1
|
Tubiolo PN, Williams JC, Gil RB, Cassidy C, Haubold NK, Patel Y, Abeykoon SK, Zheng ZJ, Pham DT, Ojeil N, Bobchin K, Silver-Frankel EB, Perlman G, Weinstein JJ, Kellendonk C, Horga G, Slifstein M, Abi-Dargham A, Van Snellenberg JX. Translational Evidence for Dopaminergic Rewiring of the Basal Ganglia in Persons with Schizophrenia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.31.25324962. [PMID: 40236399 PMCID: PMC11998822 DOI: 10.1101/2025.03.31.25324962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Importance In prior work, a transgenic mouse model of the striatal dopamine dysfunction observed in persons with schizophrenia (PSZ) exhibited dopamine-related neuroplasticity in the basal ganglia. This phenotype has never been demonstrated in human PSZ. Objective To identify a specific dopamine-related alteration of basal ganglia connectivity via task-based and resting-state functional magnetic resonance imaging (fMRI), neuromelanin-sensitive MRI (NM-MRI), and positron emission tomography (PET), in unmedicated PSZ. Design This case-control study of unmedicated PSZ and healthy controls (HC) occurred between November 2014 and June 2018, with analyses performed between April 2023 and February 2025. Setting fMRI and NM-MRI were collected at New York State Psychiatric Institute. [11C]-(+)-PHNO PET was collected at Yale University. Participants Participants were aged 18-55, and demographically matched. PSZ were antipsychotic drug-naïve or drug-free for at least three weeks prior to recruitment. Main Outcomes and Measures 1) task-state and resting-state functional connectivity (FC) between dorsal caudate (DCa) and globus pallidus externus (GPe), 2) NM-MRI contrast ratio in substantia nigra voxels associated with psychotic symptom severity, and 3) baseline and amphetamine-induced change in [11C]-(+)-PHNO binding potential in DCa. Results 37 PSZ (mean±SD age, 32.7±12.7 years, 29.7% female) and 30 HC (32.5±9.7 years, 26.7% female) underwent resting-state fMRI; 29 PSZ (33.4±12.7 years, 31% female) and 29 HC (32.4±9.7 years, 31% female) underwent working memory task-based fMRI. 22 PSZ (35.1±13.9 years, 36.4% female) and 20 HC (29.4±8.5 years, 35% female) underwent NM-MRI. 7 PSZ (23.1±6.3 years, 57.1% female) and 4 HC (31.5±11.9 years, 25% female) underwent [11C]-(+)-PHNO PET with amphetamine challenge. PSZ displayed elevated task-state FC (0.11±0.10 versus 0.05±0.09 in HC; P=0.0252), which was associated with increased NM-MRI contrast ratio (β* [SE] = 0.40 [0.17]; P=0.023), decreased baseline D2 receptor availability (β* [SE] = -0.45 [0.17]; P=0.039), greater amphetamine-induced dopamine release (β* [SE] = -0.82 [0.27]; P=0.021), and worse task performance (β* [SE] = -0.31 [0.13]; P=0.020). Conclusions and Relevance This study provides in-vivo evidence of a dopamine-associated neural abnormality of DCa and GPe connectivity in unmedicated PSZ. This phenotype suggests a potential neurodevelopmental mechanism of working memory deficits in schizophrenia, representing a critical step towards developing treatments for cognitive deficits.
Collapse
Affiliation(s)
- Philip N. Tubiolo
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Scholars in BioMedical Sciences Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - John C. Williams
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Roberto B. Gil
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
| | - Clifford Cassidy
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Natalka K. Haubold
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Yash Patel
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Sameera K. Abeykoon
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Zu Jie Zheng
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- College of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203
| | - Dathy T. Pham
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853
| | - Najate Ojeil
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
| | - Kelly Bobchin
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Eilon B. Silver-Frankel
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Greg Perlman
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Jodi J. Weinstein
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
| | - Christoph Kellendonk
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
- Department of Molecular Pharmacology & Therapeutics, Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
| | - Guillermo Horga
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
| | - Mark Slifstein
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
| | - Anissa Abi-Dargham
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
- Department of Radiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
| | - Jared X. Van Snellenberg
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian / Columbia University Irving Medical Center, New York, NY 10032
- New York State Psychiatric Institute, New York, NY 10032
- Department of Psychology, Stony Brook University, Stony Brook, NY 11794
| |
Collapse
|
2
|
Fraser SD, Harvey RJ. The emerging role of glycine receptor α2 subunit defects in neurodevelopmental disorders. Front Mol Neurosci 2025; 18:1550863. [PMID: 40007572 PMCID: PMC11850347 DOI: 10.3389/fnmol.2025.1550863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Rare neurodevelopmental disorders (NDDs) are one of the most significant unmet challenges in healthcare due to their lifelong nature, high management costs, and recurrence within families. This review will focus on newly-emerging genetic forms of NDDs resulting from variants in the glycine receptor (GlyR) α2 subunit gene. Studies using Glra2 knockout mice have convincingly demonstrated that GlyR α2 is essential for cortical interneuron migration and progenitor homeostasis. Genetic inactivation of GlyR α2 impairs the capacity of apical progenitors to generate basal progenitors, resulting in an overall reduction of projection neurons in the cerebral cortex. As a result, microcephaly is observed in newborn Glra2 knockout mice, as well as defects in neuronal morphology, increased susceptibility to seizures, and defects in novel object recognition, motor memory consolidation, righting reflexes, novelty-induced locomotion in the open field test, and motivational reward tasks. Consistent with these findings, we and others have identified missense variants and microdeletions in the human GlyR α2 subunit gene (GLRA2) in individuals with autism spectrum disorder (ASD), developmental delay (DD) and/or intellectual disability (ID), often accompanied by microcephaly, language delay and epilepsy. In this review, we highlight the critical role of the GlyR α2 subunit revealed by knockout mice and our current understanding of GlyR α2 pathomechanisms in human NDDs. Finally, we will consider the current gaps in our knowledge, which include: (i) Limited functional validation for GlyR α2 missense variants associated with human NDDs; (ii) The lack of gain-of-function GlyR α2 mouse models; (iii) Our limited knowledge of GlyR α2 interacting proteins. We also highlight potential future developments in the field, including routes to personalized medicines for individuals with GlyR α2 mutations.
Collapse
Affiliation(s)
- Sean D. Fraser
- School of Health, University of the Sunshine Coast, Maroochydore, QLD, Australia
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
| | - Robert J. Harvey
- School of Health, University of the Sunshine Coast, Maroochydore, QLD, Australia
- National PTSD Research Centre, Thompson Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
| |
Collapse
|
3
|
Ceder MM, Magnusson KA, Weman HM, Henriksson K, Andréasson L, Lindström T, Wiggins O, Lagerström MC. The mRNA expression profile of glycine receptor subunits alpha 1, alpha 2, alpha 4 and beta in female and male mice. Mol Cell Neurosci 2024; 131:103976. [PMID: 39580061 DOI: 10.1016/j.mcn.2024.103976] [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: 04/25/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 11/25/2024] Open
Abstract
Glycine receptors are ligand-gated chloride-selective channels that control excitability in the central nervous system (CNS). Herein, we have investigated the mRNA expression of the glycine receptor alpha 1 (Glra1), alpha 2 (Glra2), alpha 4 (Glra4) and the beta (Glrb) subunits, in adult female and male mice. Single-cell RNA sequencing data re-analysis of the Zeisel et al. (2018) dataset indicated widespread expression of Glra1, Glra2 and Glrb in the CNS, while only a few cells in the cortex, striatum, thalamus, midbrain and the spinal cord expressed Glra4. Highest occurrence of Glra1, Glra2 and Glrb were found in the brainstem. Moreover, Glra1 and Glrb were revealed to have the highest occurrences in the spinal cord of the investigated subunits. However, both Glra2 and Glrb had a more widespread expression in the CNS compared with Glra1 and Glra4. Bulk quantitative real-time-PCR (qRT-PCR) analysis revealed Glra1 expression in the hypothalamus, thalamus, brainstem and the spinal cord, and widespread, but low, Glra2 and Glrb expression in the CNS. Moreover, Glrb could be detected in a few visceral organs. Additionally, females and males were found to express Glra1, Glra2 and Glrb differently in certain brain areas such as the brainstem. Expression levels of Glra4 were too low to be detected using qRT-PCR. Lastly, RNAscope spatially validated the expression of Glra1, Glra2 and Glrb in the areas indicated by the single-cell and bulk analyses, and further revealed that Glra4 can be detected in the cortex, amygdala, hypothalamus, thalamus, brainstem, especially the cochlear nucleus, and in the spinal cord.
Collapse
Affiliation(s)
- Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hannah M Weman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linn Andréasson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Teresa Lindström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Wiggins
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Armijo-Weingart L, San Martin L, Gallegos S, Araya A, Konar-Nie M, Fernandez-Pérez E, Aguayo LG. Loss of glycine receptors in the nucleus accumbens and ethanol reward in an Alzheimer´s Disease mouse model. Prog Neurobiol 2024; 237:102616. [PMID: 38723884 PMCID: PMC11163974 DOI: 10.1016/j.pneurobio.2024.102616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Alterations in cognitive and non-cognitive cerebral functions characterize Alzheimer's disease (AD). Cortical and hippocampal impairments related to extracellular accumulation of Aβ in AD animal models have been extensively investigated. However, recent reports have also implicated intracellular Aβ in limbic regions, such as the nucleus accumbens (nAc). Accumbal neurons express high levels of inhibitory glycine receptors (GlyRs) that are allosterically modulated by ethanol and have a role in controlling its intake. In the present study, we investigated how GlyRs in the 2xTg mice (AD model) affect nAc functions and ethanol intake behavior. Using transgenic and control aged-matched litter mates, we found that the GlyRα2 subunit was significantly decreased in AD mice (6-month-old). We also examined intracellular calcium dynamics using the fluorescent calcium protein reporter GCaMP in slice photometry. We also found that the calcium signal mediated by GlyRs, but not GABAAR, was also reduced in AD neurons. Additionally, ethanol potentiation was significantly decreased in accumbal neurons in the AD mice. Finally, we performed drinking in the dark (DID) experiments and found that 2xTg mice consumed less ethanol on the last day of DID, in agreement with a lower blood ethanol concentration. 2xTg mice also showed lower sucrose consumption, indicating that overall food reward was altered. In conclusion, the data support the role of GlyRs in nAc neuron excitability and a decreased glycinergic activity in the 2xTg mice that might lead to impairment in reward processing at an early stage of the disease.
Collapse
Affiliation(s)
- Lorena Armijo-Weingart
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Loreto San Martin
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Scarlet Gallegos
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Anibal Araya
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Macarena Konar-Nie
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Eduardo Fernandez-Pérez
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile.
| |
Collapse
|
5
|
Devoght J, Comhair J, Morelli G, Rigo JM, D'Hooge R, Touma C, Palme R, Dewachter I, vandeVen M, Harvey RJ, Schiffmann SN, Piccart E, Brône B. Dopamine-mediated striatal activity and function is enhanced in GlyRα2 knockout animals. iScience 2023; 26:107400. [PMID: 37554441 PMCID: PMC10404725 DOI: 10.1016/j.isci.2023.107400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/27/2023] [Accepted: 07/12/2023] [Indexed: 08/10/2023] Open
Abstract
The glycine receptor alpha 2 (GlyRα2) is a ligand-gated ion channel which upon activation induces a chloride conductance. Here, we investigated the role of GlyRα2 in dopamine-stimulated striatal cell activity and behavior. We show that depletion of GlyRα2 enhances dopamine-induced increases in the activity of putative dopamine D1 receptor-expressing striatal projection neurons, but does not alter midbrain dopamine neuron activity. We next show that the locomotor response to d-amphetamine is enhanced in GlyRα2 knockout animals, and that this increase correlates with c-fos expression in the dorsal striatum. 3-D modeling revealed an increase in the neuronal ensemble size in the striatum in response to D-amphetamine in GlyRα2 KO mice. Finally, we show enhanced appetitive conditioning in GlyRα2 KO animals that is likely due to increased motivation, but not changes in associative learning or hedonic response. Taken together, we show that GlyRα2 is an important regulator of dopamine-stimulated striatal activity and function.
Collapse
Affiliation(s)
- Jens Devoght
- Department of Neuroscience, UHasselt, 3500 Hasselt, Belgium
| | - Joris Comhair
- Department of Neuroscience, UHasselt, 3500 Hasselt, Belgium
| | - Giovanni Morelli
- Brain Development and Disease Laboratory, Instituto Italiano di Tecnologia, 16163 Genova, Italy
| | | | - Rudi D'Hooge
- Laboratory for Biological Psychology, University of Leuven, 3000 Leuven, Belgium
| | - Chadi Touma
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Rupert Palme
- Institute of Biochemistry, University of Veterinary Medicine Vienna, Vienna A-1210, Austria
| | - Ilse Dewachter
- Department of Neuroscience, UHasselt, 3500 Hasselt, Belgium
| | | | - Robert J. Harvey
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Serge N. Schiffmann
- Laboratory of Neurophysiology, Université libre de Bruxelles, 1070 Brussels, Belgium
| | | | - Bert Brône
- Department of Neuroscience, UHasselt, 3500 Hasselt, Belgium
| |
Collapse
|
6
|
de Carvalho G, Khoja S, Haile MT, Chen LY. Early life adversity impaired dorsal striatal synaptic transmission and behavioral adaptability to appropriate action selection in a sex-dependent manner. Front Synaptic Neurosci 2023; 15:1128640. [PMID: 37091877 PMCID: PMC10116150 DOI: 10.3389/fnsyn.2023.1128640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/10/2023] [Indexed: 04/25/2023] Open
Abstract
Early life adversity (ELA) is a major health burden in the United States, with 62% of adults reporting at least one adverse childhood experience. These experiences during critical stages of brain development can perturb the development of neural circuits that mediate sensory cue processing and behavioral regulation. Recent studies have reported that ELA impaired the maturation of dendritic spines on neurons in the dorsolateral striatum (DLS) but not in the dorsomedial striatum (DMS). The DMS and DLS are part of two distinct corticostriatal circuits that have been extensively implicated in behavioral flexibility by regulating and integrating action selection with the reward value of those actions. To date, no studies have investigated the multifaceted effects of ELA on aspects of behavioral flexibility that require alternating between different action selection strategies or higher-order cognitive processes, and the underlying synaptic transmission in corticostriatal circuitries. To address this, we employed whole-cell patch-clamp electrophysiology to assess the effects of ELA on synaptic transmission in the DMS and DLS. We also investigated the effects of ELA on the ability to update action control in response to outcome devaluation in an instrumental learning paradigm and reversal of action-outcome contingency in a water T-maze paradigm. At the circuit level, ELA decreased corticostriatal glutamate transmission in male but not in female mice. Interestingly, in DMS, glutamate transmission is decreased in male ELA mice, but increased in female ELA mice. ELA impaired the ability to update action control in response to reward devaluation in a context that promotes goal-directedness in male mice and induced deficits in reversal learning. Overall, our findings demonstrate the sex- and region-dependent effects of ELA on behavioral flexibility and underlying corticostriatal glutamate transmission. By establishing a link between ELA and circuit mechanisms underlying behavioral flexibility, our findings will begin to identify novel molecular mechanisms that can represent strategies for treating behavioral inflexibility in individuals who experienced early life traumatic incidents.
Collapse
Affiliation(s)
- Gregory de Carvalho
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Sheraz Khoja
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Mulatwa T Haile
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Lulu Y Chen
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, United States
- UCI-Conte Center, UCI-NIMH, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
7
|
Oliva CA, Stehberg J, Barra R, Mariqueo T. Neuropathic Pain Induces Interleukin-1β Sensitive Bimodal Glycinergic Activity in the Central Amygdala. Int J Mol Sci 2022; 23:ijms23137356. [PMID: 35806360 PMCID: PMC9266833 DOI: 10.3390/ijms23137356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Neuropathic pain reduces GABA and glycine receptor (GlyR)-mediated activity in spinal and supraspinal regions associated with pain processing. Interleukin-1β (IL-1β) alters Central Amygdala (CeA) excitability by reducing glycinergic inhibition in a mechanism that involves the auxiliary β-subunit of GlyR (βGlyR), which is highly expressed in this region. However, GlyR activity and its modulation by IL-1β in supraspinal brain regions under neuropathic pain have not been studied. We performed chronic constriction injury (CCI) of the sciatic nerve in male Sprague Dawley rats, a procedure that induces hind paw plantar hyperalgesia and neuropathic pain. Ten days later, the rats were euthanized, and their brains were sliced. Glycinergic spontaneous inhibitory currents (sIPSCs) were recorded in the CeA slices. The sIPSCs from CeA neurons of CCI animals show a bimodal amplitude distribution, different from the normal distribution in Sham animals, with small and large amplitudes of similar decay constants. The perfusion of IL-1β (10 ng/mL) in these slices reduced the amplitudes within the first five minutes, with a pronounced effect on the largest amplitudes. Our data support a possible role for CeA GlyRs in pain processing and in the neuroimmune modulation of pain perception.
Collapse
Affiliation(s)
- Carolina A. Oliva
- Facultad de Educación, Universidad de Las Américas, República 71, Santiago 8370040, Chile
- Correspondence: (C.A.O.); (T.M.)
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Rafael Barra
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Av. Libertador Bernardo O’Higgins 3677, Santiago 8320000, Chile;
| | - Trinidad Mariqueo
- Laboratorio de Neurofarmacología, Centro de Investigaciones Médicas, Facultad de Medicina, Universidad de Talca, Av. Lircay S/N, Talca 3460000, Chile
- Correspondence: (C.A.O.); (T.M.)
| |
Collapse
|
8
|
Chen X, Wilson KA, Schaefer N, De Hayr L, Windsor M, Scalais E, van Rijckevorsel G, Stouffs K, Villmann C, O’Mara ML, Lynch JW, Harvey RJ. Loss, Gain and Altered Function of GlyR α2 Subunit Mutations in Neurodevelopmental Disorders. Front Mol Neurosci 2022; 15:886729. [PMID: 35571374 PMCID: PMC9103196 DOI: 10.3389/fnmol.2022.886729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022] Open
Abstract
Glycine receptors (GlyRs) containing the α2 subunit govern cell fate, neuronal migration and synaptogenesis in the developing cortex and spinal cord. Rare missense variants and microdeletions in the X-linked GlyR α2 subunit gene (GLRA2) have been associated with human autism spectrum disorder (ASD), where they typically cause a loss-of-function via protein truncation, reduced cell-surface trafficking and/or reduced glycine sensitivity (e.g., GLRA2Δex8-9 and extracellular domain variants p.N109S and p.R126Q). However, the GlyR α2 missense variant p.R323L in the intracellular M3-M4 domain results in a gain-of-function characterized by slower synaptic decay times, longer duration active periods and increases in channel conductance. This study reports the functional characterization of four missense variants in GLRA2 associated with ASD or developmental disorders (p.V-22L, p.N38K, p.K213E, p.T269M) using a combination of bioinformatics, molecular dynamics simulations, cellular models of GlyR trafficking and electrophysiology in artificial synapses. The GlyR α2V–22L variant resulted in altered predicted signal peptide cleavage and a reduction in cell-surface expression, suggestive of a partial loss-of-function. Similarly, GlyR α2N38K homomers showed reduced cell-surface expression, a reduced affinity for glycine and a reduced magnitude of IPSCs in artificial synapses. By contrast, GlyR α2K213E homomers showed a slight reduction in cell-surface expression, but IPSCs were larger, with faster rise/decay times, suggesting a gain-of-function. Lastly, GlyR α2T269M homomers exhibited a high glycine sensitivity accompanied by a substantial leak current, suggestive of an altered function that could dramatically enhance glycinergic signaling. These results may explain the heterogeneity of clinical phenotypes associated with GLRA2 mutations and reveal that missense variants can result in a loss, gain or alteration of GlyR α2 function. In turn, these GlyR α2 missense variants are likely to either negatively or positively deregulate cortical progenitor homeostasis and neuronal migration in the developing brain, leading to changes in cognition, learning, and memory.
Collapse
Affiliation(s)
- Xiumin Chen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Katie A. Wilson
- Research School of Chemistry, The Australian National University, Canberra, ACT, Australia
| | - Natascha Schaefer
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Lachlan De Hayr
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Mark Windsor
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Emmanuel Scalais
- Neurologie Pédiatrique, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | | | - Katrien Stouffs
- Center for Medical Genetics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Megan L. O’Mara
- Research School of Chemistry, The Australian National University, Canberra, ACT, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Joseph W. Lynch
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Robert J. Harvey
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
- *Correspondence: Robert J. Harvey,
| |
Collapse
|
9
|
San Martín VP, Sazo A, Utreras E, Moraga-Cid G, Yévenes GE. Glycine Receptor Subtypes and Their Roles in Nociception and Chronic Pain. Front Mol Neurosci 2022; 15:848642. [PMID: 35401105 PMCID: PMC8984470 DOI: 10.3389/fnmol.2022.848642] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 01/23/2023] Open
Abstract
Disruption of the inhibitory control provided by the glycinergic system is one of the major mechanisms underlying chronic pain. In line with this concept, recent studies have provided robust proof that pharmacological intervention of glycine receptors (GlyRs) restores the inhibitory function and exerts anti-nociceptive effects on preclinical models of chronic pain. A targeted regulation of the glycinergic system requires the identification of the GlyR subtypes involved in chronic pain states. Nevertheless, the roles of individual GlyR subunits in nociception and in chronic pain are yet not well defined. This review aims to provide a systematic outline on the contribution of GlyR subtypes in chronic pain mechanisms, with a particular focus on molecular pathways of spinal glycinergic dis-inhibition mediated by post-translational modifications at the receptor level. The current experimental evidence has shown that phosphorylation of synaptic α1β and α3β GlyRs are involved in processes of spinal glycinergic dis-inhibition triggered by chronic inflammatory pain. On the other hand, the participation of α2-containing GlyRs and of β subunits in pain signaling have been less studied and remain undefined. Although many questions in the field are still unresolved, future progress in GlyR research may soon open new exciting avenues into understanding and controlling chronic pain.
Collapse
Affiliation(s)
- Victoria P. San Martín
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Anggelo Sazo
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Elías Utreras
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile
| | - Gustavo Moraga-Cid
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Gonzalo E. Yévenes
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- *Correspondence: Gonzalo E. Yévenes,
| |
Collapse
|
10
|
Araya A, Gallegos S, Viveros R, San Martin L, Muñoz B, Harvey RJ, Zeilhofer HU, Aguayo LG. Presence of ethanol sensitive and insensitive glycine receptors in the ventral tegmental area and prefrontal cortex in mice. Br J Pharmacol 2021; 178:4691-4707. [PMID: 34378188 PMCID: PMC9293192 DOI: 10.1111/bph.15649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/22/2021] [Accepted: 07/10/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Previous studies showed that glycine receptors (GlyRs) composed of α1 and β subunits are primarily found in spinal cord and brainstem and are potentiated by ethanol (10-100 mM). However, much less is known about the presence, composition, and ethanol sensitivity of GlyRs in higher CNS regions. In the present study, we examined two regions of the brain reward system, the ventral tegmental area (VTA) and the prefrontal cortex (PFC), to determine their GlyR subunit composition and sensitivity to ethanol. EXPERIMENTAL APPROACH To achieve these aims, we used Western blot, immunohistochemistry and electrophysiological techniques in three different models: Wild-type C57BL/6, GlyR α1 knock-in and GlyR α2 knockout mice. KEY RESULTS Similar levels of α and β GlyR subunits were detected in both brain regions, and electrophysiological recordings demonstrated the presence of glycine-activated currents in both areas. The sensitivity of GlyRs to glycine was lower in the PFC compared to VTA. Picrotoxin blocked the glycine-activated current in the PFC and VTA only partially, indicating that both regions express heteromeric αβ receptors. Interestingly, GlyRs in VTA neurons, but not in PFC neurons, were potentiated by ethanol. CONCLUSION AND IMPLICATIONS GlyRs in VTA neurons from WT and α2 KO mice were potentiated by ethanol, but not in neurons from the α1 KI mice, supporting the conclusion that α1 GlyRs are predominantly expressed in the VTA. By contrast, GlyRs in PFC neurons were not potentiated in any of the mouse models studied, suggesting the presence of either α2/α3/α4 rather than α1 GlyR subunits.
Collapse
Affiliation(s)
- Anibal Araya
- Department of Physiology, Universidad de Concepción, Concepción, Chile.,PhD Program in Pharmacology, Universidad de Chile, Santiago, Chile
| | - Scarlet Gallegos
- Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Rodrigo Viveros
- Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Loreto San Martin
- Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Braulio Muñoz
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert J Harvey
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Sippy Downs, Australia.,Sunshine Coast Health Institute, Birtinya, Queensland, Australia
| | - Hanns U Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, and Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Luis G Aguayo
- Department of Physiology, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
11
|
Solorza J, Oliva CA, Castillo K, Amestica G, Maldifassi MC, López-Cortés XA, Barra R, Stehberg J, Piesche M, Sáez-Briones P, González W, Arenas-Salinas M, Mariqueo TA. Effects of Interleukin-1β in Glycinergic Transmission at the Central Amygdala. Front Pharmacol 2021; 12:613105. [PMID: 33746753 PMCID: PMC7973117 DOI: 10.3389/fphar.2021.613105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/19/2021] [Indexed: 01/22/2023] Open
Abstract
Interleukin-1β (IL-1β) is an important cytokine that modulates peripheral and central pain sensitization at the spinal level. Among its effects, it increases spinal cord excitability by reducing inhibitory Glycinergic and GABAergic neurotransmission. In the brain, IL-1β is released by glial cells in regions associated with pain processing during neuropathic pain. It also has important roles in neuroinflammation and in regulating NMDA receptor activity required for learning and memory. The modulation of glycine-mediated inhibitory activity via IL-1β may play a critical role in the perception of different levels of pain. The central nucleus of the amygdala (CeA) participates in receiving and processing pain information. Interestingly, this nucleus is enriched in the regulatory auxiliary glycine receptor (GlyR) β subunit (βGlyR); however, no studies have evaluated the effect of IL-1β on glycinergic neurotransmission in the brain. Hence, we hypothesized that IL-1β may modulate GlyR-mediated inhibitory activity via interactions with the βGlyR subunit. Our results show that the application of IL-1β (10 ng/ml) to CeA brain slices has a biphasic effect; transiently increases and then reduces sIPSC amplitude of CeA glycinergic currents. Additionally, we performed molecular docking, site-directed mutagenesis, and whole-cell voltage-clamp electrophysiological experiments in HEK cells transfected with GlyRs containing different GlyR subunits. These data indicate that IL-1β modulates GlyR activity by establishing hydrogen bonds with at least one key amino acid residue located in the back of the loop C at the ECD domain of the βGlyR subunit. The present results suggest that IL-1β in the CeA controls glycinergic neurotransmission, possibly via interactions with the βGlyR subunit. This effect could be relevant for understanding how IL-1β released by glia modulates central processing of pain, learning and memory, and is involved in neuroinflammation.
Collapse
Affiliation(s)
- Jocelyn Solorza
- Center for Medical Research, Laboratory of Neuropharmacology, School of Medicine, Universidad de Talca, Talca, Chile.,Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - Carolina A Oliva
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Gabriela Amestica
- Center for Medical Research, Laboratory of Neuropharmacology, School of Medicine, Universidad de Talca, Talca, Chile
| | - María Constanza Maldifassi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Xaviera A López-Cortés
- Department of Computer Science and Industries, Faculty of Engineering Science, Universidad Católica del Maule, Talca, Chile
| | - Rafael Barra
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago, Chile
| | - Jimmy Stehberg
- Faculty of Biological Sciences and Faculty of Medicine, Instituto de Ciencias Biomédicas, Universidad Andres Bello, Santiago, Chile
| | - Matthias Piesche
- Laboratory of Biomedical Research, Medicine Faculty, Universidad Católica del Maule, Talca, Chile.,Oncology Center, Medicine Faculty, Universidad Católica del Maule, Talca, Chile
| | - Patricio Sáez-Briones
- Laboratory of Neuropharmacology and Behavior, School of Medicine, Faculty of Medical Sciences, Universidad de Santiago de Chile (USACH), Santiago, Chile
| | - Wendy González
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Mauricio Arenas-Salinas
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - Trinidad A Mariqueo
- Center for Medical Research, Laboratory of Neuropharmacology, School of Medicine, Universidad de Talca, Talca, Chile
| |
Collapse
|
12
|
Reduced sedation and increased ethanol consumption in knock-in mice expressing an ethanol insensitive alpha 2 subunit of the glycine receptor. Neuropsychopharmacology 2021; 46:528-536. [PMID: 32357359 PMCID: PMC8026987 DOI: 10.1038/s41386-020-0689-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/20/2020] [Accepted: 04/23/2020] [Indexed: 01/24/2023]
Abstract
Previous studies have shown the presence of several subunits of the inhibitory glycine receptor (GlyR) in the reward system, specifically in medium spiny neurons (MSNs) of the nucleus Accumbens (nAc). It was suggested that GlyR α1 subunits regulate nAc excitability and ethanol consumption. However, little is known about the role of the α2 subunit in the adult brain since it is a subunit highly expressed during early brain development. In this study, we used genetically modified mice with a mutation (KR389-390AA) in the intracellular loop of the GlyR α2 subunit which results in a heteromeric α2β receptor that is insensitive to ethanol. Using this mouse model denoted knock-in α2 (KI α2), our electrophysiological studies showed that neurons in the adult nAc expressed functional KI GlyRs that were rather insensitive to ethanol when compared with WT GlyRs. In behavioral tests, the KI α2 mice did not show any difference in basal motor coordination, locomotor activity, or conditioned place preference compared with WT littermate controls. In terms of ethanol response, KI α2 male mice recovered faster from the administration of ataxic and sedative doses of ethanol. Furthermore, KI α2 mice consumed higher amounts of ethanol in the first days of the drinking in the dark protocol, as compared with WT mice. These results show that the α2 subunit is important for the potentiation of GlyRs in the adult brain and this might result in reduced sedation and increased ethanol consumption.
Collapse
|
13
|
Subunit-Specific Photocontrol of Glycine Receptors by Azobenzene-Nitrazepam Photoswitcher. eNeuro 2021; 8:ENEURO.0294-20.2020. [PMID: 33298457 PMCID: PMC7877471 DOI: 10.1523/eneuro.0294-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Photopharmacology is a unique approach that through a combination of photochemistry methods and advanced life science techniques allows the study and control of specific biological processes, ranging from intracellular pathways to brain circuits. Recently, a first photochromic channel blocker of anion-selective GABAA receptors, the azobenzene-nitrazepam-based photochromic compound (Azo-NZ1), has been described. In the present study, using patch-clamp technique in heterologous system and in mice brain slices, site-directed mutagenesis and molecular modeling we provide evidence of the interaction of Azo-NZ1 with glycine receptors (GlyRs) and determine the molecular basis of this interaction. Glycinergic synaptic neurotransmission determines an important inhibitory drive in the vertebrate nervous system and plays a crucial role in the control of neuronal circuits in the spinal cord and brain stem. GlyRs are involved in locomotion, pain sensation, breathing, and auditory function, as well as in the development of such disorders as hyperekplexia, epilepsy, and autism. Here, we demonstrate that Azo-NZ1 blocks in a UV-dependent manner the activity of α2 GlyRs (GlyR2), while being barely active on α1 GlyRs (GlyR1). The site of Azo-NZ1 action is in the chloride-selective pore of GlyR at the 2’ position of transmembrane helix 2 and amino acids forming this site determine the difference in Azo-NZ1 blocking activity between GlyR2 and GlyR1. This subunit-specific modulation is also shown on motoneurons of brainstem slices from neonatal mice that switch during development from expressing “fetal” GlyR2 to “adult” GlyR1 receptors.
Collapse
|
14
|
Li Q, Jin R, Zhang S, Sun X, Wu J. Transient receptor potential vanilloid four channels modulate inhibitory inputs through differential regulation of GABA and glycine receptors in rat retinal ganglion cells. FASEB J 2020; 34:14521-14538. [PMID: 32892440 DOI: 10.1096/fj.201902937rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 01/06/2023]
Abstract
The transient receptor potential vanilloid 4 (TRPV4) channel is widely distributed in the retina. Activation of the TRPV4 channel enhances excitatory signaling from bipolar cells to retinal ganglion cells (RGCs), thereby increasing RGC firing rate and membrane excitability. In this study, we investigated the effect of TRPV4 channel activation on the miniature inhibitory postsynaptic current (mIPSC) in rat RGCs. Our results showed that perfusion with HC-067047, a TRPV4-channel antagonist, significantly reduced the amplitude of RGC mIPSCs. Extracellular application of the TRPV4 channel agonist GSK1016790A (GSK101) enhanced the frequency and amplitude of mIPSCs in ON- and OFF-type RGCs; pre-application of HC-067047 blocked the effect of GSK101 on mIPSCs. Furthermore, TRPV4 channels were able to enhance the frequency and amplitude of glycine receptor (GlyR)-mediated mIPSCs and inhibit the frequency of type A γ-aminobutyric acid receptor (GABAA R)-mediated mIPSCs. Upon intracellular administration or intravitreal injection of GSK101, TRPV4 channel activation reduced the release of presynaptic glycine and enhanced the function and expression of postsynaptic GlyRs; however, it inhibited presynaptic release of GABA, but did not affect postsynaptic GABAA Rs. Our study results provide insight regarding the effect of TRPV4 channel activation on RGCs and offer a potential interventional target for retinal diseases involving TRPV4 channels.
Collapse
Affiliation(s)
- Qian Li
- Eye Institute, Eye and ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ruiri Jin
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shenghai Zhang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinghuai Sun
- Eye Institute, Eye and ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Shanghai, China.,Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
15
|
San Martin L, Gallegos S, Araya A, Romero N, Morelli G, Comhair J, Harvey RJ, Rigo J, Brone B, Aguayo LG. Ethanol consumption and sedation are altered in mice lacking the glycine receptor α2 subunit. Br J Pharmacol 2020; 177:3941-3956. [PMID: 32436225 PMCID: PMC7429487 DOI: 10.1111/bph.15136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/31/2020] [Accepted: 05/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The precise mechanism/s of action of ethanol, although studied for many years, are not well understood. Like other drugs of abuse, ethanol affects dopamine levels in the nucleus accumbens (nAc), an important region of the mesolimbic system, causing a reinforcing effect. It has been shown that glycine receptors (GlyRs) present in the nAc are potentiated by clinically relevant concentrations of ethanol, where α1 and α2 are the predominant subunits expressed. EXPERIMENTAL APPROACH Using a combination of electrophysiology and behavioural assays, we studied the involvement of GlyR α2 subunits on the effects of low and high doses of ethanol, as well as on consumption using mice lacking the GlyR α2 subunit (male Glra2-/Y and female Glra2-/- ). KEY RESULTS GlyR α2 subunits exist in accumbal neurons, since the glycine-evoked currents and glycinergic miniature inhibitory postsynaptic currents (mIPSCs) in Glra2-/Y mice were drastically decreased. In behavioural studies, differences in ethanol consumption and sedation were observed between wild-type (WT) and Glra2 knockout (KO) mice. Using the drinking in the dark (DID) paradigm, we found that Glra2-/Y mice presented a binge-like drinking behaviour immediately when exposed to ethanol rather than the gradual consumption seen in WT animals. Interestingly, the effect of knocking out Glra2 in female (Glra2-/- ) mice was less evident, since WT female mice already showed higher DID. CONCLUSION AND IMPLICATIONS The differences in ethanol consumption between WT and KO mice provide additional evidence supporting the conclusion that GlyRs are biologically relevant targets for the sedative and rewarding properties of ethanol.
Collapse
Affiliation(s)
- Loreto San Martin
- Laboratory of Neurophysiology, Department of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Scarlet Gallegos
- Laboratory of Neurophysiology, Department of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Anibal Araya
- Laboratory of Neurophysiology, Department of PhysiologyUniversidad de ConcepciónConcepciónChile
| | - Nicol Romero
- Laboratory of Neurophysiology, Department of PhysiologyUniversidad de ConcepciónConcepciónChile
| | | | | | - Robert J. Harvey
- School of Health and Sport SciencesUniversity of the Sunshine CoastMaroochydore DCQueenslandAustralia
- Sunshine Coast Health InstituteBirtinyaQueenslandAustralia
| | | | | | - Luis G. Aguayo
- Laboratory of Neurophysiology, Department of PhysiologyUniversidad de ConcepciónConcepciónChile
| |
Collapse
|
16
|
Nakamura M, Jang IS, Yamaga T, Kotani N, Akaike N. Effects of nitrous oxide on glycinergic transmission in rat spinal neurons. Brain Res Bull 2020; 162:191-198. [PMID: 32599127 DOI: 10.1016/j.brainresbull.2020.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/16/2020] [Accepted: 06/22/2020] [Indexed: 11/19/2022]
Abstract
We investigated the effects of nitrous oxide (N2O) on glycinergic inhibitory whole-cell and synaptic responses using a "synapse bouton preparation," dissociated mechanically from rat spinal sacral dorsal commissural nucleus (SDCN) neurons. This technique can evaluate pure single- or multi-synaptic responses from native functional nerve endings and enable us to accurately quantify how N2O influences pre- and postsynaptic transmission. We found that 70 % N2O enhanced exogenous glycine-induced whole-cell currents (IGly) at glycine concentrations lower than 3 × 10-5 M, but did not affect IGly at glycine concentrations higher than 10-4 M. N2O did not affect the amplitude and 1/e decay-time of both spontaneous and miniature glycinergic inhibitory postsynaptic currents recorded in the absence and presence of tetrodotoxin (sIPSCs and mIPSCs, respectively). The decrease in frequency induced by N2O was observed in sIPSCs but not in mIPSCs, which was recorded in the presence of both tetrodotoxin and Cd2+, which block voltage-gated Na+ and Ca2+ channels, respectively. N2O also decreased the amplitude and increased the failure rate and paired-pulse ratio of action potential-evoked glycinergic inhibitory postsynaptic currents. N2O slightly decreased the Ba2+ currents mediated by voltage-gated Ca2+ channels in SDCN neurons. We found that N2O suppresses glycinergic responses at synaptic levels with presynaptic effect having much more predominant role. The difference between glycinergic whole-cell and synaptic responses suggests that extrasynaptic responses seriously modulate whole-cell currents. Our results strongly suggest that these responses may thus in part explain analgesic effects of N2O via marked glutamatergic inhibition by glycinergic responses in the spinal cord.
Collapse
Affiliation(s)
- Michiko Nakamura
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea
| | - Toshitaka Yamaga
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama 343-0821, Japan
| | - Norio Akaike
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama 343-0821, Japan; Research Division for Clinical Pharmacology, Medical Corporation, Juryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kita-ku, Kumamoto 860-8518, Japan.
| |
Collapse
|
17
|
Gazan A, Rial D, Schiffmann SN. Ablation of striatal somatostatin interneurons affects MSN morphology and electrophysiological properties, and increases cocaine‐induced hyperlocomotion in mice. Eur J Neurosci 2020; 51:1388-1402. [DOI: 10.1111/ejn.14581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Adeline Gazan
- Laboratory of Neurophysiology ULB Neuroscience Institute Université Libre de Bruxelles (ULB) Brussels Belgium
| | - Daniel Rial
- Laboratory of Neurophysiology ULB Neuroscience Institute Université Libre de Bruxelles (ULB) Brussels Belgium
| | - Serge N. Schiffmann
- Laboratory of Neurophysiology ULB Neuroscience Institute Université Libre de Bruxelles (ULB) Brussels Belgium
| |
Collapse
|
18
|
Comhair J, Devoght J, Morelli G, Harvey RJ, Briz V, Borrie SC, Bagni C, Rigo JM, Schiffmann SN, Gall D, Brône B, Molchanova SM. Alpha2-Containing Glycine Receptors Promote Neonatal Spontaneous Activity of Striatal Medium Spiny Neurons and Support Maturation of Glutamatergic Inputs. Front Mol Neurosci 2018; 11:380. [PMID: 30374290 PMCID: PMC6196267 DOI: 10.3389/fnmol.2018.00380] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 09/24/2018] [Indexed: 12/16/2022] Open
Abstract
Glycine receptors (GlyRs) containing the α2 subunit are highly expressed in the developing brain, where they regulate neuronal migration and maturation, promote spontaneous network activity and subsequent development of synaptic connections. Mutations in GLRA2 are associated with autism spectrum disorder, but the underlying pathophysiology is not described yet. Here, using Glra2-knockout mice, we found a GlyR-dependent effect on neonatal spontaneous activity of dorsal striatum medium spiny neurons (MSNs) and maturation of the incoming glutamatergic innervation. Our data demonstrate that functional GlyRs are highly expressed in MSNs of one-week-old mice, but they do not generate endogenous chloride-mediated tonic or phasic current. Despite of that, knocking out the Glra2 severely affects the shape of action potentials and impairs spontaneous activity and the frequency of miniature AMPA receptor-mediated currents in MSNs. This reduction in spontaneous activity and glutamatergic signaling can attribute to the observed changes in neonatal behavioral phenotypes as seen in ultrasonic vocalizations and righting reflex. In adult Glra2-knockout animals, the glutamatergic synapses in MSNs remain functionally underdeveloped. The number of glutamatergic synapses and release probability at presynaptic site remain unaffected, but the amount of postsynaptic AMPA receptors is decreased. This deficit is a consequence of impaired development of the neuronal circuitry since acute inhibition of GlyRs by strychnine in adult MSNs does not affect the properties of glutamatergic synapses. Altogether, these results demonstrate that GlyR-mediated signaling supports neonatal spontaneous MSN activity and, in consequence, promotes the functional maturation of glutamatergic synapses on MSNs. The described mechanism might shed light on the pathophysiological mechanisms in GLRA2-linked autism spectrum disorder cases.
Collapse
Affiliation(s)
- Joris Comhair
- Laboratory of Neurophysiology, ULB-Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium.,BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| | - Jens Devoght
- BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| | - Giovanni Morelli
- BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| | - Robert J Harvey
- School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, QLD, Australia.,Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Victor Briz
- Center for Human Genetics and Leuven Research Institute for Neuroscience and Disease, KU Leuven, Leuven, Belgium.,VIB Center for the Biology of Disease, Leuven, Belgium
| | - Sarah C Borrie
- Center for Human Genetics and Leuven Research Institute for Neuroscience and Disease, KU Leuven, Leuven, Belgium.,VIB Center for the Biology of Disease, Leuven, Belgium
| | - Claudia Bagni
- Center for Human Genetics and Leuven Research Institute for Neuroscience and Disease, KU Leuven, Leuven, Belgium.,VIB Center for the Biology of Disease, Leuven, Belgium
| | - Jean-Michel Rigo
- BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| | - Serge N Schiffmann
- Laboratory of Neurophysiology, ULB-Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - David Gall
- Laboratory of Neurophysiology, ULB-Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Bert Brône
- BIOMED Research Institute, University of Hasselt, Hasselt, Belgium
| | - Svetlana M Molchanova
- Laboratory of Neurophysiology, ULB-Neuroscience Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|