1
|
Fehrentz T, Amin E, Görldt N, Strasdeit T, Moussavi-Torshizi SE, Leippe P, Trauner D, Meyer C, Frey N, Sasse P, Klöcker N. Optical control of cardiac electrophysiology by the photochromic ligand azobupivacaine 2. Br J Pharmacol 2025; 182:1125-1142. [PMID: 39543799 DOI: 10.1111/bph.17394] [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: 05/17/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Patients suffering from ischaemic heart disease and heart failure are at high risk of recurrent ventricular arrhythmias (VAs), eventually leading to sudden cardiac death. While high-voltage shocks delivered by an implantable defibrillator may prevent sudden cardiac death, these interventions themselves impair quality of life and raise both morbidity and mortality, which accentuates the need for developing novel defibrillation techniques. EXPERIMENTAL APPROACH Photopharmacology allows for reversible control of biological processes by light. When relying on synthetic and externally applied chromophores, it renders genetic modification of target cells dispensable and may hence be advantageous over optogenetic approaches. Here, the photochromic ligand azobupivacaine 2 (AB2) was probed as a modulator of cardiac electrophysiology in an ex vivo intact mouse heart model. KEY RESULTS By reversibly blocking voltage-gated Na+ and K+ channels, photoswitching of AB2 modulated both the ventricular effective refractory period and the conduction velocity in native heart tissue. Moreover, photoswitching of AB2 was able to convert VA into sinus rhythm. CONCLUSION AND IMPLICATIONS The present study provides the first proof of concept that AB2 enables gradual control of cardiac electrophysiology by light. AB2 may hence open the door to the development of an optical defibrillator based on photopharmacology.
Collapse
Affiliation(s)
- Timm Fehrentz
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ehsan Amin
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nicole Görldt
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Strasdeit
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Seyed-Erfan Moussavi-Torshizi
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Leippe
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Dirk Trauner
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christian Meyer
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Division of Cardiology, Angiology and Intensive Care, EVK Düsseldorf, Cardiac Neuro- and Electrophysiology Research Consortium (cNEP), Düsseldorf, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
2
|
Munguba H, Srivastava I, Gutzeit VA, Singh A, Vijay A, Kristt M, Arefin A, Thukral S, Broichhagen J, Stujenske JM, Liston C, Levitz J. Projection-targeted photopharmacology reveals distinct anxiolytic roles for presynaptic mGluR2 in prefrontal- and insula-amygdala synapses. Neuron 2025:S0896-6273(25)00006-6. [PMID: 39879977 DOI: 10.1016/j.neuron.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/11/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025]
Abstract
Dissecting how membrane receptors regulate neural circuits is critical for deciphering principles of neuromodulation and mechanisms of drug action. Here, we use a battery of optical approaches to determine how presynaptic metabotropic glutamate receptor 2 (mGluR2) in the basolateral amygdala (BLA) controls anxiety-related behavior in mice. Using projection-specific photopharmacological activation, we find that mGluR2-mediated presynaptic inhibition of ventromedial prefrontal cortex (vmPFC)-BLA, but not posterior insular cortex (pIC)-BLA, connections produces a long-lasting decrease in spatial avoidance. In contrast, presynaptic inhibition of pIC-BLA connections decreases social avoidance and novelty-induced hypophagia without impairing working memory, establishing this projection as a novel target for the treatment of anxiety disorders. Fiber photometry and viral mapping reveal distinct activity patterns and anatomical organization of vmPFC-BLA and pIC-BLA circuits. Together, this work reveals new aspects of BLA neuromodulation with therapeutic implications while establishing a powerful approach for optical mapping of drug action.
Collapse
Affiliation(s)
- Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ipsit Srivastava
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Vanessa A Gutzeit
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ashna Singh
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Akshara Vijay
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melanie Kristt
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anisul Arefin
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sonal Thukral
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Joseph M Stujenske
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
3
|
Michałowski MA, Kłopotowski K, Wiera G, Czyżewska MM, Mozrzymas JW. Molecular mechanisms of the GABA type A receptor function. Q Rev Biophys 2025; 58:e3. [PMID: 39806800 DOI: 10.1017/s0033583524000179] [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] [Indexed: 01/16/2025]
Abstract
The GABA type A receptor (GABAAR) belongs to the family of pentameric ligand-gated ion channels and plays a key role in inhibition in adult mammalian brains. Dysfunction of this macromolecule may lead to epilepsy, anxiety disorders, autism, depression, and schizophrenia. GABAAR is also a target for multiple physiologically and clinically relevant modulators, such as benzodiazepines (BDZs), general anesthetics, and neurosteroids. The first GABAAR structure appeared in 2014, but the past years have brought a particularly abundant surge in structural data for these receptors with various ligands and modulators. Although the open conformation remains elusive, this novel information has pushed the structure-function studies to an unprecedented level. Electrophysiology, mutagenesis, photolabeling, and in silico simulations, guided by novel structural information, shed new light on the molecular mechanisms of receptor functioning. The main goal of this review is to present the current knowledge of GABAAR functional and structural properties. The review begins with an outline of the functional and structural studies of GABAAR, accompanied by some methodological considerations, especially biophysical methods, enabling the reader to follow how major breakthroughs in characterizing GABAAR features have been achieved. The main section provides a comprehensive analysis of the functional significance of specific structural elements in GABAARs. We additionally summarize the current knowledge on the binding sites for major GABAAR modulators, referring to the molecular underpinnings of their action. The final chapter of the review moves beyond examining GABAAR as an isolated macromolecule and describes the interactions of the receptor with other proteins in a broader context of inhibitory plasticity. In the final section, we propose a general conclusion that agonist binding to the orthosteric binding sites appears to rely on local interactions, whereas conformational transitions of bound macromolecule (gating) and allosteric modulation seem to reflect more global phenomena involving vast portions of the macromolecule.
Collapse
Affiliation(s)
- Michał A Michałowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Karol Kłopotowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Grzegorz Wiera
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Marta M Czyżewska
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Jerzy W Mozrzymas
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
4
|
Welle TM, Smith KR. Release your inhibitions: The cell biology of GABAergic postsynaptic plasticity. Curr Opin Neurobiol 2024; 90:102952. [PMID: 39721557 DOI: 10.1016/j.conb.2024.102952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
GABAergic synaptic inhibition controls circuit function by regulating neuronal plasticity, excitability, and firing. To achieve these goals, inhibitory synapses themselves undergo several forms of plasticity via diverse mechanisms, strengthening and weakening phasic inhibition in response to numerous activity-induced stimuli. These mechanisms include changing the number and arrangement of functional GABAARs within the inhibitory postsynaptic domain (iPSD), which can profoundly regulate inhibitory synapse strength. Here, we explore recent advances in our molecular understanding of inhibitory postsynaptic plasticity, with a focus on modulation of the trafficking, protein-protein interactions, nanoscale-organization, and posttranscriptional regulation of GABAARs and iPSD proteins. What has emerged is a complex mechanistic picture of how synaptic inhibition is controlled, with critical ramifications for cognition under typical and pathogenic conditions.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
5
|
Guet-McCreight A, Mazza F, Prevot TD, Sibille E, Hay E. Therapeutic dose prediction of α5-GABA receptor modulation from simulated EEG of depression severity. PLoS Comput Biol 2024; 20:e1012693. [PMID: 39729407 DOI: 10.1371/journal.pcbi.1012693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
Treatment for major depressive disorder (depression) often has partial efficacy and a large portion of patients are treatment resistant. Recent studies implicate reduced somatostatin (SST) interneuron inhibition in depression, and new pharmacology boosting this inhibition via positive allosteric modulators of α5-GABAA receptors (α5-PAM) offers a promising effective treatment. However, testing the effect of α5-PAM on human brain activity is limited, meriting the use of detailed simulations. We utilized our previous detailed computational models of human depression microcircuits with reduced SST interneuron inhibition and α5-PAM effects, to simulate EEG of individual microcircuits across depression severity and α5-PAM doses. We developed machine learning models that predicted optimal dose from EEG with high accuracy and recovered microcircuit activity and EEG. This study provides dose prediction models for α5-PAM administration based on EEG biomarkers of depression severity. Given limitations in doing the above in the living human brain, the results and tools we developed will facilitate translation of α5-PAM treatment to clinical use.
Collapse
Affiliation(s)
| | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Maleeva G, Nin-Hill A, Wirth U, Rustler K, Ranucci M, Opar E, Rovira C, Bregestovski P, Zeilhofer HU, König B, Alfonso-Prieto M, Gorostiza P. Light-Activated Agonist-Potentiator of GABA A Receptors for Reversible Neuroinhibition in Wildtype Mice. J Am Chem Soc 2024; 146:28822-28831. [PMID: 39383450 PMCID: PMC11503767 DOI: 10.1021/jacs.4c08446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024]
Abstract
Gamma aminobutyric acid type A receptors (GABAARs) play a key role in the mammalian central nervous system (CNS) as drivers of neuroinhibitory circuits, which are commonly targeted for therapeutic purposes with potentiator drugs. However, due to their widespread expression and strong inhibitory action, systemic pharmaceutical potentiation of GABAARs inevitably causes adverse effects regardless of the drug selectivity. Therefore, therapeutic guidelines must often limit or exclude clinically available GABAAR potentiators, despite their high efficacy, good biodistribution, and favorable molecular properties. One solution to this problem is to use drugs with light-dependent activity (photopharmacology) in combination with on-demand, localized illumination. However, a suitable light-activated potentiator of GABAARs has been elusive so far for use in wildtype mammals. We have met this need by developing azocarnil, a diffusible GABAergic agonist-potentiator based on the anxiolytic drug abecarnil that is inactive in the dark and activated by visible violet light. Azocarnil can be rapidly deactivated with green light and by thermal relaxation in the dark. We demonstrate that it selectively inhibits neuronal currents in hippocampal neurons in vitro and in the dorsal horns of the spinal cord of mice, decreasing the mechanical sensitivity as a function of illumination without displaying systemic adverse effects. Azocarnil expands the in vivo photopharmacological toolkit with a novel chemical scaffold and achieves a milestone toward future phototherapeutic applications to safely treat muscle spasms, pain, anxiety, sleep disorders, and epilepsy.
Collapse
Affiliation(s)
- Galyna Maleeva
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
| | - Alba Nin-Hill
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica) & Institut de Química
Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08020, Spain
| | - Ulrike Wirth
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Karin Rustler
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Matteo Ranucci
- Institute
of Pharmacology and Toxicology, University
of Zurich, Zürich 8057, Switzerland
| | - Ekin Opar
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
- Doctorate
program of the University of Barcelona, Barcelona 08020, Spain
| | - Carme Rovira
- Departament
de Química Inorgànica i Orgànica (Secció
de Química Orgànica) & Institut de Química
Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Barcelona 08020, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Piotr Bregestovski
- Institut
de Neurosciences des Systèmes, UMR INSERM 1106, Aix-Marseille Université, Marseille 13005, France
| | - Hanns Ulrich Zeilhofer
- Institute
of Pharmacology and Toxicology, University
of Zurich, Zürich 8057, Switzerland
- Institute
of Pharmaceutical Sciences, Swiss Federal
Institute of Technology (ETH) Zürich, Zürich 8093, Switzerland
| | - Burkhard König
- Institute
of Organic Chemistry, University of Regensburg, Regensburg 93053, Germany
| | - Mercedes Alfonso-Prieto
- Institute
of Neuroscience and Medicine INM-9 Computational Biomedicine, Forschungszentrum Jülich GmbH, D-52428 Jülich, Germany
| | - Pau Gorostiza
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona 08028, Spain
- Networking
Biomedical Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), ISCIII, Barcelona 08028, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| |
Collapse
|
7
|
Zeng L, Yu T, Liu H, Li M, Wang J, Wang C, Xu P. IGF-1's protective effect on OSAS rats' learning and memory. Sleep Breath 2024; 28:1919-1928. [PMID: 38858326 PMCID: PMC11450044 DOI: 10.1007/s11325-024-03047-8] [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: 08/04/2023] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 06/12/2024]
Abstract
PURPOSE Patients with obstructive sleep apnea syndrome (OSAS) frequently experience cognitive dysfunction, which may be connected to chronic intermittent hypoxia (CIH). Insulin-like growth factor-1 (IGF-1) is thought to be closely associated with cognitive function, but its role in cognitive impairment caused by OSAS is unclear. The purpose of this study was to investigate the potential protective effect of IGF-1 on cognitive impairment in OSAS rats. METHODS Healthy male SD rats (n = 40) were randomly assigned into four groups: control group, CIH group, NS + CIH group, and IGF-1 + CIH group. All experimental rats except for those in the control group were exposed to intermittent hypoxic (IH) environments for 8 h per day over 28 days. Prior to daily exposure to IH, rats in the IGF-1 + CIH group received subcutaneous injections of IGF-1. The Morris water maze test was conducted on all experimental rats. Brain tissue testing methods included Enzyme-Linked Immunosorbent Assay, Hematoxylin and eosin staining, Immunohistochemistry, and Western blotting. RESULTS The rat model of OSAS was successfully established following exposure to CIH and exhibited significant cognitive impairment. However, daily subcutaneous injections of IGF-1 partially restored the impaired cognitive function in OSAS rats. Compared with the control group, there was a significant decrease in the expression levels of IGF-1, p-IGF-IR, and SYP in the CIH group; however, these expression levels increased significantly in the IGF-I + CIH group. CONCLUSION In OSAS rats, IGF-1 enhances learning memory; this effect may be linked to increased p-IGF-1R and SYP protein production in the hippocampus.
Collapse
Affiliation(s)
- Ling Zeng
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Ting Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Haijun Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Mi Li
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Jin Wang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Changsheng Wang
- Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, Guizhou, 563000, China.
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
8
|
Long P, Ma Q, Wang Z, Wang G, Jiang J, Gao L. Genetic patterning in hippocampus of rat undergoing impaired spatial memory induced by long-term heat stress. Heliyon 2024; 10:e37319. [PMID: 39296065 PMCID: PMC11408118 DOI: 10.1016/j.heliyon.2024.e37319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
The organism's normal physiological function is greatly impacted in a febrile environment, leading to the manifestation of pathological conditions including elevated body temperature, dehydration, gastric bleeding, and spermatogenic dysfunction. Numerous lines of evidence indicate that heat stress significantly impacts the brain's structure and function. Previous studies have demonstrated that both animals and humans experience cognitive impairment as a result of exposure to high temperatures. However, there is a lack of research on the effects of prolonged exposure to high-temperature environments on learning and memory function, as well as the underlying molecular regulatory mechanisms. In this study, we examined the impact of long-term heat stress exposure on spatial memory function in rats and conducted transcriptome sequencing analysis of rat hippocampal tissues to identify the crucial molecular targets affected by prolonged heat stress exposure. It was found that the long-term heat stress impaired rats' spatial memory function due to the pathological damages and apoptosis of hippocampal neurons at the CA3 region, which is accompanied with the decrease of growth hormone level in peripheral blood. RNA sequencing analysis revealed the signaling pathways related to positive regulation of external stimulation response and innate immune response were dramatically affected by heat stress. Among the verified differentially expressed genes, the knockdown of Arhgap36 in neuronal cell line HT22 significantly enhances the cell apoptosis, suggesting the impaired spatial memory induced by long-term heat stress may at least partially be mediated by the dysregulation of Arhgap36 in hippocampal neurons. The uncovered relationship between molecular changes in the hippocampus and behavioral alterations induced by long-term heat stress may offer valuable insights for the development of therapeutic targets and protective drugs to enhance memory function in heat-exposed individuals.
Collapse
Affiliation(s)
- Peihua Long
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Qunfei Ma
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Zhe Wang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Guanqin Wang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Jianan Jiang
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
| | - Lu Gao
- Department of Physiology, Naval Medical University, Shanghai, 200433, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200120, PR China
| |
Collapse
|
9
|
Xiang F, Zhang S, Tang M, Li P, Zhang H, Xiong J, Zhang Q, Li X. Optogenetics Neuromodulation of the Nose. Behav Neurol 2024; 2024:2627406. [PMID: 39165250 PMCID: PMC11335419 DOI: 10.1155/2024/2627406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/22/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Recently developed optogenetic technology, which allows high-fidelity control of neuronal activity, has been applied to investigate the neural circuits underlying sensory processing and behavior. The nasal cavity is innervated by the olfactory nerve and trigeminal nerve, which are closely related to common symptoms of rhinitis, such as impairment of smell, itching, and sneezing. The olfactory system has an amazing ability to distinguish thousands of odorant molecules at trace levels. However, there are many issues in olfactory sensing mechanisms that need to be addressed. Optogenetics offers a novel technical approach to solve this dilemma. Therefore, we review the recent advances in olfactory optogenetics to clarify the mechanisms of chemical sensing, which may help identify the mechanism of dysfunction and suggest possible treatments for impaired smell. Additionally, in rhinitis patients, alterations in the other nerve (trigeminal nerve) that innervates the nasal cavity can lead to hyperresponsiveness to various nociceptive stimuli and central sensitization, causing frequent and persistent itching and sneezing. In the last several years, the application of optogenetics in regulating nociceptive receptors, which are distributed in sensory nerve endings, and amino acid receptors, which are distributed in vital brain regions, to alleviate overreaction to nociceptive stimuli, has gained significant attention. Therefore, we focus on the progress in optogenetics and its application in neuromodulation of nociceptive stimuli and discuss the potential clinical translation for treating rhinitis in the future.
Collapse
Affiliation(s)
- Feng Xiang
- TCM DepartmentChongqing University Cancer HospitalChongqing Cancer Hospital, Chongqing, China
| | - Shipeng Zhang
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mi Tang
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peijia Li
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Zhang
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiahui Xiong
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinxiu Zhang
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- E.N.T. DepartmentChengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinrong Li
- E.N.T. DepartmentHospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Ge Y, Craig AM. Haploinsufficiency of GABA A Receptor-Associated Clptm1 Enhances Phasic and Tonic Inhibitory Neurotransmission, Suppresses Excitatory Synaptic Plasticity, and Impairs Memory. J Neurosci 2024; 44:e0521242024. [PMID: 38942471 PMCID: PMC11308325 DOI: 10.1523/jneurosci.0521-24.2024] [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: 03/15/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024] Open
Abstract
The mechanisms utilized by neurons to regulate the efficacy of phasic and tonic inhibition and their impacts on synaptic plasticity and behavior are incompletely understood. Cleft lip and palate transmembrane protein 1 (Clptm1) is a membrane-spanning protein that interacts with multiple γ-aminobutyric acid type A receptor (GABAAR) subunits, trapping them in the endoplasmic reticulum and Golgi network. Overexpression and knock-down studies suggest that Clptm1 modulates GABAAR-mediated phasic inhibition and tonic inhibition as well as activity-induced inhibitory synaptic homeostasis in cultured hippocampal neurons. To investigate the role of Clptm1 in the modulation of GABAARs in vivo, we generated Clptm1 knock-out (KO) mice. Here, we show that genetic KO of Clptm1 elevated phasic and tonic inhibitory transmission in both male and female heterozygous mice. Although basal excitatory synaptic transmission was not affected, Clptm1 haploinsufficiency significantly blocked high-frequency stimulation-induced long-term potentiation (LTP) in hippocampal CA3→CA1 synapses. In the hippocampus-dependent contextual fear-conditioning behavior task, both male and female Clptm1 heterozygous KO mice exhibited impairment in contextual fear memory. In addition, LTP and contextual fear memory were rescued by application of L-655,708, a negative allosteric modulator of the extrasynaptic GABAAR α5 subunit. These results suggest that haploinsufficiency of Clptm1 contributes to cognitive deficits through altered synaptic transmission and plasticity by elevation of inhibitory neurotransmission, with tonic inhibition playing a major role.
Collapse
Affiliation(s)
- Yuan Ge
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| |
Collapse
|
11
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [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: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
12
|
Almeida VN. Somatostatin and the pathophysiology of Alzheimer's disease. Ageing Res Rev 2024; 96:102270. [PMID: 38484981 DOI: 10.1016/j.arr.2024.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-β plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aβ production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aβ to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aβ release, fibrillation and plaque formation. Aβ plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.
Collapse
Affiliation(s)
- Victor N Almeida
- Institute of Psychiatry, Faculty of Medicine, University of São Paulo (USP), Brazil; Faculty of Languages, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
13
|
McCoy AM, Prevot TD, Mian MY, Sharmin D, Ahmad AN, Cook JM, Sibille EL, Lodge DJ. Extrasynaptic localization is essential for α5GABA A receptor modulation of dopamine system function. eNeuro 2024; 11:ENEURO.0344-23.2023. [PMID: 38413199 PMCID: PMC10972738 DOI: 10.1523/eneuro.0344-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 02/29/2024] Open
Abstract
Dopamine system dysfunction, observed in animal models with psychosis-like symptomatology, can be restored by targeting Gamma-Aminobutyric Acid type A receptors (GABAAR) containing the α5, but not α1, subunit in the ventral hippocampus (vHipp). The reason for this discrepancy in efficacy remains elusive; however, one key difference is that α1GABAARs are primarily located in the synapse, whereas α5GABAARs are mostly extrasynaptic. To test whether receptor location is responsible for this difference in efficacy, we injected a small interfering ribonucleic acid (siRNA) into the vHipp to knock down radixin, a scaffolding protein that holds α5GABAARs in the extrasynaptic space. We then administered GL-II-73, a positive allosteric modulator of α5GABAARs (α5-PAM) known to reverse shock-induced deficits in dopamine system function, to determine if shifting α5GABAARs from the extrasynaptic space to the synapse would prevent the effects of α5-PAM on dopamine system function. As expected, knockdown of radixin significantly decreased radixin-associated α5GABAARs and increased the proportion of synaptic α5GABAARs, without changing the overall expression of α5GABAARs. Importantly, GL-II-73 was no longer able to modulate dopamine neuron activity in radixin-knockdown rats, indicating that the extrasynaptic localization of α5GABAARs is critical for hippocampal modulation of the dopamine system. These results may have important implications for clinical use of GL-II-73, as periods of high hippocampal activity appear to favor synaptic α5GABAARs, thus efficacy may be diminished in conditions where aberrant hippocampal activity is present.Significance Statement Currently available treatments for psychosis, a debilitating symptom linked with several brain disorders, are inadequate. While they can help manage symptoms in some patients, they do so imperfectly. They are also associated with severe side effects that can cause discontinuation of medication. This study provides preclinical evidence that the drug, GL-II-73, possesses the ability to modulate dopamine activity, a key player in psychosis symptoms, and further provides some mechanistic details regarding these effects. Overall, this work contributes to the growing body of literature suggesting that GL-II-73 and similar compounds may possess antipsychotic efficacy.
Collapse
Affiliation(s)
- Alexandra M. McCoy
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas 78229
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas 78229
| | - Thomas D. Prevot
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario M5G 2C1, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Md Yeunus Mian
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - Adeeba N. Ahmad
- University of Texas, Rio Grande Valley, Edinburg, Texas 78539
| | - James M. Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - Etienne L. Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Ontario M5G 2C1, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas 78229
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas 78229
| |
Collapse
|
14
|
Guet-McCreight A, Chameh HM, Mazza F, Prevot TD, Valiante TA, Sibille E, Hay E. In-silico testing of new pharmacology for restoring inhibition and human cortical function in depression. Commun Biol 2024; 7:225. [PMID: 38396202 PMCID: PMC10891083 DOI: 10.1038/s42003-024-05907-1] [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: 08/30/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Reduced inhibition by somatostatin-expressing interneurons is associated with depression. Administration of positive allosteric modulators of α5 subunit-containing GABAA receptor (α5-PAM) that selectively target this lost inhibition exhibit antidepressant and pro-cognitive effects in rodent models of chronic stress. However, the functional effects of α5-PAM on the human brain in vivo are unknown, and currently cannot be assessed experimentally. We modeled the effects of α5-PAM on tonic inhibition as measured in human neurons, and tested in silico α5-PAM effects on detailed models of human cortical microcircuits in health and depression. We found that α5-PAM effectively recovered impaired cortical processing as quantified by stimulus detection metrics, and also recovered the power spectral density profile of the microcircuit EEG signals. We performed an α5-PAM dose-response and identified simulated EEG biomarker candidates. Our results serve to de-risk and facilitate α5-PAM translation and provide biomarkers in non-invasive brain signals for monitoring target engagement and drug efficacy.
Collapse
Affiliation(s)
- Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | | | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Taufik A Valiante
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, Toronto, ON, Canada
- Max Planck-University of Toronto Center for Neural Science and Technology, Toronto, ON, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Munguba H, Gutzeit VA, Srivastava I, Kristt M, Singh A, Vijay A, Arefin A, Thukral S, Broichhagen J, Stujenske JM, Liston C, Levitz J. Projection-Targeted Photopharmacology Reveals Distinct Anxiolytic Roles for Presynaptic mGluR2 in Prefrontal- and Insula-Amygdala Synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575699. [PMID: 38293136 PMCID: PMC10827048 DOI: 10.1101/2024.01.15.575699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Dissecting how membrane receptors regulate neural circuit function is critical for deciphering basic principles of neuromodulation and mechanisms of therapeutic drug action. Classical pharmacological and genetic approaches are not well-equipped to untangle the roles of specific receptor populations, especially in long-range projections which coordinate communication between brain regions. Here we use viral tracing, electrophysiological, optogenetic, and photopharmacological approaches to determine how presynaptic metabotropic glutamate receptor 2 (mGluR2) activation in the basolateral amygdala (BLA) alters anxiety-related behavior. We find that mGluR2-expressing neurons from the ventromedial prefrontal cortex (vmPFC) and posterior insular cortex (pIC) preferentially target distinct cell types and subregions of the BLA to regulate different forms of avoidant behavior. Using projection-specific photopharmacological activation, we find that mGluR2-mediated presynaptic inhibition of vmPFC-BLA, but not pIC-BLA, connections can produce long-lasting decreases in spatial avoidance. In contrast, presynaptic inhibition of pIC-BLA connections decreased social avoidance, novelty-induced hypophagia, and increased exploratory behavior without impairing working memory, establishing this projection as a novel target for the treatment of anxiety disorders. Overall, this work reveals new aspects of BLA neuromodulation with therapeutic implications while establishing a powerful approach for optical mapping of drug action via photopharmacology.
Collapse
Affiliation(s)
- Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Vanessa A. Gutzeit
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ipsit Srivastava
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melanie Kristt
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ashna Singh
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Akshara Vijay
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anisul Arefin
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sonal Thukral
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Joseph M. Stujenske
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
16
|
Feng YF, Zhou YY, Duan KM. The Role of Extrasynaptic GABA Receptors in Postpartum Depression. Mol Neurobiol 2024; 61:385-396. [PMID: 37612480 DOI: 10.1007/s12035-023-03574-7] [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: 05/31/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Postpartum depression is a serious disease with a high incidence and severe impact on pregnant women and infants, but its mechanism remains unclear. Recent studies have shown that GABA receptors, especially extrasynaptic receptors, are closely associated with postpartum depression. There are many different structures of GABA receptors, so different types of receptors have different functions, even though they transmit information primarily through GABA. In this review, we focus on the function of GABA receptors, especially extrasynaptic GABA receptors, and their association with postpartum depression. We have shown that the extrasynaptic GABA receptor has a significant impact on the activity and function of neurons through tonic inhibition. The extrasynaptic receptor and its ligands undergo drastic changes during pregnancy and childbirth. Abnormal changes or the body's inability to adjust and recover may be an important cause of postpartum depression. Finally, by reviewing the mechanisms of several novel antidepressants, we suggest that extrasynaptic receptors may be potential targets for the treatment of postpartum depression.
Collapse
Affiliation(s)
- Yun Fei Feng
- Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yin Yong Zhou
- Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Kai Ming Duan
- Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
17
|
Zhang P, Yan J, Wei J, Li Y, Sun C. Disrupted synaptic homeostasis and partial occlusion of associative long-term potentiation in the human cortex during social isolation. J Affect Disord 2024; 344:207-218. [PMID: 37832738 DOI: 10.1016/j.jad.2023.10.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
Social isolation often occurs in the military mission of soldiers but has increased in the general population since the COVID-19 epidemic. Overall synaptic homeostasis along with associative plasticity for the activity-dependent refinement of transmission across single synapses represent basic neural network function and adaptive behavior mechanisms. Here, we use electrophysiological and behavioral indices to non-invasively study the net synaptic strength and long-term potentiation (LTP)-like plasticity of humans in social isolation environments. The theta activity of electroencephalography (EEG) signals and transcranial magnetic stimulation (TMS) intensity to elicit a predefined amplitude of motor-evoked potential (MEP) demonstrate the disrupted synaptic homeostasis in the human cortex during social isolation. Furthermore, the induced MEP change by paired associative stimulation (PAS) demonstrates the partial occlusion of LTP-like plasticity, further behavior performances in a word-pair task are also identified as a potential index. Our study indicates that social isolation disrupts synaptic homeostasis and occludes associative LTP-like plasticity in the human cortex, decreasing behavior performance related to declarative memory.
Collapse
Affiliation(s)
- Peng Zhang
- School of Psychology, Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing 100048, China
| | - Juan Yan
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Jiao Wei
- The First Affiliated Hospital of Shandong First Medical University, Neurosurgery, Jinan 250013, China
| | - Yane Li
- College of Mathematics and Computer Science, Zhejiang A&F University, Hangzhou 311300, China
| | - Chuancai Sun
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China; The First Affiliated Hospital of Shandong First Medical University, Nephrology, Jinan 250013, China.
| |
Collapse
|
18
|
Nuwer JL, Povysheva N, Jacob TC. Long-term α5 GABA A receptor negative allosteric modulator treatment reduces NMDAR-mediated neuronal excitation and maintains basal neuronal inhibition. Neuropharmacology 2023; 237:109587. [PMID: 37270156 PMCID: PMC10527172 DOI: 10.1016/j.neuropharm.2023.109587] [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: 03/27/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 06/05/2023]
Abstract
α5 subunit-containing GABA type-A receptors (α5 GABAARs) are enriched in the hippocampus and play critical roles in neurodevelopment, synaptic plasticity, and cognition. α5 GABAAR preferring negative allosteric modulators (α5 NAMs) show promise mitigating cognitive impairment in preclinical studies of conditions characterized by excess GABAergic inhibition, including Down syndrome and memory deficits post-anesthesia. However, previous studies have primarily focused on acute application or single-dose α5 NAM treatment. Here, we measured the effects of chronic (7-day) in vitro treatment with L-655,708 (L6), a highly selective α5 NAM, on glutamatergic and GABAergic synapses in rat hippocampal neurons. We previously showed that 2-day in vitro treatment with L6 enhanced synaptic levels of the glutamate NMDA receptor (NMDAR) GluN2A subunit without modifying surface α5 GABAAR expression, inhibitory synapse function, or L6 sensitivity. We hypothesized that chronic L6 treatment would further increase synaptic GluN2A subunit levels while maintaining GABAergic inhibition and L6 efficacy, thus increasing neuronal excitation and glutamate-evoked intracellular calcium responses. Immunofluorescence experiments revealed that 7-day L6 treatment slightly increased the synaptic levels of gephyrin and surface α5 GABAARs. Functional studies showed that chronic α5 NAM treatment did not alter inhibition or α5 NAM sensitivity. Surprisingly, chronic L6 exposure decreased surface levels of GluN2A and GluN2B subunits, concurrent with reduced NMDAR-mediated neuronal excitation as seen by faster synaptic decay rates and reduced glutamate-evoked calcium responses. Together, these results show that chronic in vitro treatment with an α5 NAM leads to subtle homeostatic changes in inhibitory and excitatory synapses that suggest an overall dampening of excitability.
Collapse
Affiliation(s)
- Jessica L Nuwer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadya Povysheva
- Department of 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
|
19
|
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
|
20
|
Deng Y, Song D, Ni J, Qing H, Quan Z. Reward prediction error in learning-related behaviors. Front Neurosci 2023; 17:1171612. [PMID: 37662112 PMCID: PMC10471312 DOI: 10.3389/fnins.2023.1171612] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Learning is a complex process, during which our opinions and decisions are easily changed due to unexpected information. But the neural mechanism underlying revision and correction during the learning process remains unclear. For decades, prediction error has been regarded as the core of changes to perception in learning, even driving the learning progress. In this article, we reviewed the concept of reward prediction error, and the encoding mechanism of dopaminergic neurons and the related neural circuities. We also discussed the relationship between reward prediction error and learning-related behaviors, including reversal learning. We then demonstrated the evidence of reward prediction error signals in several neurological diseases, including Parkinson's disease and addiction. These observations may help to better understand the regulatory mechanism of reward prediction error in learning-related behaviors.
Collapse
Affiliation(s)
- Yujun Deng
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
- Department of Biology, Shenzhen MSU-BIT University, Shenzhen, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
21
|
McCoy AM, Prevot TD, Sharmin D, Cook JM, Sibille EL, Lodge DJ. GL-II-73, a Positive Allosteric Modulator of α5GABA A Receptors, Reverses Dopamine System Dysfunction Associated with Pilocarpine-Induced Temporal Lobe Epilepsy. Int J Mol Sci 2023; 24:11588. [PMID: 37511346 PMCID: PMC10380722 DOI: 10.3390/ijms241411588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/15/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
Although seizures are a hallmark feature of temporal lobe epilepsy (TLE), psychiatric comorbidities, including psychosis, are frequently associated with TLE and contribute to decreased quality of life. Currently, there are no defined therapeutic protocols to manage psychosis in TLE patients, as antipsychotic agents may induce epileptic seizures and are associated with severe side effects and pharmacokinetic and pharmacodynamic interactions with antiepileptic drugs. Thus, novel treatment strategies are necessary. Several lines of evidence suggest that hippocampal hyperactivity is central to the pathology of both TLE and psychosis; therefore, restoring hippocampal activity back to normal levels may be a novel therapeutic approach for treating psychosis in TLE. In rodent models, increased activity in the ventral hippocampus (vHipp) results in aberrant dopamine system function, which is thought to underlie symptoms of psychosis. Indeed, we have previously demonstrated that targeting α5-containing γ-aminobutyric acid receptors (α5GABAARs), an inhibitory receptor abundant in the hippocampus, with positive allosteric modulators (PAMs), can restore dopamine system function in rodent models displaying hippocampal hyperactivity. Thus, we posited that α5-PAMs may be beneficial in a model used to study TLE. Here, we demonstrate that pilocarpine-induced TLE is associated with increased VTA dopamine neuron activity, an effect that was completely reversed by intra-vHipp administration of GL-II-73, a selective α5-PAM. Further, pilocarpine did not alter the hippocampal α5GABAAR expression or synaptic localization that may affect the efficacy of α5-PAMs. Taken together, these results suggest augmenting α5GABAAR function as a novel therapeutic modality for the treatment of psychosis in TLE.
Collapse
Affiliation(s)
- Alexandra M. McCoy
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA;
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| | - Thomas D. Prevot
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON M5S 2S1, Canada; (T.D.P.); (E.L.S.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA; (D.S.); (J.M.C.)
| | - James M. Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA; (D.S.); (J.M.C.)
| | - Etienne L. Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON M5S 2S1, Canada; (T.D.P.); (E.L.S.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, TX 78229, USA;
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78229, USA
| |
Collapse
|
22
|
McCoy AM, Prevot TD, Mian MY, Sharmin D, Ahmad AN, Cook JM, Sibille EL, Lodge DJ. Extrasynaptic localization is essential for α5GABA A receptor modulation of dopamine system function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548744. [PMID: 37502875 PMCID: PMC10370028 DOI: 10.1101/2023.07.12.548744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dopamine system dysfunction, observed in animal models with psychosis-like symptomatology, can be restored by targeting Gamma-Aminobutyric Acid type A receptors (GABA A R) containing the α5, but not α1, subunit in the ventral hippocampus (vHipp). The reason for this discrepancy in efficacy remains elusive; however, one key difference is that α1GABA A Rs are primarily located in the synapse, whereas α5GABA A Rs are mostly extrasynaptic. To test whether receptor location is responsible for this difference in efficacy, we injected a small interfering ribonucleic acid (siRNA) into the vHipp to knock down radixin, a scaffolding protein that holds α5GABA A Rs in the extrasynaptic space. We then administered GL-II-73, a positive allosteric modulator of α5GABA A Rs (α5-PAM) known to reverse shock-induced deficits in dopamine system function, to determine if shifting α5GABA A Rs from the extrasynaptic space to the synapse would prevent the effects of α5-PAM on dopamine system function. As expected, knockdown of radixin significantly decreased radixin-associated α5GABA A Rs and increased the proportion of synaptic α5GABA A Rs, without changing the overall expression of α5GABA A Rs. Importantly, GL-II-73 was no longer able to modulate dopamine neuron activity in radixin-knockdown rats, indicating that the extrasynaptic localization of α5GABA A Rs is critical for hippocampal modulation of the dopamine system. These results may have important implications for clinical use of GL-II-73, as periods of high hippocampal activity appear to favor synaptic α5GABA A Rs, thus efficacy may be diminished in conditions where aberrant hippocampal activity is present. Significance Statement Dopamine activity is known to be altered in both psychosis patients and in animal models, with promising new antipsychotics restoring normal dopamine system function. One such drug is GL-II-73, a positive allosteric modulator of α5GABA A Rs (α5-PAM). Interestingly, previous research has shown that a positive allosteric modulator of α1GABA A Rs (α1-PAM) does not share this ability, even when directly given to the ventral hippocampus, a region known to modulate dopamine activity. One potential explanation for this difference we examined in this study is that α1GABA A Rs are primarily located in the synapse, whereas α5GABA A Rs are mostly extrasynaptic. Determining the mechanism of this differential efficacy could lead to the refinement of antipsychotic treatment and improve patient outcomes overall.
Collapse
|
23
|
Wyroślak M, Dobrzański G, Mozrzymas JW. Bidirectional plasticity of GABAergic tonic inhibition in hippocampal somatostatin- and parvalbumin-containing interneurons. Front Cell Neurosci 2023; 17:1193383. [PMID: 37448697 PMCID: PMC10336215 DOI: 10.3389/fncel.2023.1193383] [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: 03/24/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
GABAA receptors present in extrasynaptic areas mediate tonic inhibition in hippocampal neurons regulating the performance of neural networks. In this study, we investigated the effect of NMDA-induced plasticity on tonic inhibition in somatostatin- and parvalbumin-containing interneurons. Using pharmacological methods and transgenic mice (SST-Cre/PV-Cre x Ai14), we induced the plasticity of GABAergic transmission in somatostatin- and parvalbumin-containing interneurons by a brief (3 min) application of NMDA. In the whole-cell patch-clamp configuration, we measured tonic currents enhanced by specific agonists (etomidate or gaboxadol). Furthermore, in both the control and NMDA-treated groups, we examined to what extent these changes depend on the regulation of distinct subtypes of GABAA receptors. Tonic conductance in the somatostatin-containing (SST+) interneurons is enhanced after NMDA application, and the observed effect is associated with an increased content of α5-containing GABAARs. Both fast-spiking and non-fast-spiking parvalbumin-positive (PV+) cells showed a reduction of tonic inhibition after plasticity induction. This effect was accompanied in both PV+ interneuron types by a strongly reduced proportion of δ-subunit-containing GABAARs and a relatively small increase in currents mediated by α5-containing GABAARs. Both somatostatin- and parvalbumin-containing interneurons show cell type-dependent and opposite sign plasticity of tonic inhibition. The underlying mechanisms depend on the cell-specific balance of plastic changes in the contents of α5 and δ subunit-containing GABAARs.
Collapse
Affiliation(s)
- Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | | | - Jerzy W. Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
24
|
Ma X, Johnson DA, He XJ, Layden AE, McClain SP, Yung JC, Rizzo A, Bonaventura J, Banghart MR. In vivo photopharmacology with a caged mu opioid receptor agonist drives rapid changes in behavior. Nat Methods 2023; 20:682-685. [PMID: 36973548 PMCID: PMC10569260 DOI: 10.1038/s41592-023-01819-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 02/16/2023] [Indexed: 03/29/2023]
Abstract
Photoactivatable drugs and peptides can drive quantitative studies into receptor signaling with high spatiotemporal precision, yet few are compatible with behavioral studies in mammals. We developed CNV-Y-DAMGO-a caged derivative of the mu opioid receptor-selective peptide agonist DAMGO. Photoactivation in the mouse ventral tegmental area produced an opioid-dependent increase in locomotion within seconds of illumination. These results demonstrate the power of in vivo photopharmacology for dynamic studies into animal behavior.
Collapse
Affiliation(s)
- Xiang Ma
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Desiree A Johnson
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Xinyi Jenny He
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Aryanna E Layden
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Shannan P McClain
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jean C Yung
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Arianna Rizzo
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
- Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Catalonia, Spain
| | - Matthew R Banghart
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
25
|
Fogarty MJ. Inhibitory Synaptic Influences on Developmental Motor Disorders. Int J Mol Sci 2023; 24:ijms24086962. [PMID: 37108127 PMCID: PMC10138861 DOI: 10.3390/ijms24086962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
During development, GABA and glycine play major trophic and synaptic roles in the establishment of the neuromotor system. In this review, we summarise the formation, function and maturation of GABAergic and glycinergic synapses within neuromotor circuits during development. We take special care to discuss the differences in limb and respiratory neuromotor control. We then investigate the influences that GABAergic and glycinergic neurotransmission has on two major developmental neuromotor disorders: Rett syndrome and spastic cerebral palsy. We present these two syndromes in order to contrast the approaches to disease mechanism and therapy. While both conditions have motor dysfunctions at their core, one condition Rett syndrome, despite having myriad symptoms, has scientists focused on the breathing abnormalities and their alleviation-to great clinical advances. By contrast, cerebral palsy remains a scientific quagmire or poor definitions, no widely adopted model and a lack of therapeutic focus. We conclude that the sheer abundance of diversity of inhibitory neurotransmitter targets should provide hope for intractable conditions, particularly those that exhibit broad spectra of dysfunction-such as spastic cerebral palsy and Rett syndrome.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
26
|
Brzdąk P, Lebida K, Wyroślak M, Mozrzymas JW. GABAergic synapses onto SST and PV interneurons in the CA1 hippocampal region show cell-specific and integrin-dependent plasticity. Sci Rep 2023; 13:5079. [PMID: 36977728 PMCID: PMC10050003 DOI: 10.1038/s41598-023-31882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
It is known that GABAergic transmission onto pyramidal neurons shows different forms of plasticity. However, GABAergic cells innervate also other inhibitory interneurons and plasticity phenomena at these projections remain largely unknown. Several mechanisms underlying plastic changes, both at inhibitory and excitatory synapses, show dependence on integrins, key proteins mediating interaction between intra- and extracellular environment. We thus used hippocampal slices to address the impact of integrins on long-term plasticity of GABAergic synapses on specific inhibitory interneurons (containing parvalbumin, PV + or somatostatin, SST +) known to innervate distinct parts of principal cells. Administration of RGD sequence-containing peptide induced inhibitory long-term potentiation (iLTP) at fast-spiking (FS) PV + as well as on SST + interneurons. Interestingly, treatment with a more specific peptide GA(C)RRETAWA(C)GA (RRETAWA), affecting α5β1 integrins, resulted in iLTP in SST + and iLTD in FS PV + interneurons. Brief exposure to NMDA is known to induce iLTP at GABAergic synapses on pyramidal cells. Intriguingly, application of this protocol for considered interneurons evoked iLTP in SST + and iLTD in PV + interneurons. Moreover, we showed that in SST + cells, NMDA-evoked iLTP depends on the incorporation of GABAA receptors containing α5 subunit to the synapses, and this iLTP is occluded by RRETAWA peptide, indicating a key role of α5β1 integrins. Altogether, our results revealed that plasticity of inhibitory synapses at GABAergic cells shows interneuron-specificity and show differences in the underlying integrin-dependent mechanisms. This is the first evidence that neuronal disinhibition may be a highly plastic process depending on interneuron type and integrins' activity.
Collapse
Affiliation(s)
- Patrycja Brzdąk
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland
| |
Collapse
|
27
|
Gatta E, Camussi D, Auta J, Guidotti A, Pandey SC. Neurosteroids (allopregnanolone) and alcohol use disorder: From mechanisms to potential pharmacotherapy. Pharmacol Ther 2022; 240:108299. [PMID: 36323379 PMCID: PMC9810076 DOI: 10.1016/j.pharmthera.2022.108299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments - disulfiram, acamprosate, and naltrexone - are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.
Collapse
Affiliation(s)
- Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Diletta Camussi
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - James Auta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA; Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612, USA.
| |
Collapse
|
28
|
Hausrat TJ, Vogl C, Neef J, Schweizer M, Yee BK, Strenzke N, Kneussel M. Monoallelic loss of the F-actin-binding protein radixin facilitates startle reactivity and pre-pulse inhibition in mice. Front Cell Dev Biol 2022; 10:987691. [DOI: 10.3389/fcell.2022.987691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Hearing impairment is one of the most common disorders with a global burden and increasing prevalence in an ever-aging population. Previous research has largely focused on peripheral sensory perception, while the brain circuits of auditory processing and integration remain poorly understood. Mutations in the rdx gene, encoding the F-actin binding protein radixin (Rdx), can induce hearing loss in human patients and homozygous depletion of Rdx causes deafness in mice. However, the precise physiological function of Rdx in hearing and auditory information processing is still ill-defined. Here, we investigated consequences of rdx monoallelic loss in the mouse. Unlike the homozygous (−/−) rdx knockout, which is characterized by the degeneration of actin-based stereocilia and subsequent hearing loss, our analysis of heterozygous (+/−) mutants has revealed a different phenotype. Specifically, monoallelic loss of rdx potentiated the startle reflex in response to acoustic stimulation of increasing intensities, suggesting a gain of function relative to wildtype littermates. The monoallelic loss of the rdx gene also facilitated pre-pulse inhibition of the acoustic startle reflex induced by weak auditory pre-pulse stimuli, indicating a modification to the circuit underlying sensorimotor gating of auditory input. However, the auditory brainstem response (ABR)-based hearing thresholds revealed a mild impairment in peripheral sound perception in rdx (+/-) mice, suggesting minor aberration of stereocilia structural integrity. Taken together, our data suggest a critical role of Rdx in the top-down processing and/or integration of auditory signals, and therefore a novel perspective to uncover further Rdx-mediated mechanisms in central auditory information processing.
Collapse
|
29
|
Sleep and wake cycles dynamically modulate hippocampal inhibitory synaptic plasticity. PLoS Biol 2022; 20:e3001812. [PMID: 36318572 PMCID: PMC9624398 DOI: 10.1371/journal.pbio.3001812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/30/2022] [Indexed: 01/01/2023] Open
Abstract
Sleep is an essential process that consolidates memories by modulating synapses through poorly understood mechanisms. Here, we report that GABAergic synapses in hippocampal CA1 pyramidal neurons undergo daily rhythmic alterations. Specifically, wake inhibits phasic inhibition, whereas it promotes tonic inhibition compared to sleep. We further utilize a model of chemically induced inhibitory long-term potentiation (iLTP) to examine inhibitory plasticity. Intriguingly, while CA1 pyramidal neurons in both wake and sleep mice undergo iLTP, wake mice have a much higher magnitude. We also employ optogenetics and observe that inhibitory inputs from parvalbumin-, but not somatostatin-, expressing interneurons contribute to dynamic iLTP during sleep and wake. Finally, we demonstrate that synaptic insertion of α5-GABAA receptors underlies the wake-specific enhancement of iLTP at parvalbumin-synapses, which is independent of time of the day. These data reveal a previously unappreciated daily oscillation of inhibitory LTP in hippocampal neurons and uncover a dynamic contribution of inhibitory synapses in memory mechanisms across sleep and wake.
Collapse
|
30
|
Iggena D, Maier PM, Häußler SM, Menk M, Olze H, Larkum ME, Finke C, Ploner CJ. Post-encoding modulation of spatial memory consolidation by propofol. Cortex 2022; 156:1-12. [DOI: 10.1016/j.cortex.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/09/2022] [Accepted: 08/10/2022] [Indexed: 11/03/2022]
|
31
|
Li Z, Liu X, Li C, Wei Z, Shi Y, Song H, Chen X, Zhang Y, Li J, Zhu R, Hu B, Ye W, Huo D, Jiang G, Sasaki T, Zhang L, Han F, Lu Y. Decreased synapse-associated proteins are associated with the onset of epileptic memory impairment in endothelial CDK5-deficient mice. MedComm (Beijing) 2022; 3:e128. [PMID: 35770064 PMCID: PMC9209881 DOI: 10.1002/mco2.128] [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: 12/09/2021] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/07/2022] Open
Abstract
Accumulating evidence indicates that epilepsy has a higher risk of inducing memory impairment and dementia. However, the underlying onset mechanism remains unclear. Here, we found that mice with spontaneous epilepsy induced by endothelial CDK5 deficiency exhibited hippocampal-dependent memory impairment at 6 months of age, but not at 2 months of age. Moreover, the persistent epileptic seizures induce aberrant changes in phosphorylation of CaMKII protein in the hippocampus of spontaneous epileptic mice. Using genome-wide RNA sequencing and intergenic interaction analysis of STRING, we found that in addition to epilepsy-related genes, there are changes in synaptic organization pathway node genes, such as Bdnf and Grin1. The synapse-related proteins by Western blot analysis, such as NMDA receptors (NR1 and NR2B), PSD95, and the phosphorylation of synapsin1, are progressively decreased during epileptic seizures in Cdh5-CreERT2;CDK5f/f mice. Notably, we found that valproate (VPA) and phenytoin (PHT) augment mRNA expression and protein levels of synapse-related genes and ameliorate memory impairment in Cdh5-CreERT2;CDK5f/f mice. Our study elucidates a potential mechanism of memory deficits in epilepsy, and pharmacological reversal of synaptic pathology targeting might provide a new therapeutic intervention for epileptic memory deficits.
Collapse
Affiliation(s)
- Zheng‐Mao Li
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Xiu‐Xiu Liu
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Chen Li
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Zhao‐Cong Wei
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Yi Shi
- Department of PhysiologyNanjing Medical UniversityNanjingChina
| | - Heng‐Yi Song
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Yu Zhang
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Jia‐Wei Li
- The First Clinical Medical College of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Rui‐Fang Zhu
- The First Clinical Medical College of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Ben‐Hui Hu
- Key Laboratory of Clinical and Medical EngineeringSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjingChina
| | - Wei‐Feng Ye
- Department of PharmacyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Da Huo
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
| | - Guo‐Jun Jiang
- Department of PharmacyZhejiang Xiaoshan HospitalHangzhouChina
| | - Takuya Sasaki
- Department of PharmacologyGraduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
| | - Li Zhang
- Institute of Brain ScienceThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular MedicineSchool of PharmacyNanjing Medical UniversityNanjingChina
- Institute of Brain ScienceThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
- Gusu SchoolNanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhouChina
| | - Ying‐Mei Lu
- Department of PhysiologyNanjing Medical UniversityNanjingChina
- Institute of Brain ScienceThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
32
|
McCoy AM, Prevot TD, Mian MY, Cook JM, Frazer A, Sibille EL, Carreno FR, Lodge DJ. Positive Allosteric Modulation of α5-GABAA Receptors Reverses Stress-Induced Alterations in Dopamine System Function and Prepulse Inhibition of Startle. Int J Neuropsychopharmacol 2022; 25:688-698. [PMID: 35732272 PMCID: PMC9380714 DOI: 10.1093/ijnp/pyac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Up to 64% of patients diagnosed with posttraumatic stress disorder (PTSD) experience psychosis, likely attributable to aberrant dopamine neuron activity. We have previously demonstrated that positive allosteric modulators of α5-GABAARs can selectively decrease hippocampal activity and reverse psychosis-like physiological and behavioral alterations in a rodent model used to study schizophrenia; however, whether this approach translates to a PTSD model remains to be elucidated. METHODS We utilized a 2-day inescapable foot shock (IS) procedure to induce stress-related pathophysiology in male Sprague-Dawley rats. We evaluated the effects of intra-ventral hippocampus (vHipp) administration GL-II-73, an α5-GABAAR, or viral overexpression of the α5 subunit, using in vivo electrophysiology and behavioral measures in control and IS-treated rats. RESULTS IS significantly increased ventral tegmental area dopamine neuron population activity, or the number of dopamine neurons firing spontaneously (n = 6; P = .016), consistent with observation in multiple rodent models used to study psychosis. IS also induced deficits in sensorimotor gating, as measured by reduced prepulse inhibition of startle (n = 12; P = .039). Interestingly, intra-vHipp administration of GL-II-73 completely reversed IS-induced increases in dopamine neuron population activity (n = 6; P = .024) and deficits in prepulse inhibition (n = 8; P = .025), whereas viral overexpression of the α5 subunit in the vHipp was not effective. CONCLUSIONS Our results demonstrate that pharmacological intervention augmenting α5-GABAAR function, but not α5 overexpression in itself, can reverse stress-induced deficits related to PTSD in a rodent model, providing a potential site of therapeutic intervention to treat comorbid psychosis in PTSD.
Collapse
Affiliation(s)
- Alexandra M McCoy
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas, USA
| | - Thomas D Prevot
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Md Yenus Mian
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Alan Frazer
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas, USA
| | - Etienne L Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Flavia R Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas, USA
| | - Daniel J Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, Texas, USA
| |
Collapse
|
33
|
Troppoli TA, Zanos P, Georgiou P, Gould TD, Rudolph U, Thompson SM. Negative Allosteric Modulation of Gamma-Aminobutyric Acid A Receptors at α5 Subunit-Containing Benzodiazepine Sites Reverses Stress-Induced Anhedonia and Weakened Synaptic Function in Mice. Biol Psychiatry 2022; 92:216-226. [PMID: 35120711 PMCID: PMC9198111 DOI: 10.1016/j.biopsych.2021.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Abnormal reward processing, typically anhedonia, is a hallmark of human depression and is accompanied by altered functional connectivity in reward circuits. Negative allosteric modulators of GABAA (gamma-aminobutyric acid A) receptors (GABA-NAMs) have rapid antidepressant-like properties in rodents and exert few adverse effects, but molecular targets underlying their behavioral and synaptic effects remain undetermined. We hypothesized that GABA-NAMs act at the benzodiazepine site of GABAA receptors containing α5 subunits to increase gamma oscillatory activity, strengthen synapses in reward circuits, and reverse anhedonia. METHODS Anhedonia was induced by chronic stress in male mice and assayed by preferences for sucrose and female urine (n = 5-7 mice/group). Hippocampal slices were then prepared for electrophysiological recording (n = 1-6 slices/mouse, 4-6 mice/group). Electroencephalography power was quantified in response to GABA-NAM and ketamine administration (n = 7-9 mice/group). RESULTS Chronic stress reduced sucrose and female urine preferences and hippocampal temporoammonic-CA1 synaptic strength. A peripheral injection of the GABA-NAM MRK-016 restored hedonic behavior and AMPA-to-NMDA ratios in wild-type mice. These actions were prevented by pretreatment with the benzodiazepine site antagonist flumazenil. MRK-016 administration increased gamma power over the prefrontal cortex in wild-type mice but not α5 knockout mice, whereas ketamine promoted gamma power in both genotypes. Hedonic behavior and AMPA-to-NMDA ratios were only restored by MRK-016 in stressed wild-type mice but not α5 knockout mice. CONCLUSIONS α5-Selective GABA-NAMs exert rapid anti-anhedonic actions and restore the strength of synapses in reward regions by acting at the benzodiazepine site of α5-containing GABAA receptors. These results encourage human studies using GABA-NAMs to treat depression by providing readily translatable measures of target engagement.
Collapse
Affiliation(s)
- Timothy A. Troppoli
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Molecular Medicine Program, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201
| | - Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Current address: Department of Psychology, University of Cyprus, 1 Panepistimiou Avenue, Aglantzia, 2109, PO Box 1678, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychiatry, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Department of Anatomy & Neurobiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Department of Pharmacology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201
| | - Uwe Rudolph
- Department of Comparative Biosciences and Carl R. Woese Institute for Genomic Biology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, 2001 S Lincoln Ave, Urbana, IL 61802-6178
| | - Scott M. Thompson
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,Department of Psychiatry, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201,To whom correspondence should be addressed:
| |
Collapse
|
34
|
The general anaesthetic propofol prevents cerebrocortical potentiation in neocortical mouse brain slices. Brain Res 2022; 1792:148018. [PMID: 35850186 DOI: 10.1016/j.brainres.2022.148018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Propofol is well known to cause amnesia independent of its sedative effect. Memory consolidation processes in the hippocampus have been proposed as a target - however the neural substrates for propofol's amnesic actions remain understudied and poorly described. In particular, the potential role of the cerebral cortex has not been investigated. As an in vitro experimental model of cortical memory consolidation, potentiated cerebral cortex evoked responses were generated in mouse neocortical slices using high frequency (20 Hz) stimulation to layer IV cortical grey matter or subcortical white matter. In separate experiments, slices were pretreated with propofol at two concentrations, 2 µg/mL and 4 µg/mL, to determine the effect of clinically relevant propofol levels on the potentiation response. Only grey matter stimulation induced a significant and lasting increase in cortical evoked potential amplitude in the drug-free condition. Propofol at 2 µg/mL completely inhibited cortical evoked response potentiation, while the 4 µg/mL concentration caused a small but significant depressant effect consequent to the high frequency stimulation. These findings support the hypothesis that propofol disrupts memory consolidation and actively facilitates memory decay in the cerebral cortex. The results further highlight the importance of the cerebral cortex in the early phase of long term memory consolidation.
Collapse
|
35
|
Cerne R, Lippa A, Poe MM, Smith JL, Jin X, Ping X, Golani LK, Cook JM, Witkin JM. GABAkines - Advances in the discovery, development, and commercialization of positive allosteric modulators of GABA A receptors. Pharmacol Ther 2022; 234:108035. [PMID: 34793859 PMCID: PMC9787737 DOI: 10.1016/j.pharmthera.2021.108035] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022]
Abstract
Positive allosteric modulators of γ-aminobutyric acid-A (GABAA) receptors or GABAkines have been widely used medicines for over 70 years for anxiety, epilepsy, sleep, and other disorders. Traditional GABAkines like diazepam have safety and tolerability concerns that include sedation, motor-impairment, respiratory depression, tolerance and dependence. Multiple GABAkines have entered clinical development but the issue of side-effects has not been fully solved. The compounds that are presently being developed and commercialized include several neuroactive steroids (an allopregnanolone formulation (brexanolone), an allopregnanolone prodrug (LYT-300), Sage-324, zuranolone, and ganaxolone), the α2/3-preferring GABAkine, KRM-II-81, and the α2/3/5-preferring GABAkine PF-06372865 (darigabat). The neuroactive steroids are in clinical development for post-partum depression, intractable epilepsy, tremor, status epilepticus, and genetic epilepsy disorders. Darigabat is in development for epilepsy and anxiety. The imidazodiazepine, KRM-II-81 is efficacious in animal models for the treatment of epilepsy and post-traumatic epilepsy, acute and chronic pain, as well as anxiety and depression. The efficacy of KRM-II-81 in models of pharmacoresistant epilepsy, preventing the development of seizure sensitization, and in brain tissue of intractable epileptic patients bodes well for improved therapeutics. Medicinal chemistry efforts are also ongoing to identify novel and improved GABAkines. The data document gaps in our understanding of the molecular pharmacology of GABAkines that drive differential pharmacological profiles, but emphasize advancements in the ability to successfully utilize GABAA receptor potentiation for therapeutic gain in neurology and psychiatry.
Collapse
Affiliation(s)
- Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN USA,Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia.,RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA,Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA
| | | | - Jodi L. Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN USA
| | - Xiaoming Jin
- Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Xingjie Ping
- Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Lalit K. Golani
- Department of Chemistry and Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - James M. Cook
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA,Department of Chemistry and Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jeffrey M. Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN USA,RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA,Department of Chemistry and Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
36
|
Chapman CA, Nuwer JL, Jacob TC. The Yin and Yang of GABAergic and Glutamatergic Synaptic Plasticity: Opposites in Balance by Crosstalking Mechanisms. Front Synaptic Neurosci 2022; 14:911020. [PMID: 35663370 PMCID: PMC9160301 DOI: 10.3389/fnsyn.2022.911020] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/26/2022] [Indexed: 01/12/2023] Open
Abstract
Synaptic plasticity is a critical process that regulates neuronal activity by allowing neurons to adjust their synaptic strength in response to changes in activity. Despite the high proximity of excitatory glutamatergic and inhibitory GABAergic postsynaptic zones and their functional integration within dendritic regions, concurrent plasticity has historically been underassessed. Growing evidence for pathological disruptions in the excitation and inhibition (E/I) balance in neurological and neurodevelopmental disorders indicates the need for an improved, more "holistic" understanding of synaptic interplay. There continues to be a long-standing focus on the persistent strengthening of excitation (excitatory long-term potentiation; eLTP) and its role in learning and memory, although the importance of inhibitory long-term potentiation (iLTP) and depression (iLTD) has become increasingly apparent. Emerging evidence further points to a dynamic dialogue between excitatory and inhibitory synapses, but much remains to be understood regarding the mechanisms and extent of this exchange. In this mini-review, we explore the role calcium signaling and synaptic crosstalk play in regulating postsynaptic plasticity and neuronal excitability. We examine current knowledge on GABAergic and glutamatergic synapse responses to perturbances in activity, with a focus on postsynaptic plasticity induced by short-term pharmacological treatments which act to either enhance or reduce neuronal excitability via ionotropic receptor regulation in neuronal culture. To delve deeper into potential mechanisms of synaptic crosstalk, we discuss the influence of synaptic activity on key regulatory proteins, including kinases, phosphatases, and synaptic structural/scaffolding proteins. Finally, we briefly suggest avenues for future research to better understand the crosstalk between glutamatergic and GABAergic synapses.
Collapse
Affiliation(s)
| | | | - Tija C. Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
37
|
Frank JA. Optofluidic neural interfaces for in vivo photopharmacology. Curr Opin Pharmacol 2022; 63:102195. [DOI: 10.1016/j.coph.2022.102195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 11/03/2022]
|
38
|
Kramer RH, Rajappa R. Interrogating the function of GABA A receptors in the brain with optogenetic pharmacology. Curr Opin Pharmacol 2022; 63:102198. [PMID: 35276498 DOI: 10.1016/j.coph.2022.102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/26/2022]
Abstract
To better understand neural circuits and behavior, microbial opsins have been developed as optogenetic tools for stimulating or inhibiting action potentials with high temporal and spatial precision. However, if we seek a more reductionist understanding of how neuronal circuits operate, we also need high-resolution tools for perturbing the function of synapses. By combining photochemical tools and molecular biology, a wide variety of light-regulated neurotransmitter receptors have been developed, enabling photo-control of excitatory, inhibitory, and modulatory synaptic transmission. Here we focus on photo-control of GABAA receptors, ligand-gated Cl- channels that underlie almost all synaptic inhibition in the mammalian brain. By conjugating a photoswitchable tethered ligand onto a genetically-modified subunit of the GABAA receptor, light-sensitivity can be conferred onto specific isoforms of the receptor. Through gene editing, this attachment site can be knocked into the genome, enabling photocontrol of endogenous GABAA receptors. This strategy can be employed to explore the cell biology and neurophysiology of GABAA receptors. This includes investigating how specific isoforms contribute to synaptic and tonic inhibition and understanding the roles they play in brain development, long-term synaptic plasticity, and learning and memory.
Collapse
Affiliation(s)
- Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States.
| | - Rajit Rajappa
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, United States; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| |
Collapse
|
39
|
Advances in tethered photopharmacology for precise optical control of signaling proteins. Curr Opin Pharmacol 2022; 63:102196. [PMID: 35245800 DOI: 10.1016/j.coph.2022.102196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/25/2022]
Abstract
To overcome the limitations of traditional pharmacology, the field of photopharmacology has developed around the central concept of using light to endow drug action with spatiotemporal precision. Tethered photopharmacology, where a photoswitchable ligand is covalently attached to a target protein, offers a particularly high degree of spatiotemporal control, as well as the ability to genetically target drug action and limit effects to specific protein subtypes. In this review, we describe the core engineering concepts of tethered pharmacology and highlight recent advances in harnessing the power of tethered photopharmacology for an expanded palette of targets and conjugation modes using new, complementary strategies. We also discuss the various applications, including mechanistic studies from the molecular biophysical realm to in vivo studies in behaving animals, that demonstrate the power of tethered pharmacology.
Collapse
|
40
|
Ravasenga T, Ruben M, Regio V, Polenghi A, Petrini EM, Barberis A. Spatial regulation of coordinated excitatory and inhibitory synaptic plasticity at dendritic synapses. Cell Rep 2022; 38:110347. [PMID: 35139381 PMCID: PMC8844559 DOI: 10.1016/j.celrep.2022.110347] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 09/16/2021] [Accepted: 01/14/2022] [Indexed: 12/02/2022] Open
Abstract
The induction of synaptic plasticity at an individual dendritic glutamatergic spine can affect neighboring spines. This local modulation generates dendritic plasticity microdomains believed to expand the neuronal computational capacity. Here, we investigate whether local modulation of plasticity can also occur between glutamatergic synapses and adjacent GABAergic synapses. We find that the induction of long-term potentiation at an individual glutamatergic spine causes the depression of nearby GABAergic inhibitory synapses (within 3 μm), whereas more distant ones are potentiated. Notably, L-type calcium channels and calpain are required for this plasticity spreading. Overall, our data support a model whereby input-specific glutamatergic postsynaptic potentiation induces a spatially regulated rearrangement of inhibitory synaptic strength in the surrounding area through short-range heterosynaptic interactions. Such local coordination of excitatory and inhibitory synaptic plasticity is expected to influence dendritic information processing and integration. LTP of individual dendritic spines causes iLTD at neighboring GABAergic synapses Interaction between single-spine LTP and iLTD occurs in the spatial range of ±3 μm This iLTD depends on the local dendritic calcium increase and calpain activation iLTD is associated with reduced gephyrin clustering and increased GABAAR mobility
Collapse
Affiliation(s)
- Tiziana Ravasenga
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Massimo Ruben
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Vincenzo Regio
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Alice Polenghi
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Enrica Maria Petrini
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Andrea Barberis
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
41
|
Belelli D, Phillips GD, Atack JR, Lambert JJ. Relating neurosteroid modulation of inhibitory neurotransmission to behaviour. J Neuroendocrinol 2022; 34:e13045. [PMID: 34644812 DOI: 10.1111/jne.13045] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022]
Abstract
Studies in the 1980s revealed endogenous metabolites of progesterone and deoxycorticosterone to be potent, efficacious, positive allosteric modulators (PAMs) of the GABAA receptor (GABAA R). The discovery that such steroids are locally synthesised in the central nervous system (CNS) promoted the thesis that neural inhibition in the CNS may be "fine-tuned" by these neurosteroids to influence behaviour. In preclinical studies, these neurosteroids exhibited anxiolytic, anticonvulsant, analgesic and sedative properties and, at relatively high doses, induced a state of general anaesthesia, a profile consistent with their interaction with GABAA Rs. However, realising the therapeutic potential of either endogenous neurosteroids or synthetic "neuroactive" steroids has proven challenging. Recent approval by the Food and Drug Administration of the use of allopregnanolone (brexanolone) to treat postpartum depression has rekindled enthusiasm for exploring their potential as new medicines. Although neurosteroids are selective for GABAA Rs, they exhibit little or no selectivity across the many GABAA R subtypes. Nevertheless, a relatively minor population of receptors incorporating the δ-subunit (δ-GABAA Rs) appears to be an important contributor to their behavioural effects. Here, we consider how neurosteroids acting upon GABAA Rs influence neuronal signalling, as well as how such effects may acutely and persistently influence behaviour, and explore the case for developing selective PAMs of δ-GABAA R subtypes for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Delia Belelli
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Grant D Phillips
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - John R Atack
- Medicines Discovery Institute, Cardiff University, Cardiff, UK
| | - Jeremy J Lambert
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
42
|
Perez SM, McCoy AM, Prevot TD, Mian MY, Carreno FR, Frazer A, Cook JM, Sibille E, Lodge DJ. Hippocampal α5-GABA A Receptors Modulate Dopamine Neuron Activity in the Rat Ventral Tegmental Area. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 3:78-86. [PMID: 36712569 PMCID: PMC9874136 DOI: 10.1016/j.bpsgos.2021.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 02/01/2023] Open
Abstract
Background Aberrant dopamine neuron activity is attributable to hyperactivity in hippocampal subfields driving a pathological increase in dopamine neuron activity, which is positively correlated with psychosis in humans. Evidence indicates that hippocampal hyperactivity is due to loss of intrinsic GABAergic (gamma-aminobutyric acidergic) inhibition. We have previously demonstrated that hippocampal GABAergic neurotransmission can be modulated by targeting α5-GABAA receptors, which are preferentially expressed in hippocampal regions. Positive and negative allosteric modulators of α5-GABAA receptors (α5-PAMs and α5-NAMs) elicit effects on hippocampal-dependent behaviors. We posited that the selective manipulation of hippocampal inhibition, using α5-PAMs or α5-NAMs, would modulate dopamine activity in control rats. Further, α5-PAMs would reverse aberrant dopamine neuron activity in a rodent model with schizophrenia-related pathophysiologies (methylazoxymethanol acetate [MAM] model). Methods We performed in vivo extracellular recordings of ventral tegmental area dopamine neurons in anesthetized rats to compare the effects of two novel, selective α5-PAMs (GL-II-73, MP-III-022), a nonselective α-PAM (midazolam), and two selective α5-NAMs (L-655,708, TB 21007) in control and MAM-treated male Sprague Dawley rats (n = 5-9). Results Systemic or intracranial administration of selective α5-GABAA receptor modulators regulated dopamine activity. Specifically, both α5-NAMs increased dopamine neuron activity in control rats, whereas GL-II-73, MP-III-022, and L-655,708 attenuated aberrant dopamine neuron activity in MAM-treated rats, an effect mediated by the ventral hippocampus. Conclusions This study demonstrated that α5-GABAA receptor modulation can regulate dopamine neuron activity under control or abnormal activity, providing additional evidence that α5-PAMs and α5-NAMs may have therapeutic applications in psychosis and other psychiatric diseases where aberrant hippocampal activity is present.
Collapse
Affiliation(s)
- Stephanie M. Perez
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas,Audie L. Murphy Memorial Veterans Hospital, South Texas Veterans Health Care System, San Antonio, Texas,Address correspondence to Stephanie M. Perez, Ph.D.
| | - Alexandra M. McCoy
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas,Audie L. Murphy Memorial Veterans Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Thomas D. Prevot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Md Yeunus Mian
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Flavia R. Carreno
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas
| | - Alan Frazer
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas,Audie L. Murphy Memorial Veterans Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - James M. Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, UT Health San Antonio, San Antonio, Texas,Audie L. Murphy Memorial Veterans Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
43
|
Yao HK, Guet-McCreight A, Mazza F, Moradi Chameh H, Prevot TD, Griffiths JD, Tripathy SJ, Valiante TA, Sibille E, Hay E. Reduced inhibition in depression impairs stimulus processing in human cortical microcircuits. Cell Rep 2022; 38:110232. [PMID: 35021088 DOI: 10.1016/j.celrep.2021.110232] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/07/2021] [Accepted: 12/16/2021] [Indexed: 12/01/2022] Open
Abstract
Cortical processing depends on finely tuned excitatory and inhibitory connections in neuronal microcircuits. Reduced inhibition by somatostatin-expressing interneurons is a key component of altered inhibition associated with treatment-resistant major depressive disorder (depression), which is implicated in cognitive deficits and rumination, but the link remains to be better established mechanistically in humans. Here we test the effect of reduced somatostatin interneuron-mediated inhibition on cortical processing in human neuronal microcircuits using a data-driven computational approach. We integrate human cellular, circuit, and gene expression data to generate detailed models of human cortical microcircuits in health and depression. We simulate microcircuit baseline and response activity and find a reduced signal-to-noise ratio and increased false/failed detection of stimuli due to a higher baseline activity in depression. We thus apply models of human cortical microcircuits to demonstrate mechanistically how reduced inhibition impairs cortical processing in depression, providing quantitative links between altered inhibition and cognitive deficits.
Collapse
Affiliation(s)
- Heng Kang Yao
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada
| | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada
| | - John D Griffiths
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Shreejoy J Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Taufik A Valiante
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 1A1; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada; Max Planck-University of Toronto Center for Neural Science and Technology, University of Toronto, Toronto, ON M5S 1A1, Canada; Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON M5T 1R7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada.
| |
Collapse
|
44
|
Non-linear GABA A receptors promote synaptic inhibition in developing neurons : Commentary on Lodge et al. (2021) Sparsification of AP firing in adult-born hippocampal granule cells via voltage-dependent α5-GABA A receptors. Cell Rep 37:109768. Pflugers Arch 2021; 474:181-183. [PMID: 34907453 PMCID: PMC8766374 DOI: 10.1007/s00424-021-02652-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/25/2022]
|
45
|
Genetically encoded intrabodies as high-precision tools to visualize and manipulate neuronal function. Semin Cell Dev Biol 2021; 126:117-124. [PMID: 34782184 DOI: 10.1016/j.semcdb.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/24/2022]
Abstract
Basic neuroscience research employs numerous forms of antibodies as key reagents in diverse applications. While the predominant use of antibodies is as immunolabeling reagents, neuroscientists are making increased use of intracellular antibodies or intrabodies. Intrabodies are recombinant antibodies genetically encoded for expression within neurons. These can be used to target various cargo (fluorescent proteins, reporters, enzymes, etc.) to specific molecules and subcellular domains to report on and manipulate neuronal function with high precision. Intrabodies have the advantages inherent in all genetically encoded recombinant antibodies but represent a distinct subclass in that their structure allows for their expression and function within cells. The high precision afforded by the ability to direct their expression to specific cell types, and the selective binding of intrabodies to targets within these allows intrabodies to offer unique advantages for neuroscience research, given the tremendous molecular, cellular and morphological complexity of brain neurons. Intrabodies expressed within neurons have been used for a variety of purposes in basic neuroscience research. Here I provide a general background to intrabodies and their development, and examples of their emerging utility as valuable basic neuroscience research tools.
Collapse
|
46
|
Chen L, Yan H, Wang Y, He Z, Leng Q, Huang S, Wu F, Feng X, Yan J. The Mechanisms and Boundary Conditions of Drug Memory Reconsolidation. Front Neurosci 2021; 15:717956. [PMID: 34421529 PMCID: PMC8377231 DOI: 10.3389/fnins.2021.717956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Drug addiction can be seen as a disorder of maladaptive learning characterized by relapse. Therefore, disrupting drug-related memories could be an approach to improving therapies for addiction. Pioneering studies over the last two decades have revealed that consolidated memories are not static, but can be reconsolidated after retrieval, thereby providing candidate pathways for the treatment of addiction. The limbic-corticostriatal system is known to play a vital role in encoding the drug memory engram. Specific structures within this system contribute differently to the process of memory reconsolidation, making it a potential target for preventing relapse. In addition, as molecular processes are also active during memory reconsolidation, amnestic agents can be used to attenuate drug memory. In this review, we focus primarily on the brain structures involved in storing the drug memory engram, as well as the molecular processes involved in drug memory reconsolidation. Notably, we describe reports regarding boundary conditions constraining the therapeutic potential of memory reconsolidation. Furthermore, we discuss the principles that could be employed to modify stored memories. Finally, we emphasize the challenge of reconsolidation-based strategies, but end with an optimistic view on the development of reconsolidation theory for drug relapse prevention.
Collapse
Affiliation(s)
- Liangpei Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - He Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Yufang Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Ziping He
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qihao Leng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shihao Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Feilong Wu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiangyang Feng
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China.,Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
47
|
Giorgi C, Marinelli S. Roles and Transcriptional Responses of Inhibitory Neurons in Learning and Memory. Front Mol Neurosci 2021; 14:689952. [PMID: 34211369 PMCID: PMC8239217 DOI: 10.3389/fnmol.2021.689952] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence supports a model whereby memories are encoded by sparse ensembles of neurons called engrams, activated during memory encoding and reactivated upon recall. An engram consists of a network of cells that undergo long-lasting modifications of their transcriptional programs and connectivity. Ground-breaking advancements in this field have been made possible by the creative exploitation of the characteristic transcriptional responses of neurons to activity, allowing both engram labeling and manipulation. Nevertheless, numerous aspects of engram cell-type composition and function remain to be addressed. As recent transcriptomic studies have revealed, memory encoding induces persistent transcriptional and functional changes in a plethora of neuronal subtypes and non-neuronal cells, including glutamatergic excitatory neurons, GABAergic inhibitory neurons, and glia cells. Dissecting the contribution of these different cellular classes to memory engram formation and activity is quite a challenging yet essential endeavor. In this review, we focus on the role played by the GABAergic inhibitory component of the engram through two complementary lenses. On one hand, we report on available physiological evidence addressing the involvement of inhibitory neurons to different stages of memory formation, consolidation, storage and recall. On the other, we capitalize on a growing number of transcriptomic studies that profile the transcriptional response of inhibitory neurons to activity, revealing important clues on their potential involvement in learning and memory processes. The picture that emerges suggests that inhibitory neurons are an essential component of the engram, likely involved in engram allocation, in tuning engram excitation and in storing the memory trace.
Collapse
Affiliation(s)
- Corinna Giorgi
- CNR, Institute of Molecular Biology and Pathology, Rome, Italy.,European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
48
|
Wyroślak M, Lebida K, Mozrzymas JW. Induction of Inhibitory Synaptic Plasticity Enhances Tonic Current by Increasing the Content of α5-Subunit Containing GABA A Receptors in Hippocampal Pyramidal Neurons. Neuroscience 2021; 467:39-46. [PMID: 34033868 DOI: 10.1016/j.neuroscience.2021.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022]
Abstract
It is known that besides synaptic inhibition, there is a persistent component of inhibitory drive mediated by tonic currents which is believed to mediate majority of the total inhibitory charge in hippocampal neurons. Tonic currents, depending on cell types, can be mediated by a variety of GABAA receptor (GABAAR) subtypes but in pyramidal neurons, α5-subunit containing receptors were found to be predominant. Importantly, α5-GABAARs were implicated in both inhibitory and excitatory synaptic plasticity as well as in a variety of cognitive tasks. In the present study, we asked whether the protocol that evokes NMDAR-dependent GABAergic inhibitory long-term potentiation (iLTP) also induces the plasticity of tonic inhibition in hippocampal pyramidal neurons. Our whole-cell patch-clamp recordings revealed that the induction of this type of iLTP is associated with a marked increase in tonic current. By using the specific inverse agonist of α5-containing GABAARs (L-655,709) we provide evidence that this plastic change in tonic current is correlated with an increased proportion of this type of GABAARs. On the contrary, the iLTP induction did not affect the tonic current potentiated by THIP, indicating that the pool of δ subunit-containing GABAARs receptors remains unaffected. We conclude that the α5-GABAARs-dependent plasticity of tonic inhibition is a novel dimension of the neuroplasticity of the inhibitory drive in the hippocampal principal neurons. Overall, α5-containing GABAARs emerge as key players in a variety of plasticity mechanisms operating over a large span of time and spatial scales.
Collapse
Affiliation(s)
- Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland.
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland
| |
Collapse
|
49
|
Maingret F, Groc L. Characterization of the Functional Cross-Talk between Surface GABA A and Dopamine D5 Receptors. Int J Mol Sci 2021; 22:4867. [PMID: 34064454 PMCID: PMC8125140 DOI: 10.3390/ijms22094867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 01/17/2023] Open
Abstract
The γ-aminobutyric acid type A receptor (GABAAR) plays a major role in fast inhibitory synaptic transmission and is highly regulated by the neuromodulator dopamine. In this aspect, most of the attention has been focused on the classical intracellular signaling cascades following dopamine G-protein-coupled receptor activation. Interestingly, the GABAAR and dopamine D5 receptor (D5R) have been shown to physically interact in the hippocampus, but whether a functional cross-talk occurs is still debated. In the present study, we use a combination of imaging and single nanoparticle tracking in live hippocampal neurons to provide evidence that GABAARs and D5Rs form dynamic surface clusters. Disrupting the GABAAR-D5R interaction with a competing peptide leads to an increase in the diffusion coefficient and the explored area of both receptors, and a drop in immobile synaptic GABAARs. By means of patch-clamp recordings, we show that this fast lateral redistribution of surface GABAARs correlates with a robust depression in the evoked GABAergic currents. Strikingly, it also shifts in time the expression of long-term potentiation at glutamatergic synapses. Together, our data both set the plasma membrane as the primary stage of a functional interplay between GABAAR and D5R, and uncover a non-canonical role in regulating synaptic transmission.
Collapse
Affiliation(s)
- François Maingret
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, UMR 5297, 33076 Bordeaux, France;
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33076 Bordeaux, France
| | - Laurent Groc
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, UMR 5297, 33076 Bordeaux, France;
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33076 Bordeaux, France
| |
Collapse
|
50
|
Gutzeit VA, Acosta-Ruiz A, Munguba H, Häfner S, Landra-Willm A, Mathes B, Mony J, Yarotski D, Börjesson K, Liston C, Sandoz G, Levitz J, Broichhagen J. A fine-tuned azobenzene for enhanced photopharmacology in vivo. Cell Chem Biol 2021; 28:1648-1663.e16. [PMID: 33735619 DOI: 10.1016/j.chembiol.2021.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/23/2020] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Despite the power of photopharmacology for interrogating signaling proteins, many photopharmacological systems are limited by their efficiency, speed, or spectral properties. Here, we screen a library of azobenzene photoswitches and identify a urea-substituted "azobenzene-400" core that offers bistable switching between cis and trans with improved kinetics, light sensitivity, and a red-shift. We then focus on the metabotropic glutamate receptors (mGluRs), neuromodulatory receptors that are major pharmacological targets. Synthesis of "BGAG12,400," a photoswitchable orthogonal, remotely tethered ligand (PORTL), enables highly efficient, rapid optical agonism following conjugation to SNAP-tagged mGluR2 and permits robust optical control of mGluR1 and mGluR5 signaling. We then produce fluorophore-conjugated branched PORTLs to enable dual imaging and manipulation of mGluRs and highlight their power in ex vivo slice and in vivo behavioral experiments in the mouse prefrontal cortex. Finally, we demonstrate the generalizability of our strategy by developing an improved soluble, photoswitchable pore blocker for potassium channels.
Collapse
Affiliation(s)
- Vanessa A Gutzeit
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Amanda Acosta-Ruiz
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stephanie Häfner
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Arnaud Landra-Willm
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Bettina Mathes
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Jürgen Mony
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Dzianis Yarotski
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Karl Börjesson
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Guillaume Sandoz
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Joshua Levitz
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Johannes Broichhagen
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany; Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany.
| |
Collapse
|