. Phospholipase C-related inactive protein type-1 (PRIP-1) is important in GABAA-R phosphorylation and membrane trafficking. In this study, we investigated the role of PRIP-1 in general anesthetic action. The anesthetic effects of propofol, etomidate, and pentobarbital were evaluated in wild-type and PRIP-1 knockout (PRIP-1 KO) mice by measuring the latency and duration of loss of righting reflex (LORR) and loss of tail-pinch withdrawal response (LTWR). The effect of pretreatment with okadaic acid (OA), a protein phosphatase 1/2A inhibitor, on propofol- and etomidate-induced LORR was also examined. PRIP-1 deficiency provided the reduction of LORR and LTWR induced by propofol but not by etomidate or pentobarbital, indicating that PRIP-1 could determine the potency of the anesthetic action of propofol. Pretreatment with OA recovered the anesthetic potency induced by propofol in PRIP-1 KO mice. OA injection enhanced phosphorylation of cortical the GABAA-R β3 subunit in PRIP-1 KO mice. These results suggest that PRIP-1-mediated GABAA-R β3 subunit phosphorylation might be involved in the general anesthetic action induced by propofol but not by etomidate or pentobarbital.
Collapse
Affiliation(s)
- Yoshikazu Nikaido
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Tomonori Furukawa
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Shuji Shimoyama
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Junko Yamada
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Keisuke Migita
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Kohei Koga
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Tetsuya Kushikata
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Kazuyoshi Hirota
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Takashi Kanematsu
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Masato Hirata
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| | - Shinya Ueno
- Graduate School of Medicine (Y.N.), Department of Neurophysiology (Y.N., T.F., K.K., S.U.) and Department of Anesthesiology, Graduate School of Medicine (Y.N., T.Ku., K.H.), Research Center for Child Mental Development, Graduate School of Medicine (S.S., S.U.), and Department of Biomedical Sciences, Division of Medical Life Sciences, Graduate School of Health Sciences (J.Y.), Hirosaki University, Hirosaki, Japan; Department of Drug Informatics, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan (K.M.); Department of Cellular and Molecular Pharmacology, Division of Basic Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (T.Ka.); Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan (M.H.); Fukuoka Dental College, Fukuoka, Japan (M.H.)
| |
Collapse
|
34
|
Modgil A, Parakala ML, Ackley MA, Doherty JJ, Moss SJ, Davies PA. Endogenous and synthetic neuroactive steroids evoke sustained increases in the efficacy of GABAergic inhibition via a protein kinase C-dependent mechanism. Neuropharmacology 2016; 113:314-322. [PMID: 27743930 DOI: 10.1016/j.neuropharm.2016.10.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 10/03/2016] [Accepted: 10/09/2016] [Indexed: 11/16/2022]
Abstract
The neuroactive steroid (NAS) tetrahydrodeoxycorticosterone (THDOC) increases protein kinase C (PKC) mediated phosphorylation of extrasynaptic GABAA receptor (GABAAR) subunits leading to increased surface expression of α4/β3 subunit-containing extrasynaptic GABAARs, leading to a sustained increase in GABAAR tonic current density. Whether other naturally occurring and synthetic NASs share both an allosteric and metabotropic action on GABAARs is unknown. Here, we examine the allosteric and metabotropic properties of allopregnanolone (ALLO), and synthetic NASs SGE-516 and ganaxolone. ALLO, SGE-516, and ganaxolone all allosterically enhanced prototypical synaptic and extrasynaptic recombinant GABAARs. In dentate gyrus granule cells (DGGCs) all three NASs, when applied acutely, allosterically enhanced tonic and phasic GABAergic currents. In separate experiments, slices were exposed to NASs for 15 min, and then transferred to a steroid naïve recording chamber followed by ≥ 30 min wash before tonic currents were measured. A sustained increase in tonic current was observed following exposure to ALLO, or SGE-516 and was prevented by inhibiting PKC with GF 109203X. No increase in tonic current was observed with exposure to ganaxolone. In agreement with the observations of an increased tonic current, the NASs ALLO and SGE-516 increased the phosphorylation and surface expression of the β3 subunit-containing GABAARs. Our studies demonstrate that neuroactive steroids have differential abilities to induce sustained increases in the efficacy of tonic inhibition by promoting GABAAR phosphorylation and membrane trafficking dependent on PKC activity.
Collapse
Affiliation(s)
- Amit Modgil
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA
| | - Manasa L Parakala
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA
| | | | | | - Stephen J Moss
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA; Department of Neuroscience, Physiology and Pharmacology, University College, London WC1E6BT, United Kingdom
| | - Paul A Davies
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA.
| |
Collapse
|
35
|
Mele M, Leal G, Duarte CB. Role of GABAAR trafficking in the plasticity of inhibitory synapses. J Neurochem 2016; 139:997-1018. [DOI: 10.1111/jnc.13742] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Miranda Mele
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
| | - Graciano Leal
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
| | - Carlos B. Duarte
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Department of Life Sciences; University of Coimbra; Coimbra Portugal
| |
Collapse
|
36
|
Ahring PK, Bang LH, Jensen ML, Strøbæk D, Hartiadi LY, Chebib M, Absalom N. A pharmacological assessment of agonists and modulators at α4β2γ2 and α4β2δ GABA A receptors: The challenge in comparing apples with oranges. Pharmacol Res 2016; 111:563-576. [DOI: 10.1016/j.phrs.2016.05.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 01/24/2023]
|
37
|
Boychuk CR, Smith BN. Glutamatergic drive facilitates synaptic inhibition of dorsal vagal motor neurons after experimentally induced diabetes in mice. J Neurophysiol 2016; 116:1498-506. [PMID: 27385796 DOI: 10.1152/jn.00325.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/01/2016] [Indexed: 12/31/2022] Open
Abstract
The role of central regulatory circuits in modulating diabetes-associated glucose dysregulation has only recently been under rigorous investigation. One brain region of interest is the dorsal motor nucleus of the vagus (DMV), which contains preganglionic parasympathetic motor neurons that regulate subdiaphragmatic visceral function. Previous research has demonstrated that glutamatergic and GABAergic neurotransmission are independently remodeled after chronic hyperglycemia/hypoinsulinemia. However, glutamatergic circuitry within the dorsal brain stem impinges on GABAergic regulation of the DMV. The present study investigated the role of glutamatergic neurotransmission in synaptic GABAergic control of DMV neurons after streptozotocin (STZ)-induced hyperglycemia/hypoinsulinemia by using electrophysiological recordings in vitro. The frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) was elevated in DMV neurons from STZ-treated mice. The effect was abolished in the presence of the ionotropic glutamate receptor blocker kynurenic acid or the sodium channel blocker tetrodotoxin, suggesting that after STZ-induced hyperglycemia/hypoinsulinemia, increased glutamatergic receptor activity occurs at a soma-dendritic location on local GABA neurons projecting to the DMV. Although sIPSCs in DMV neurons normally demonstrated considerable amplitude variability, this variability was significantly increased after STZ-induced hyperglycemia/hypoinsulinemia. The elevated amplitude variability was not related to changes in quantal release, but rather correlated with significantly elevated frequency of sIPSCs in these mice. Taken together, these findings suggest that GABAergic regulation of central vagal circuitry responsible for the regulation of energy homeostasis undergoes complex functional reorganization after several days of hyperglycemia/hypoinsulinemia, including both glutamate-dependent and -independent forms of plasticity.
Collapse
Affiliation(s)
- Carie R Boychuk
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Bret N Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
38
|
Nakamura Y, Morrow DH, Modgil A, Huyghe D, Deeb TZ, Lumb MJ, Davies PA, Moss SJ. Proteomic Characterization of Inhibitory Synapses Using a Novel pHluorin-tagged γ-Aminobutyric Acid Receptor, Type A (GABAA), α2 Subunit Knock-in Mouse. J Biol Chem 2016; 291:12394-407. [PMID: 27044742 DOI: 10.1074/jbc.m116.724443] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Indexed: 11/06/2022] Open
Abstract
The accumulation of γ-aminobutyric acid receptors (GABAARs) at the appropriate postsynaptic sites is critical for determining the efficacy of fast inhibitory neurotransmission. Although we know that the majority of synaptic GABAAR subtypes are assembled from α1-3, β, and γ2 subunits, our understanding of how neurons facilitate their targeting to and stabilization at inhibitory synapses is rudimentary. To address these issues, we have created knock-in mice in which the pH-sensitive green fluorescent protein (GFP) and the Myc epitope were introduced to the extracellular domain of the mature receptor α2 subunit (pHα2). Using immunoaffinity purification and mass spectroscopy, we identified a stable complex of 174 proteins that were associated with pHα2, including other GABAAR subunits, and previously identified receptor-associated proteins such as gephyrin and collybistin. 149 of these proteins were novel GABAAR binding partners and included G-protein-coupled receptors and ion channel subunits, proteins that regulate trafficking and degradation, regulators of protein phosphorylation, GTPases, and a number of proteins that regulate their activity. Notably, members of the postsynaptic density family of proteins that are critical components of excitatory synapses were not associated with GABAARs. Crucially, we demonstrated for a subset of these novel proteins (including cullin1, ephexin, potassium channel tetramerization domain containing protein 12, mitofusin2, metabotropic glutamate receptor 5, p21-activated kinase 7, and Ras-related protein 5A) bind directly to the intracellular domains of GABAARs, validating our proteomic analysis. Thus, our experiments illustrate the complexity of the GABAAR proteome and enhance our understanding of the mechanisms neurons use to construct inhibitory synapses.
Collapse
Affiliation(s)
- Yasuko Nakamura
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Danielle H Morrow
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Amit Modgil
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Deborah Huyghe
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Tarek Z Deeb
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Michael J Lumb
- the Department of Neuroscience, Physiology and Pharmacology, University College, London WC1E 6BT, United Kingdom
| | - Paul A Davies
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and
| | - Stephen J Moss
- From the Department of Neuroscience, Tufts University School of Medicine, Boston Massachusetts 02111 and the Department of Neuroscience, Physiology and Pharmacology, University College, London WC1E 6BT, United Kingdom
| |
Collapse
|
39
|
Werner DF, Porcu P, Boyd KN, O'Buckley TK, Carter JM, Kumar S, Morrow AL. Ethanol-induced GABAA receptor alpha4 subunit plasticity involves phosphorylation and neuroactive steroids. Mol Cell Neurosci 2016; 72:1-8. [PMID: 26805653 DOI: 10.1016/j.mcn.2016.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 12/11/2015] [Accepted: 01/09/2016] [Indexed: 10/22/2022] Open
Abstract
GABAA receptors containing α4 subunits are widely implicated in acute ethanol sensitivity, and their spatial and temporal regulation prominently contributes to ethanol-induced neuroplasticity in hippocampus and cortex. However, it is unknown if α4-containing GABAA receptors in the thalamus, an area of high α4 expression, display similar regulatory patterns following ethanol administration, and if so, by which molecular mechanisms. In the current study, thalamic GABAA receptor α4 subunit levels were increased following a 6-week-, but not a 2-week chronic ethanol diet. Following acute high-dose ethanol administration, thalamic GABAA receptor α4 subunit levels were regulated in a temporal fashion, as a decrease was observed at 2h followed by a delayed transient increase. PKCγ and PKCδ levels paralleled α4 temporal expression patterns following ethanol exposure. Initial decreases in α4 subunit expression were associated with reduced serine phosphorylation. Delayed increases in expression were not associated with a change in phosphorylation state, but were prevented by inhibiting neuroactive steroid production with the 5α-reductase inhibitor finasteride. Overall, these studies indicate that thalamic GABAA receptor α4 subunit expression following acute and chronic ethanol administration exhibits similar regulatory patterns as other regions and that transient expression patterns following acute exposure in vivo are likely dependent on both subunit phosphorylation state and neuroactive steroids.
Collapse
Affiliation(s)
- David F Werner
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, NY 13902, USA.
| | - Patrizia Porcu
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Kevin N Boyd
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA
| | - Todd K O'Buckley
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA
| | - Jenna M Carter
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University - State University of New York, Binghamton, NY 13902, USA
| | - Sandeep Kumar
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA
| | - A Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7178, USA
| |
Collapse
|
40
|
Bohnsack JP, Carlson SL, Morrow AL. Differential regulation of synaptic and extrasynaptic α4 GABA(A) receptor populations by protein kinase A and protein kinase C in cultured cortical neurons. Neuropharmacology 2016; 105:124-132. [PMID: 26767953 DOI: 10.1016/j.neuropharm.2016.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022]
Abstract
The GABAA α4 subunit exists in two distinct populations of GABAA receptors. Synaptic GABAA α4 receptors are localized at the synapse and mediate phasic inhibitory neurotransmission, while extrasynaptic GABAA receptors are located outside of the synapse and mediate tonic inhibitory transmission. These receptors have distinct pharmacological and biophysical properties that contribute to interest in how these different subtypes are regulated under physiological and pathological states. We utilized subcellular fractionation procedures to separate these populations of receptors in order to investigate their regulation by protein kinases in cortical cultured neurons. Protein kinase A (PKA) activation decreases synaptic α4 expression while protein kinase C (PKC) activation increases α4 subunit expression, and these effects are associated with increased β3 S408/409 or γ2 S327 phosphorylation respectively. In contrast, PKA activation increases extrasynaptic α4 and δ subunit expression, while PKC activation has no effect. Our findings suggest synaptic and extrasynaptic GABAA α4 subunit expression can be modulated by PKA to inform the development of more specific therapeutics for neurological diseases that involve deficits in GABAergic transmission.
Collapse
Affiliation(s)
- John Peyton Bohnsack
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - Stephen L Carlson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - A Leslie Morrow
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA.
| |
Collapse
|
41
|
Carlson SL, Bohnsack JP, Patel V, Morrow AL. Regulation of Extrasynaptic GABAA α4 Receptors by Ethanol-Induced Protein Kinase A, but Not Protein Kinase C Activation in Cultured Rat Cerebral Cortical Neurons. J Pharmacol Exp Ther 2016; 356:148-56. [PMID: 26483396 PMCID: PMC4702069 DOI: 10.1124/jpet.115.228056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/14/2015] [Indexed: 01/14/2023] Open
Abstract
Ethanol produces changes in GABAA receptor trafficking and function that contribute to ethanol dependence symptomatology. Extrasynaptic γ-aminobutyric acid A receptors (GABAA-R) mediate inhibitory tonic current and are of particular interest because they are potentiated by physiologically relevant doses of ethanol. Here, we isolate GABAA α4δ receptors by western blotting in subsynaptic fractions to investigate protein kinase A (PKA) and protein kinase C (PKC) modulation of ethanol-induced receptor trafficking, while extrasynaptic receptor function is determined by measurement of tonic inhibition and responses evoked by 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol (THIP). Rat cerebral cortical neurons were grown for 18 days in vitro and exposed to ethanol and/or PKA/PKC modulators. Ethanol exposure (1 hour) did not alter GABAA α4 receptor abundance, but it increased tonic current amplitude, an effect that was prevented by inhibiting PKA, but not PKC. Direct activation of PKA, but not PKC, increased the abundance and tonic current of extrasynaptic α4δ receptors. In contrast, prolonged ethanol exposure (4 hours) reduced α4δ receptor abundance as well as tonic current, and this effect was also PKA dependent. Finally, PKC activation by ethanol or phorbol-12,13-dibutyrate (PdBu) had no effect on extrasynaptic α4δ subunit abundance or activity. We conclude that ethanol alters extrasynaptic α4δ receptor function and expression in cortical neurons in a PKA-dependent manner, but ethanol activation of PKC does not influence these receptors. These results could have clinical relevance for therapeutic strategies to restore normal GABAergic functioning for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Stephen L Carlson
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - J Peyton Bohnsack
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Vraj Patel
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - A Leslie Morrow
- Bowles Center for Alcohol Studies and Departments of Psychiatry and Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
42
|
Fu YL, Wang YJ, Mu TW. Proteostasis Maintenance of Cys-Loop Receptors. ION CHANNELS AS THERAPEUTIC TARGETS, PART A 2016; 103:1-23. [DOI: 10.1016/bs.apcsb.2015.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
43
|
Yang G, Ren Z, Mei YA. Exposure to 50 Hz magnetic field modulates GABAA currents in cerebellar granule neurons through an EP receptor-mediated PKC pathway. J Cell Mol Med 2015; 19:2413-22. [PMID: 26176998 PMCID: PMC4594682 DOI: 10.1111/jcmm.12626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/12/2015] [Indexed: 12/31/2022] Open
Abstract
Previous work from both our lab and others have indicated that exposure to 50 Hz magnetic fields (ELF-MF) was able to modify ion channel functions. However, very few studies have investigated the effects of MF on γ-aminobutyric acid (GABA) type A receptors (GABA(A) Rs) channel functioning, which are fundamental to overall neuronal excitability. Here, our major goal is to reveal the potential effects of ELF-MF on GABA(A) Rs activity in rat cerebellar granule neurons (CGNs). Our results indicated that exposing CGNs to 1 mT ELF-MF for 60 min. significantly increased GABA(A) R currents without modifying sensitivity to GABA. However, activation of PKA by db-cAMP failed to do so, but led to a slight decrease instead. On the other hand, PKC activation or inhibition by PMA or Bis and Docosahexaenoic acid (DHA) mimicked or eliminated the field-induced-increase of GABA(A) R currents. Western blot analysis indicated that the intracellular levels of phosphorylated PKC (pPKC) were significantly elevated after 60 min. of ELF-MF exposure, which was subsequently blocked by application of DHA or EP1 receptor-specific (prostaglandin E receptor 1) antagonist (SC19220), but not by EP2-EP4 receptor-specific antagonists. SC19220 also significantly inhibited the ELF-MF-induced elevation on GABA(A) R currents. Together, these data obviously demonstrated for the first time that neuronal GABA(A) currents are significantly increased by ELF-MF exposure, and also suggest that these effects are mediated via an EP1 receptor-mediated PKC pathway. Future work will focus on a more comprehensive analysis of the physiological and/or pathological consequences of these effects.
Collapse
Affiliation(s)
- Guang Yang
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Zhen Ren
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| | - Yan-Ai Mei
- School of Life Sciences, Institute of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China
| |
Collapse
|
44
|
Kudryashova IV. The plasticity of inhibitory synapses as a factor of long-term modifications. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415030058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Williams AD, Jung S, Poolos NP. Protein kinase C bidirectionally modulates Ih and hyperpolarization-activated cyclic nucleotide-gated (HCN) channel surface expression in hippocampal pyramidal neurons. J Physiol 2015; 593:2779-92. [PMID: 25820761 PMCID: PMC4506181 DOI: 10.1113/jp270453] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 03/25/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, particularly that of the HCN1 isoform, are enriched in the distal dendrites of hippocampal CA1 pyramidal neurons; these channels have physiological functions with respect to decreasing neuronal excitability. In the present study, we aimed to investigate phosphorylation as a mechanism controlling Ih amplitude and HCN1 surface expression in hippocampal principal neurons under normal physiological conditions. Tyrosine phosphorylation decreased Ih amplitude at maximal activation (maximal Ih ), without altering HCN1 surface expression, in two classes of hippocampal principal neurons. Inhibition of serine/threonine protein phosphatases 1 and 2A decreased maximal Ih and HCN1 surface expression in hippocampal principal neurons. Protein kinase C (PKC) activation irreversibly diminished Ih and HCN1 surface expression, whereas PKC inhibition augmented Ih and HCN1 surface expression. PKC activation increased HCN1 channel phosphorylation. These results demonstrate the novel finding of a phosphorylation mechanism, dependent on PKC activity, which bidirectionally modulates Ih amplitude and HCN1channel surface expression in hippocampal principal neurons under normal physiological conditions. ABSTRACT Hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels attenuate excitability in hippocampal pyramidal neurons. Loss of HCN channel-mediated current (Ih ), particularly that mediated by the HCN1 isoform, occurs with the development of epilepsy. Previously, we showed that, following pilocarpine-induced status epilepticus, there are two independent changes in HCN function in dendrites: decreased Ih amplitude associated with a loss of HCN1 surface expression and a hyperpolarizing shift in voltage-dependence of activation (gating). The hyperpolarizing shift in gating was attributed to decreased phosphorylation as a result of a loss of p38 mitogen-activated protein kinase activity and increased calcineurin activity; however, the mechanisms controlling Ih amplitude and HCN1 surface expression under epileptic or normal physiological conditions are poorly understood. We aimed to investigate phosphorylation as a mechanism regulating Ih amplitude and HCN1 surface expression (i.e. as is the case for HCN gating) in hippocampal principal neurons under normal physiological conditions. We discovered that inhibition of either tyrosine phosphatases or the serine/threonine protein phosphatases 1 and 2A decreased Ih at maximal activation in hippocampal CA1 pyramidal dendrites and pyramidal-like principal neuron somata from naïve rats. Furthermore, we found that inhibition of PP1/PP2A decreased HCN1 surface expression, whereas tyrosine phosphatase inhibition did not. Protein kinase C (PKC) activation reduced Ih amplitude and HCN1 surface expression, whereas PKC inhibition produced the opposite effect. Inhibition of protein phosphatases 1 and 2A and activation of PKC increased the serine phosphorylation state of the HCN1 protein. The effect of PKC activation on Ih was irreversible. These results indicate that PKC bidirectionally modulates Ih amplitude and HCN1 surface expression in hippocampal principal neurons.
Collapse
Affiliation(s)
- Aaron D Williams
- Department of Physiology and Biophysics, University of Washington
| | - Sangwook Jung
- Department of Physiology and Biophysics, University of Washington
| | - Nicholas P Poolos
- Department of Physiology and Biophysics, University of Washington
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, USA
| |
Collapse
|
46
|
Boychuk CR, Halmos KC, Smith BN. Diabetes induces GABA receptor plasticity in murine vagal motor neurons. J Neurophysiol 2015; 114:698-706. [PMID: 25995347 DOI: 10.1152/jn.00209.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/19/2015] [Indexed: 01/07/2023] Open
Abstract
Autonomic dysregulation accompanies type-1 diabetes, and synaptic regulation of parasympathetic preganglionic motor neurons in the dorsal motor nucleus of the vagus (DMV) is altered after chronic hyperglycemia/hypoinsulinemia. Tonic gamma-aminobutyric acid A (GABAA) inhibition prominently regulates DMV neuron activity, which contributes to autonomic control of energy homeostasis. This study investigated persistent effects of chronic hyperglycemia/hypoinsulinemia on GABAA receptor-mediated inhibition in the DMV after streptozotocin-induced type-1 diabetes using electrophysiological recordings in vitro, quantitative (q)RT-PCR, and immunohistochemistry. Application of the nonspecific GABAA receptor agonist muscimol evoked an outward current of significantly larger amplitude in DMV neurons from diabetic mice than controls. Results from application of 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol hydrochloride (THIP), a δ-subunit agonist, suggested that GABAA receptors containing δ-subunits contributed to the enhanced inducible tonic GABA current in diabetic mice. Sensitivity to THIP of inhibitory postsynaptic currents in DMV neurons from diabetic mice was also increased. Results from qRT-PCR and immunohistochemical analyses indicated that the altered GABAergic inhibition may be related to increased trafficking of GABAA receptors that contain the δ-subunit, rather than an expression change. Overall these findings suggest increased sensitivity of δ-subunit containing GABAA receptors after several days of hyperglycemia/hypoinsulinemia, which dramatically alters GABAergic inhibition of DMV neurons and could contribute to diabetic autonomic dysregulation.
Collapse
Affiliation(s)
- C R Boychuk
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - K Cs Halmos
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - B N Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
47
|
Chen XQ, Zhu JX, Wang Y, Zhang X, Bao L. CaMKIIα and caveolin-1 cooperate to drive ATP-induced membrane delivery of the P2X3 receptor. J Mol Cell Biol 2015; 6:140-53. [PMID: 24755854 DOI: 10.1093/jmcb/mju011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The P2X3 receptor plays a vital role in sensory processing and transmission. The assembly and trafficking of the P2X3 receptor are important for its function in primary sensory neurons. As an important inflammation mediator, ATP is released from different cell types around primary sensory neurons, especially under pathological pain conditions. Here, we show that α, β-MeATP dramatically promoted membrane delivery of the P2X3 receptor both in HEK293T cells expressing recombinant P2X3 receptor and in rat primary sensory neurons. α, β-MeATP induced P2X3 receptor-mediated Ca²⁺ influx, which further activated Ca²⁺/calmodulin-dependent protein kinase IIα (CaMKIIα). The N terminus of the P2X3 receptor was responsible for CaMKIIα binding, whereas Thr³⁸⁸ in the C terminus was phosphorylated by CaMKIIα. Thr³⁸⁸ phosphorylation increased P2X3 receptor binding to caveolin-1. Caveolin-1 knockdown abrogated the α, β-MeATP-induced membrane insertion of the P2X3 receptor. Moreover, α, β-MeATP drove the CaMKIIα-mediated membrane coinsertion of the P2X2 receptor with the P2X3 receptor. The increased P2X3 receptors on the cell membrane that are due to Thr³⁸⁸ phosphorylation facilitated P2X3 receptor-mediated signal transduction. Together, our data indicate that CaMKIIα and caveolin-1 cooperate to drive ligand-induced membrane delivery of the P2X3 receptor and may provide a mechanism of P2X3 receptor sensitization in pain development.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
48
|
Persistent CaMKII activation mediates learning-induced long-lasting enhancement of synaptic inhibition. J Neurosci 2015; 35:128-39. [PMID: 25568108 DOI: 10.1523/jneurosci.2123-14.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Training rats in a particularly difficult olfactory-discrimination task results in acquisition of high skill to perform the task superbly, termed "rule learning" or "learning set." Such complex learning results in enhanced intrinsic neuronal excitability of piriform cortex pyramidal neurons, and in their excitatory synaptic interconnections. These changes, while subserving memory maintenance, must be counterbalanced by modifications that prevent overspreading of activity and uncontrolled synaptic strengthening. Indeed, we have previously shown that the average amplitude of GABAA-mediated miniature IPSCs (mIPSCs) in these neurons is enhanced for several days after learning, an enhancement mediated via a postsynaptic mechanism. To unravel the molecular mechanism of this long-term inhibition enhancement, we tested the role of key second-messenger systems in maintaining such long-lasting modulation. The calcium/calmodulin-dependent kinase II (CaMKII) blocker, KN93, significantly reduced the average mIPSC amplitude in neurons from trained rats only to the average pretraining level. A similar effect was obtained by the CaMKII peptide inhibitor, tatCN21. Such reduction resulted from decreased single-channel conductance and not in the number of activated channels. The PKC inhibitor, GF109203X, reduced the average mIPSC amplitude in neurons from naive, pseudo-trained, and trained animals, and the difference between the trained and control groups remained. Such reduction resulted from a decrease in the number of activated channels. The PKA inhibitor H89 dihydrochloride did not affect the average mIPSC amplitude in neurons from any of the three groups. We conclude that learning-induced enhancement of GABAA-mediated synaptic inhibition is maintained by persistent CaMKII activation.
Collapse
|
49
|
Nakamura Y, Darnieder LM, Deeb TZ, Moss SJ. Regulation of GABAARs by phosphorylation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 72:97-146. [PMID: 25600368 PMCID: PMC5337123 DOI: 10.1016/bs.apha.2014.11.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are the principal mediators of fast synaptic inhibition in the brain as well as the low persistent extrasynaptic inhibition, both of which are fundamental to proper brain function. Thus unsurprisingly, deficits in GABAARs are implicated in a number of neurological disorders and diseases. The complexity of GABAAR regulation is determined not only by the heterogeneity of these receptors but also by its posttranslational modifications, the foremost, and best characterized of which is phosphorylation. This review will explore the details of this dynamic process, our understanding of which has barely scratched the surface. GABAARs are regulated by a number of kinases and phosphatases, and its phosphorylation plays an important role in governing its trafficking, expression, and interaction partners. Here, we summarize the progress in understanding the role phosphorylation plays in the regulation of GABAARs. This includes how phosphorylation can affect the allosteric modulation of GABAARs, as well as signaling pathways that affect GABAAR phosphorylation. Finally, we discuss the dysregulation of GABAAR phosphorylation and its implication in disease processes.
Collapse
|
50
|
Adams JM, Thomas P, Smart TG. Modulation of neurosteroid potentiation by protein kinases at synaptic- and extrasynaptic-type GABAA receptors. Neuropharmacology 2015; 88:63-73. [PMID: 25278033 PMCID: PMC4239298 DOI: 10.1016/j.neuropharm.2014.09.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/08/2014] [Accepted: 09/18/2014] [Indexed: 01/21/2023]
Abstract
GABAA receptors are important for inhibition in the CNS where neurosteroids and protein kinases are potent endogenous modulators. Acting individually, these can either enhance or depress receptor function, dependent upon the type of neurosteroid or kinase and the receptor subunit combination. However, in vivo, these modulators probably act in concert to fine-tune GABAA receptor activity and thus inhibition, although how this is achieved remains unclear. Therefore, we investigated the relationship between these modulators at synaptic-type α1β3γ2L and extrasynaptic-type α4β3δ GABAA receptors using electrophysiology. For α1β3γ2L, potentiation of GABA responses by tetrahydro-deoxycorticosterone was reduced after inhibiting protein kinase C, and enhanced following its activation, suggesting this kinase regulates neurosteroid modulation. In comparison, neurosteroid potentiation was reduced at α1β3(S408A,S409A)γ2L receptors, and unaltered by PKC inhibitors or activators, indicating that phosphorylation of β3 subunits is important for regulating neurosteroid activity. To determine whether extrasynaptic-type GABAA receptors were similarly modulated, α4β3δ and α4β3(S408A,S409A)δ receptors were investigated. Neurosteroid potentiation was reduced at both receptors by the kinase inhibitor staurosporine. By contrast, neurosteroid-mediated potentiation at α4(S443A)β3(S408A,S409A)δ receptors was unaffected by protein kinase inhibition, strongly suggesting that phosphorylation of α4 and β3 subunits is required for regulating neurosteroid activity at extrasynaptic receptors. Western blot analyses revealed that neurosteroids increased phosphorylation of β3(S408,S409) implying that a reciprocal pathway exists for neurosteroids to modulate phosphorylation of GABAA receptors. Overall, these findings provide important insight into the regulation of GABAA receptors in vivo, and into the mechanisms by which GABAergic inhibitory transmission may be simultaneously tuned by two endogenous neuromodulators.
Collapse
Affiliation(s)
- Joanna M Adams
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Philip Thomas
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Trevor G Smart
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|